Bacterially expressed full-length recombinant Plasmodium falciparum RH5 protein binds erythrocytes and elicits potent strain-transcending parasite-neutralizing antibodies

Share Embed


Descripción

IAI Accepts, published online ahead of print on 14 October 2013 Infect. Immun. doi:10.1128/IAI.00970-13 Copyright © 2013, American Society for Microbiology. All Rights Reserved.

1

Classification: Microbial Immunity & Vaccines

2 3

A bacterially expressed full-length recombinant Plasmodium falciparum RH5 protein binds

4

erythrocytes and elicits potent strain-transcending parasite neutralizing antibodies

5 6

K. Sony Reddy§, Alok K. Pandey§, Hina Singh, Tajali Sahar, Amlabu Emmanuel,

7

Chetan E. Chitnis, Virander S. Chauhan, Deepak Gaur*

8 9 10

Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, India.

11 12

*Corresponding Author

13

Please send correspondence to:

14 15 16 17 18 19 20 21 22 23

Deepak Gaur, Ph.D. Malaria Group International Centre for Genetic Engineering and Biotechnology (ICGEB) Aruna Asaf Ali Marg New Delhi, India Tel: +91-11-26741358 Fax: +91-11-26742316 E-mail: [email protected]

24

§ These authors contributed equally to the work

25 26

Running Title: Recombinant PfRH5 elicits neutralizing antibodies

27 28

Keywords: Malaria, Plasmodium, erythrocyte, invasion, ligand, receptors, vaccine, host pathogen

29

interaction.

30 31 1

34 32 Abstract 33 35 Plasmodium falciparum reticulocyte binding-like homologous protein 5 (PfRH5) is an essential 36

merozoite ligand that binds with its erythrocyte receptor, Basigin. PfRH5 is an attractive malaria

37

vaccine candidate as it is expressed by a wide number of P. falciparum strains, cannot be

38

genetically disrupted and exhibits limited sequence polymorphisms. Viral vector induced PfRH5

39

antibodies potently inhibited erythrocyte invasion. However, it has been a challenge to generate

40

full-length recombinant PfRH5 in a bacterial cell based expression system. Here, we have

41

produced full-length recombinant PfRH5 in Escherichia coli that exhibits specific erythrocyte

42

binding similar to that of the native PfRH5 parasite protein and also importantly elicits potent

43

invasion inhibitory antibodies against a number of P. falciparum strains. Anti-Basigin antibodies

44

blocked the erythrocyte binding of both native and recombinant PfRH5 further confirming that

45

they bind with Basigin. We have thus successfully produced full-length PfRH5 as a functionally

46

active erythrocyte binding recombinant protein with a conformational integrity that mimics the

47

native parasite protein and elicits potent strain-transcending parasite neutralizing antibodies. P.

48

falciparum has the capability to develop immune escape mechanisms and thus blood-stage

49

malaria vaccines that target multiple antigens or pathways may prove to be highly efficacious. In

50

this regard, antibody combinations targeting PfRH5 and other key merozoite antigens produced

51

potent additive inhibition against multiple worldwide P. falciparum strains. PfRH5 was

52

immunogenic when immunized with other antigens eliciting potent invasion inhibitory antibody

53

responses with no immune interference. Our results strongly support the development of PfRH5

54

as a component of a combination blood-stage malaria vaccine.

55 56 2

57

Introduction

58

Malaria is a global infectious disease that accounts for around one million deaths across

59

the world primarily in young children below the age of five years (1). The causative agent of the

60

most severe form of malaria that is responsible for maximum mortality is the parasite,

61

Plasmodium falciparum. Invasion of human erythrocytes by P. falciparum is a critical process

62

during the parasite’s life cycle that leads to the development of blood stage parasites, which are

63

primarily responsible for malaria pathogenesis. P. falciparum has evolved a complex, multistep

64

process of erythrocyte invasion that involves numerous ligand-receptor interactions (2-4). This

65

molecular redundancy allows the parasite to use many alternate pathways for invasion, thus

66

ensuring that the pathogen gains entry into its host erythrocyte (2-4).

67

The quest for developing a vaccine that targets blood-stage parasites has involved

68

extensive studies on identifying and characterizing key parasite molecules that mediate

69

erythrocyte invasion. Early efforts have focused on two leading candidates – MSP-142 and AMA-

70

1, which play an essential role in erythrocyte invasion (2-4) but have unfortunately not generated

71

optimal protection in field efficacy trials (5-7). Recently, the family of P. falciparum reticulocyte

72

binding-like homologous proteins (PfRH) has attracted most attention as key determinants of

73

merozoite invasion (2-4, 8, 9). The PfRH family comprises of five members - PfRH1, PfRH2a,

74

PfRH2b, PfRH4 and PfRH5 that bind with either sialic acid dependent or sialic acid independent

75

erythrocyte receptors (10-22). However, most of these proteins are not essential for erythrocyte

76

invasion and can be genetically disrupted (4, 8, 9) with the exception of PfRH5 (22).

77

PfRH5 (Accession Number: XP_001351544; PlasmoDB ID: PF3D7_0424100) was first

78

identified by genetic mapping as a key determinant of species specific erythrocyte invasion (21). 3

79

Genetic analysis of the progeny of a P. falciparum cross between two parental clones 7G8 x GB4

80

had mapped the PfRH5 gene on chromosome 4 as the locus responsible for mediating invasion

81

of Aotus nancymaae erythrocytes as well as infectivity of Aotus monkeys (21). It was also

82

demonstrated that PfRH5 is an erythrocyte binding ligand in which single point mutations

83

critically affected the specificity of its binding with Aotus erythrocytes (21). Recently, PfRH5

84

has also been shown to play a role in the invasion of both owl monkey and rat erythrocytes by P.

85

falciparum (23). Further, PfRH5 was found to be unique in being the only erythrocyte binding

86

ligand among the EBA/PfRH families that is essential for the parasite as it cannot be genetically

87

knocked out (22), suggesting a crucial role in erythrocyte invasion. PfRH5 is also exceptional

88

compared to other PfRH homologues as it is smaller in size (63 kDa) and lacks a transmembrane

89

domain (21, 22). PfRH5 has been shown to be localized on the merozoite surface in association

90

with another parasite molecule, PfRipr (P. falciparum PfRH5 interacting protein) (24). While,

91

PfRH proteins are differentially expressed among different P. falciparum clones that exhibit

92

phenotypic variation in their invasion properties (11, 13, 16-19), the expression of PfRH5 was

93

found to be consistent among these parasite clones (21, 22).

94

Recently, PfRH5 was reported to bind with the CD147 IgG super family member,

95

Basigin (BSG) on the erythrocyte surface (25). For this study, a mutated version of PfRH5 was

96

produced in mammalian HEK293 cells as a biotinylated fusion protein with the CD4 domains

97

3+4(CD4d3+4) of rat origin, which was found to bind with a pentamer of Basigin (25). The

98

mutations were necessary to produce a non-glycosylated protein in the mammalian cells similar

99

to that of the native parasite protein, which like P. falciparum native proteins remains essentially

100

unglycosylated. The significance of the PfRH5-BSG interaction was highlighted by the 4

101

demonstration that anti-BSG antibodies blocked erythrocyte invasion by a large number of P.

102

falciparum clones that were known to exhibit different invasion phenotypes (25). However, no

103

data was reported in this elegant study on the interaction of native PfRH5 with Basigin.

104

A heterologous prime-boost strategy based on the adenoviral/MVA viral vector platform

105

was used to generate anti-PfRH5 antibodies that efficiently inhibited erythrocyte invasion by

106

multiple heterologous P. falciparum clones (26). It has also been recently reported that

107

antibodies against a fusion protein comprising of a mutated PfRH5 with the (d3+4) domains of

108

the rat CD4 protein exhibited potent inhibition of erythrocyte invasion (27). This study further

109

substantiated the importance of the PfRH5-BSG interaction during erythrocyte invasion and

110

strongly supported PfRH5 as a blood stage vaccine candidate. However, polymorphisms in

111

PfRH5 as well as other P. falciparum adhesins have been shown to induce changes in their

112

receptor specificity (2, 3, 21, 23) and could also possibly alter native structure. Thus, the

113

production of a functionally active recombinant wild-type full-length PfRH5 protein, which

114

would elicit similar potent invasion inhibitory antibodies, in a cell based expression system that

115

could be scalable for mass production still remained a challenge not only from a vaccine

116

perspective but also to facilitate the basic structure-function analysis of PfRH5.

117

Also considering that the target population of a prophylactic malaria vaccine is young

118

infants and children, it is important to test different expression platforms so as to identify the

119

safest and most efficacious antigen or delivery mechanism that would be most feasible to

120

administer as a vaccine for mass immunization. In this regard, the subunit vaccine approach

121

based on formulations of recombinant proteins and adjuvant pose a safe and effective platform

122

for administering a vaccine for large masses. However, the test here lies in being able to produce 5

123

the recombinant protein with a structural integrity that yields potent neutralizing antibodies

124

against the respective pathogens.

125

Expression in Escherichia coli provides a cost-effective method for production of

126

recombinant proteins for use as biologics and vaccines. Previous reports of production of

127

recombinant proteins against PfRH5 in E. coli have focused on expressing smaller fragments of

128

143-168 amino acids (22, 28). Both these recombinant fragments failed to elicit invasion

129

inhibitory antibodies (22, 28). This is consistent with the recent report using the Adeno-MVA

130

prime boost approach that demonstrated potent invasion inhibitory antibodies only against full-

131

length PfRH5 and not against the 168 amino acid fragment of PfRH5 (26).

132

In light of these reports, in our study we have demonstrated that the full-length wild-type

133

PfRH5 recombinant protein produced in E. coli was efficacious in eliciting potent invasion

134

inhibition consistent with that observed with the viral vector delivery platform (26) or with the

135

ratCD4(d3+4) fusion construct (27). In the current report, we have successfully produced full-

136

length PfRH5 as a recombinant protein in E. coli that exhibits specific erythrocyte binding

137

activity similar to that of the native PfRH5 parasite protein. We also demonstrated that anti-

138

Basigin antibodies blocked the erythrocyte binding of both native and recombinant PfRH5

139

proteins, further confirming that Basigin acts as their erythrocyte receptor. Our recombinant

140

PfRH5 elicited potent strain-transcending invasion inhibitory antibodies that blocked a number

141

of heterologous parasite clones. Importantly, the PfRH5 antibodies produced additive invasion

142

inhibition in combination with antibodies against other key merozoite antigens. Thus, our study

143

strongly supports the development of PfRH5 based antigen combinations as malaria vaccine

144

candidates. 6

145

Materials and Methods:

146 147

Ethics statement

148

The animal studies described below were approved by the International Centre for

149

Genetic Engineering and Biotechnology (ICGEB) Institutional Animal Ethics Committee

150

(IAEC) (reference no.MAL-51) according to the guidelines of the Department of Biotechnology,

151

Government of India.

152 153

Cloning, expression and purification of the full-length recombinant PfRH5

154

The 1500 bp PfRH5 gene that encodes the 500 amino acid full length parasite protein

155

excluding the signal sequence (rPfRH563, Asp27-Gln526) was PCR amplified from the genomic

156

DNA of the P. falciparum clone 3D7 using the following primers, RH5-Fwd: 5’-

157

ATATATAATTCATATGAATGCAATAAAAAAAACGAAGAAT-3’

158

AGCACTCGAGTTGTGTAAGTGGTTTATTTTTTT-3’. The PCR product encoding rPfRH563

159

were digested with Nde I and Xho I (New England Biolabs, Beverly, MA) and inserted

160

downstream of the T7 promoter in the E. coli expression vector, pET-24b (Novagen, San Diego,

161

CA) with a C-terminal 6-histidine (6-His) tag to obtain the plasmid pPfRH5-pET24b.

162

Sequencing of the ligated plasmid confirmed the correct sequence of the PfRh5 gene and that the

163

insertions were in the correct reading frame.

and

RH5-Rev:

5’-

164

E. coli BL21(DE3) was transformed with pPfRH5-pET24b and was used to produce the

165

recombinant protein rPfRH563. Transformed E. coli BL21(DE3) were cultured in superbroth at

166

37°C and later were induced with 1mM IPTG when the OD600 was around 0.8-0.9. Cells were 7

167

harvested by centrifugation at 3000g, after 4 hours of induction at 37°C. Cell pellets were lyzed

168

by sonication and rPfRH563 was found to be expressed as inclusion bodies. The inclusion bodies

169

were washed, collected by centrifugation at 15000g and solubilized in a buffer containing 20

170

mM Tris pH 8.0, 6 M Guanidium-HCl, 300 mM NaCl, 10 mM Imidazole and 5 mM beta

171

mercaptoethanol. rPfRH563 was purified from solubilized inclusion bodies by metal affinity

172

chromatography using the Ni-NTA (nitrilotriacetic acid) resin. Metal affinity purified rPfRH563

173

was refolded in redox conditions by rapid dilution (30-fold) in an MES based buffer pH 6.5

174

comprising of 1 mM GSH (reduced Glutathione), 0.1 mM GSSG (oxidized Glutathione) and 440

175

mM sucrose. The refolded protein was dialyzed against 25 mM MES (pH 6.5), 200mM sucrose

176

and further purified to homogeneity by cation exchange chromatography using an SP-sepharose

177

column (GE Healthcare, Piscataway, NJ). The dialyzed protein was loaded on the SP-sepharose

178

column and eluted with an increasing concentration of NaCl (0-1 M) in the MES based buffer,

179

pH 6.5. The purified recombinant rPfRH563 protein was characterized by SDS-PAGE, Western

180

blotting, Edman degradation, Mass spectrometric (LC-MS) analysis (Orbitrap VELOS PRO,

181

Thermofisher Scientific), Reverse phase RP-HPLC (C8 column; Waters) and Size exclusion

182

chromatography (Superdex 75 10/300 GL; GE Healthcare).

183 184

Animal Immunization and antibody generation

185

Animal immunizations and total IgG purification were done as reported previously (29).

186

Briefly, five rats and three rabbits were immunized intramuscularly with 50 µg and 100 µg

187

rPfRH563, respectively. The rPfRH563 protein was emulsified with complete Freund’s adjuvant

188

(Sigma, St. Louis, MO) for immunization on day 0 followed by two boosts emulsified with 8

189

incomplete Freund’s Adjuvant on day 28 and 56. The sera were collected on day 70. Antibody

190

levels were measured by ELISA.

191

For co-immunogenicity, a group of six BALB/c mice were immunized with antigen

192

mixtures PfF2+PfRH5+PfAARP, PfRH2+PfRH5+PfAARP as well as individual antigens

193

emulsified with complete Freund’s adjuvant on day 0 followed by two boosts emulsified with

194

incomplete Freund’s adjuvant on days 28 and 56. 17 µg of each antigen was immunized in each

195

mouse, whether used alone or as a co-immunization triple antigen mixture (total 51 µg).

196

Terminal bleeds were collected on Day 70. Sera were tested for antibody titers and specific

197

recognition of each recombinant protein by ELISA. For the GIA, the sera from the six mice in

198

each group were pooled for IgG purification.

199

Total IgG was purified from the mice, rat or rabbit sera using Protein G affinity column

200

(GE Healthcare, Uppsala, Sweden), dialyzed with RPMI medium and further tested in invasion

201

inhibition assays as described below. As an adjuvant negative control, we also had raised

202

antibodies in mice, rats and rabbits against a non-related peptide (KESRAKKFQR

203

KHITNTRDVD from human pancreatic RNase) that was also formulated with the same adjuvant

204

(CFA/IFA) and injected in animals with the same schedule used for raising the PfRH5 antibodies

205

as described above.

206 207

Erythrocytes and enzymatic treatment:

208

Packed RBCs were procured from the Rotary Blood Bank (Tughlaqabad), New Delhi,

209

India. Erythrocytes were washed in RPMI, and stored at 50% haematocrit. Enzymatic treatments

210

of erythrocytes were done as stated previously (16, 20, 29). 9

211

Erythrocyte binding assays:

212

Soluble parasite proteins were obtained from 3D7 culture supernatants as described

213

previously (16, 20, 29). Briefly, 500µl of culture supernatant were incubated with 100µl packed

214

volume of human erythrocytes at 37 C, following which the suspension was centrifuged through

215

Dibutyl phthalate (Sigma). The supernatant and oil were removed by aspiration and bound

216

parasite proteins were eluted using 1.5M NaCl. For rPfRH563 binding, 0.04µM of rPfRH5 was

217

incubated with 100µl packed volume of human erythrocytes at 37 C in similar erythrocyte

218

binding assays as described above. The eluate fractions were analyzed for the presence of native

219

PfRH5 and rPfRH563 by immunoblotting using anti-rPfRH563 antibodies.

220 221

Invasion inhibition assays

222

Invasion inhibition assays were performed as described previously (16, 29) using total

223

IgG purified from the sera of different animals immunized with PfRH5. Briefly, schizont-stage

224

parasites at an initial parasitemia of 0.3% at 2% hematocrit were incubated with purified total

225

IgG for one cycle of parasite growth (40 h post invasion). The parasite-infected erythrocytes

226

were stained with ethidium bromide dye and measured by FACS as described previously (16,

227

29). The percent invasion inhibition for each immune IgG was calculated with respect to the

228

control pre-immune IgG from the same animal. As another negative control, we used immune

229

IgG raised against a non-related peptide from human pancreatic RNase that was also immunized

230

with the same freund’s adjuvant (CFA/IFA) used for raising the PfRH5 antibodies. The results

231

represent the average of three independent experiments performed in duplicate and the error bars

232

represent the standard error of the mean. Statistical significance was calculated using the 10

233

Student’s t- test (Graph Pad Prism software, version 6.03). P values < 0.05 were considered

234

statistically significant.

235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 11

255

Results

256

Expression of the full-length recombinant PfRH5 protein and generation of specific PfRH5

257

antibodies

258

A 500 amino acid sequence (Asn27-Gln526) of PfRH5 from the P. falciparum clone

259

3D7, comprising of the full-length protein excluding the signal peptide, was chosen for

260

recombinant protein expression in E. coli (Fig. 1A). The recombinant 63kDa PfRH5 (rPfRH563)

261

was expressed with a C-terminal 6-His tag. In E. coli, the rPfRH563 protein got expressed in

262

inclusion bodies, which were then refolded after purification under denaturing conditions by

263

metal affinity chromatography. rPfRH563 was further purified to homogeneity using ion-

264

exchange chromatography (Fig. 1B).

265

Recombinant rPfRH563 comprises of 6 cysteines that could lead to three potential

266

disulphide linkages. Initial SDS-PAGE analysis of the purified protein on a 12% gel under both

267

reducing and non-reducing conditions did not reflect a mobility shift (Fig. 1B). However, when

268

the rPfRH563 protein was run on the same gel for a longer period of time such that the 35 KDa

269

pre-stained marker protein reached the end of the gel, a mobility shift could be detected (Fig.

270

S1A). The SDS-PAGE analysis was repeated along with a highly cysteine rich recombinant

271

protein (rEBA-175 RII) that has 24 cysteines leading to 12 potential disulphide bonds (Fig. S1B,

272

S1C). rEBA-175 RII exhibited a significant mobility shift compared to rPfRH563 that showed no

273

shift when run normally on the 12% SDS-PAGE gel (Fig. S1B). On running the gel for a longer

274

time, the mobility shift increased for rEBA-175 RII and became detectable for rPfRH563 (Fig.

275

S1C). Edman degradation analysis of the full-length rPfRH563 protein yielded an N-terminal

276

sequence (MNAIKK) that matched with the protein sequence of the PfRH5 native parasite 12

277

protein after the signal sequence from amino acid Asn 27 onwards (Fig. S1C). Recombinant

278

rPfRH563 was identified in immunoblots using a specific anti-His tag antibody (Fig. 1C)

279

confirming expression of the full-length 500 amino acid protein with the C-terminal His-tag.

280

While, the SDS-PAGE analysis of the rPfRH563 protein showed a highly pure protein preparation

281

with the predominant band at 63 kDa, traces of a smaller 45 kDa protein were also faintly

282

visible. These two protein bands were excised from the gel and subjected to trypsin digestion

283

followed by LC-MS (liquid chromatography-mass spectrometry) analysis (Orbitrap VELOS

284

PRO, Thermofisher scientific). The LC-MS analysis revealed a large number of unique high

285

scoring peptides for both proteins (32 peptides for 63 kDa protein; 31 peptides for 45 kDa

286

protein) that confirmed the identity of both proteins to be PfRH5 (Table S1, S2). The detection of

287

a smaller 45 kDa protein is consistent with previous reports on the production of recombinant

288

PfRH5 in HEK293 cells (27) and that observed in the parasite lysate (22). We further analyzed

289

the recombinant protein on Size exclusion chromatography (SEC) and confirmed that our

290

recombinant protein eluted at the expected molecular size with respect to the BSA standard

291

protein, which also has a similar molecular weight (66 kDa) (Fig. S1D). The SEC profile also

292

showed that our recombinant protein was primarily in a monomeric state. The recombinant

293

rPfRH563 protein was further analyzed using Reverse Phase HPLC (RP-HPLC) that showed a

294

single symmetrical peak reflecting a highly pure protein preparation (Fig. S1E).

295

Rats and rabbits were immunized with rPfRH563 to raise PfRH5-specific antibodies. High

296

titer antibodies against rPfRH563 were detected in both rats and rabbits with end points observed

297

at dilutions of 1:320,000 (data not shown). The specificity of the PfRH5 antibodies was analyzed

298

by immunoblotting studies to detect the PfRH5 native protein in parasite lysates and localization 13

299

studies in merozoites by immunofluorescence super resolution confocal microscopy (N-SIM,

300

Nikon, Japan). Consistent with previous reports, native parasite PfRH5 was detected in

301

immunoblots at the expected size for the full-length protein (63kDa) only in the schizont stages

302

and not in early rings or trophozoite stages (Fig. S2A, S2B). PfRH5 has been reported to be

303

localized in the rhoptry bulb by immunoelectron microscopy (22). With our antibodies, PfRH5

304

was also found to co-localize with the known rhoptry bulb protein, PTRAMP (30) (Fig. S3A).

305

On the other hand, there was no co-localization observed with the rhoptry neck protein, PfRH2

306

(Fig. S3B) or the micronemal protein, PfEBA-175 (Fig. S3C). Our data was consistent with

307

previous reports (22) and confirmed the specificity of our PfRH5 antibodies.

308 309

Recombinant PfRH5 exhibits specific erythrocyte binding activity

310

Standard erythrocyte binding assays were performed with parasite culture supernatant

311

(3D7) incubated with human erythrocytes as described previously (16, 20, 29). Native PfRH5

312

parasite protein (63kDa) has been reported to be processed into smaller fragments of 45 kDa and

313

28 kDa (21, 22). The full-length native PfRH5 and its processed fragments bind erythrocytes

314

with the same specificity (21, 22, 28). In our assay, we prepared 3D7 culture supernatants and

315

observed that both the native 63 kDa full-length PfRH5 protein and the 45 kDa processed protein

316

bound erythrocytes in a sialic acid independent, trypsin and chymotrypsin resistant manner (Fig.

317

2A), which is consistent with previous reports (21, 22). While, in this culture supernatant we did

318

not observe the 28 kDa fragment, in another culture supernatant preparation in which the

319

parasites were incubated for a longer period of time, we detected the 28 kDa fragment and

320

observed it to bind erythrocytes with the same specificity (Fig. S2C). Culture supernatants are 14

321

prepared in the absence of any protease inhibitors as they would impede parasite egress itself and

322

this does lead to the possibility of parasite proteins undergoing proteolytic cleavage yielding

323

fragments of different sizes (21, 22, 28).

324

The full-length, wild type PfRH5 recombinant protein, rPfRH563, expressed in E. coli

325

also exhibited an erythrocyte binding specificity that matched with that of the native parasite

326

protein (Fig. 2B). rPfRH563 specifically bound erythrocytes in a sialic acid independent, trypsin

327

and chymotrypsin resistant manner (Fig. 2B). Since, the native and recombinant PfRH5 proteins

328

bound erythrocytes treated with each of the three enzymes (neuraminidase, trypsin,

329

chymotrypsin), we also tested the effect of proteinase K treatment and found that binding of both

330

native and recombinant PfRH5 was sensitive to proteinase K (Fig.2A, 2B, S2C)

331

As controls, both native PfEBA-175 from the parasite culture supernatant and

332

recombinant PfF2, the receptor-binding domain of PfEBA-175, were found to bind erythrocytes

333

in a sialic acid dependent, trypsin sensitive and chymotrypsin resistant manner (Fig. 2C, 2D,

334

S2D) consistent with previous reports (31-33). In addition, no bound proteins were detected

335

when the erythrocytes were incubated with PBS alone (Fig. 2), thus confirming that no non-

336

specific erythrocyte protein was being detected in our assay and that the binding of PfRH5 or

337

PfEBA-175 was specific.

338 339

Antibodies against recombinant PfRH5 and Basigin block the erythrocyte binding activity

340

of native PfRH5

341

After demonstrating that rPfRH563 specifically bound erythrocytes, we determined

342

whether anti-rPfRH563 antibodies could block the erythrocyte binding of native PfRH5 parasite 15

343

protein. We demonstrated that total IgG purified from the sera of rabbits immunized with

344

rPfRH563 blocked the erythrocyte binding of native PfRH5 (Fig. 3A). Total IgG containing anti-

345

PfRH5 antibodies blocked binding of both the native and recombinant PfRH5 proteins with

346

erythrocytes in a dose-dependent manner (Fig. 3A, 3B). At a total IgG concentration of 200

347

µg/ml, the anti-PfRH5 IgG potently blocked the erythrocyte binding of both the native and

348

recombinant PfRH5 protein, whereas even at a concentration of 800 µg/ml the PfRH5 IgG had

349

no effect on the erythrocyte binding of another parasite ligand, PfEBA-175 (Fig. 3C) or its

350

recombinant receptor binding domain, PfF2 (Fig. 3D). This result clearly demonstrated that

351

PfRH5 antibodies specifically recognized only PfRH5 and further abrogated its interaction with

352

the erythrocyte surface.

353

As described earlier, the erythrocyte receptor of PfRH5 was recently identified as the

354

CD147 erythrocyte surface molecule, Basigin (BSG) (25). However, this elegant study had

355

demonstrated this interaction with recombinant PfRH5 and not the native parasite protein. We

356

tested the invasion inhibitory activity of the anti-BSG monoclonal antibodies (TRA-1-85; R&D

357

Systems, USA) and found them to potently inhibit invasion with 90% inhibition observed at a

358

concentration of 2.5µg/ml (Fig. 4A) consistent with the previous study (25). Further, we tested

359

the ability of the anti-BSG monoclonal TRA-1-85 antibodies to block the erythrocyte binding of

360

recombinant protein, rPfRH563 and native PfRH5 from parasite culture supernatants. Anti-BSG

361

monoclonal TRA-1-85 antibodies potently blocked erythrocyte invasion from a minimum

362

concentration of 2.5µg/ml and from the same concentration, the binding of the native PfRH5

363

protein was also observed to be significantly reduced in a dose dependent manner (Fig. 4B). 10

364

µg/ml of the anti-BSG monoclonal TRA-1-85 antibody completely abrogated the binding of both 16

365

the native PfRH5 protein (Fig. 4B) and rPfRH563 (Fig. 4C). As a control, anti-glycophorin A

366

monoclonal antibodies (Sigma-Aldrich) had no effect on the binding of the PfRH5 parasite

367

protein (Fig. 4B, 4C) suggesting that the anti-BSG TRA-1-85 monoclonal antibodies were acting

368

in a specific manner. This result substantiates the previous finding on PfRH5-BSG and

369

demonstrates for the first time the interaction between BSG with the native PfRH5 parasite

370

protein.

371 372

Antibodies against recombinant PfRH5 potently block erythrocyte invasion by multiple P.

373

falciparum clones

374

PfRH5 is the only parasite ligand among the EBA/PfRH families that is essential for

375

erythrocyte invasion (21, 22). We thus, compared the invasion inhibitory activity of PfRH5

376

antibodies with that of antibodies raised against five other parasite ligands from our antigen

377

portfolio – PfRH1, PfRH2, PfRH4, PfAARP (P. falciparum Apical Asparagine Rich Protein),

378

PfF2 (F2: receptor binding domain of PfEBA-175) as described previously (16, 20, 29, 32, 34).

379

The total rabbit IgGs (0.5-10 mg/ml) purified from the sera of rabbits individually immunized

380

with one of the six antigens were tested in standard one-cycle in vitro invasion inhibition assays

381

(Fig. 5A) as described in our previous report (29). The invasion inhibition of the six antibodies

382

was tested against the P. falciparum clone 3D7, which invades using both sialic acid dependent

383

and independent pathways (Fig. 5A). All antibodies exhibited a dose dependent inhibition that

384

suggested a specific effect (Fig.5A). The invasion inhibition for each immune IgG was

385

calculated with respect to the pre-immune IgG obtained from the same rabbit. In addition, as

386

another negative control, we tested the invasion inhibitory activity of purified rabbit total IgG 17

387

against a non-related peptide from a human pancreatic ribonuclease (HPR) that was also

388

formulated with the same freund’s adjuvant. The anti-HPR IgG failed to exhibit any invasion

389

inhibition at the maximum concentration of 10 mg/ml (Fig. 5A). The anti-HPR negative control

390

has been tested in each assay reported in the current study.

391

Among all six antibodies, PfRH5 total IgG was found to elicit maximum inhibition of

392

erythrocyte invasion with ~ 83% inhibition at 10 mg/ml followed by PfAARP IgG (51%, 10

393

mg/ml) and PfRH2 IgG (49%, 10 mg/ml) (Fig. 5A). PfRH5 total IgG exhibited an invasion

394

inhibition of 63% at 5 mg/ml and 52% at 3.3 mg/ml (Fig. 5A). The variation in inhibitory

395

activity among the different antibodies could not be attributed to any disparity in their antibody

396

titers. The end point antibody titers against all the six proteins immunized in rabbits were in the

397

range of 1:320,000 (data not shown). As mentioned above, we had raised antibodies against

398

PfRH5 in three rabbits that had shown equivalent end point titers in the range of 1:320,000. The

399

purified total IgG from the other two rabbits also exhibited a potent invasion inhibitory activity

400

against the P. falciparum clone 3D7 (Fig. S4A) similar to that observed with the anti-PfRH5 IgG

401

from rabbit 1 (Fig. 5A).

402

We further analyzed the strain-transcending invasion inhibitory activity of the PfRH5

403

antibodies (rabbit 1) against five P. falciparum strains that originate from different regions of

404

the world and express different polymorphic variants of the PfRH5 parasite protein (Table S3)

405

(21). In addition, these P. falciparum strains exhibit a variation in their invasion phenotype by

406

utilizing different ligand receptor interactions and pathways for invading human erythrocytes

407

(Table S3) (2-4,29 33,35). The invasion phenotypes of the P. falciparum clones are classified in

408

literature on the basis of their invasion sensitivity to enzymatic treatments of the target 18

409

erythrocytes (2-4, 29, 33, 35). Thus, the five parasite clones represent diversity both at the level

410

of PfRH5 antigenic polymorphisms as well as the phenotypic variation in the invasion properties

411

of the parasite clones (Table S3). The strains 3D7, HB3 and 7G8 are known to invade through

412

sialic acid independent pathways (3, 33, 35), whereas Dd2, MCamp are completely dependent on

413

sialic acids for erythrocyte invasion (3, 33, 35). The anti-PfRH5 antibodies (rabbit 1) were found

414

to potently inhibit erythrocyte invasion of all five P. falciparum clones (Fig. 5B). The purified

415

PfRH5 total IgG exhibited 81-85% invasion inhibition among the five parasite clones at a

416

concentration of 10 mg/ml (Fig.5B), which is comparable with the maximum inhibition that we

417

recently reported with an antibody combination against three antigens, PfAARP+PfRH2+PfF2

418

(29). The invasion inhibitory activity was dose dependent as reported earlier with an inhibition of

419

60-71% at 5 mg/ml, 49-59% at 3.3 mg/ml and 34-46% at 2.5 mg/ml (Fig. 5B). Similar potent

420

strain-transcending invasion inhibition was also observed with total IgG purified from the sera of

421

the other two rabbits immunized with rPfRH563 (data not shown). Our PfRH5 antibodies

422

exhibited a 50% inhibition of erythrocyte invasion at a total IgG concentration (EC50) of ~3

423

mg/ml, which is consistent with previous studies that have reported EC50 values for anti-PfRH5

424

IgG obtained from different rabbits within a concentration range of 0.7-4 mg/ml against multiple

425

P. falciparum strains (26, 27, 36).

426

The purified total IgG from five individual rats also exhibited a potent dose dependent

427

invasion inhibition of the P. falciparum clone 3D7 with the maximum inhibition of around 72-

428

77% at 10 mg/ml concentration (Fig. S4B). The specificity of the invasion inhibition by the

429

PfRH5 rat IgG was validated by reversing it with the addition of the recombinant rPfRH563 in the

430

assay. Addition of rPfRH563 at a concentration of 50µg/ml significantly reduced the invasion 19

431

inhibition observed with 5 mg/ml PfRH5 IgG by 80% (Fig. S4C). On the other hand, addition of

432

PfF2 at the same concentration had no effect on the invasion inhibitory activity of the PfRH5

433

antibodies (Fig. S4C).

434

Thus, not only within the PfRH family but also compared to other major parasite ligands,

435

PfRH5 is a potent target of specific antibody mediated blockade of erythrocyte invasion and

436

appears to have a dominant role in the erythrocyte invasion process. Therefore, antibodies raised

437

against a functional recombinant protein expressed in E. coli representing the wild-type full-

438

length sequence of PfRH5 have proven to be as potent in blocking parasite invasion as reported

439

earlier with antibodies raised by adenoviral vectors (26,36) or the mutated PfRH5-ratCD4(d3+4)

440

fusion protein (27).

441 442

PfRH5 based antibody combinations produce an additive inhibition of erythrocyte invasion

443

by P. falciparum

444

In a recent report, we identified a potent antigen combination (PfAARP+PfRH2+PfF2)

445

that elicits strain-transcending invasion inhibitory antibodies (29). Through this study, we had

446

demonstrated that triple antibody combinations were more efficacious in inhibiting erythrocyte

447

invasion compared to double antibody combinations (29). This study did not include PfRH5, so

448

we wanted to now analyze whether PfRH5 based triple antibody combinations would also be

449

effective in producing additive inhibition of invasion at lower individual IgG concentrations. In

450

this regard, we tested the invasion inhibition of ten PfRH5 based triple antibody combinations

451

that comprised of purified total IgG (3.3 mg/ml each antigen; Total: 10 mg/ml) against PfRH5

452

(rabbit 1) and two other antigens from our portfolio (PfRH1, PfRH2, PfRH4, PfAARP, PfF2) 20

453

generated previously (29). The invasion inhibition was first assessed against two P. falciparum

454

strains (3D7, Dd2) (Fig. 6) and then the six most efficacious antibody combinations were further

455

analyzed against a total of five P. falciparum strains (3D7, Dd2, 7G8, MCamp, HB3) (Fig. 7) to

456

ascertain whether the antibody combinations elicited strain-transcending activity.

457

Individually, the six antibodies at 3.3 mg/ml exhibited a broad range of invasion

458

inhibition of 3-54% against all five parasite clones (Fig. 6, 7). As described above, anti-PfRH5

459

IgG displayed maximum inhibition against all strains (44-54%), followed by anti-PfAARP IgG

460

(17-33%) (Fig. 6, 7). Consistent with previous findings, anti-PfRH1 and anti-PfF2 IgG were

461

more efficacious in blocking the sialic acid dependent clones- Dd2 (24-36%), MCamp (15-17%)

462

and not the sialic acid independent clones – 3D7 (8-12%), 7G8 (5-10%) and HB3 (5-10%)

463

(Fig.6, 7). Similarly, PfRH2 IgG and PfRH4 IgG efficiently blocked only the sialic acid

464

independent clones and not the sialic acid dependent clones, in which they are poorly expressed.

465

The different anti-PfRH5 based antibody combinations displayed potent inhibition of

466

erythrocyte invasion by the 3D7 parasite clone (Fig. 6A) with the maximum inhibition observed

467

with three antibody combinations, PfAARP+PfRH2+PfRH5 (82%), PfF2+PfRH5+PfAARP

468

(82%) and PfF2+PfRH2+PfRH5 (79%). In line with the utilization and expression of the

469

different PfRH ligands, we observed that the maximum inhibition with the Dd2 parasite clone

470

was by the antibody combinations, PfRH5+PfF2+PfAARP (81%), PfRH5+PfRH1+PfAARP

471

(79%) and PfRH5+PfRH1+PfF2 (78%) (Fig. 6B). This is consistent with the higher expression

472

and utilization of the sialic acid binding ligands (PfRH1, PfF2 and PfAARP) for erythrocyte

473

invasion by sialic acid dependent parasite clones such as Dd2. We further tested invasion

474

inhibition of the six best PfRH5 based antibody combinations against five P. falciparum clones 21

475

that originate from diverse regions of the world and exhibit different invasion phenotypes as

476

well.

477

The six antibody combinations were potent in blocking invasion of all five parasite

478

clones

with

the

antibody

combinations,

PfRH5+PfF2+PfAARP

(77-82%),

479

PfRH5+PfRH2+PfAARP (71-83%), and PfF2+PfRH2+PfRH5 (72-79%) eliciting the most

480

efficacious strain-transcending invasion inhibitory activity (Fig. 7). We also tested as a control

481

the PfAARP+PfF2+PfRH2 antibody combination, which we had previously reported (29) as our

482

most efficacious antibody combination (Fig. 7). While this antibody combination does elicit

483

strain-transcending activity, the combination PfRH5+PfF2+PfAARP appeared to be slightly

484

more efficient in its overall strain-transcending activity against all the five P. falciparum clones,

485

which is consistent with the observation that PfRH5 antibodies individually were observed to be

486

most efficient among the different parasite ligands in blocking erythrocyte invasion. However,

487

the difference in invasion inhibitory activity between the PfRH5+PfF2+PfAARP and

488

PfAARP+PfF2+PfRH2 antibody combinations was not statistically significant (p>0.05).

489 490

Co-immunized antigen mixtures also elicit potent strain-transcending invasion inhibitory

491

antibodies

492

After evaluating antibody combinations that were mixed in vitro for their invasion

493

inhibitory activity, we wanted to test whether the most potent combination identified would elicit

494

similar invasion inhibitory antibodies when co-immunized together as a single formulation. In

495

this next step, the PfRH5 based triple antigen mixtures (PfRH5+PfRH2+PfAARP and

496

PfRH5+PfF2+PfAARP) were used to immunize mice (BALB/c). The individual antigens in each 22

497

combination were also used to immunize mice separately. All antigens were formulated with the

498

adjuvant, CFA/IFA. The ELISA results (OD492) showed that the antibody titers (end point

499

1:320,000) against each protein immunized individually were not significantly altered when

500

immunized as a mixture with the two other antigens (Fig. S5). The immunogenicity curves for

501

PfRH5, PfRH2, PfF2 and PfAARP were identical and overlapping whether the antigens were

502

immunized alone or in their respective combinations (Fig. S5). Thus, our recombinant antigens

503

including rPfRH563 were immunogenic and did not elicit any significant immune interference

504

when immunized in combination.

505

Consistent with the invasion inhibition observed with the antibody combinations

506

physically added in vitro, the antibodies raised against the antigen mixtures were highly potent

507

and equally efficient in inhibiting erythrocyte invasion (Fig. 8, S6). The antibodies against the

508

two antigen mixtures displayed 71-79% and ~86% inhibition of the P. falciparum clones (3D7,

509

Dd2) at concentrations of 5 and 10 mg/ml, respectively (Fig. 8A, 8B). The antibodies displayed a

510

similar potent inhibition of three more parasite clones at an IgG concentration of 10 mg/ml (Fig.

511

S6). Thus, antibodies raised against co-immunized antigen mixtures were as potent in efficiently

512

blocking erythrocyte invasion as antibodies physically combined (Fig. 6, 7) in the in vitro

513

invasion inhibition assays.

514 515 516 517 518 23

519

Discussion

520

Parasite neutralizing antibodies that block P. falciparum erythrocyte invasion is one of

521

the key effector mechanisms known to mediate immunity against blood-stage malaria parasites

522

and is the fundamental basis for the development of blood-stage malaria vaccines. The ability to

523

impede erythrocyte invasion by P. falciparum merozoites can be quantitated in an in vitro

524

invasion inhibition or growth inhibition assay (GIA) that has been widely reported in the field. A

525

significant association of invasion inhibition measured in vitro with a reduced risk of malaria has

526

also been reported (37, 38) and thus, in vitro invasion-inhibition appears to be a useful surrogate

527

marker to predict the functional efficacy of antibodies induced by a blood-stage vaccine. In spite

528

of the extensive research on the parasite biology of P. falciparum, it has been difficult to

529

demonstrate potent invasion inhibitory activity with the exception of antibodies against Apical

530

Membrane Antigen-1, AMA-1 (39, 40). The challenge in generating potent invasion inhibitory

531

antibodies that have hindered the development of blood-stage malaria vaccines against P.

532

falciparum could be attributed to the enormous complexity of the parasite, which has evolved

533

redundancy in parasite ligands that enables invasion of diverse types of human erythrocytes (2-

534

4). As a result, no human erythrocyte is known to be totally refractory to invasion by P.

535

falciparum. Another level of complexity is the high level of antigenic polymorphisms that

536

mediates immune escape, which has proven to be a major impediment in developing leading

537

antigens such as AMA-1 and MSP-142 as blood stage vaccines (5-7). Both antigens have failed to

538

elicit optimal efficacy in field trials (5, 6),

539

Thus, to fully realize the potential of a candidate blood-stage antigen, it is not sufficient

540

to just show that the antigen elicits strong invasion inhibitory antibodies that block the parasite in 24

541

vitro. A good example is AMA-1, an essential parasite protein involved in the critical step of

542

junction formation during erythrocyte invasion (41, 42) , which has been demonstrated in vitro to

543

induce potent invasion inhibitory antibodies only against homologous parasite strains and not

544

against heterologous strains (39, 40). This inability has been attributed to high levels of antigenic

545

polymorphisms in AMA-1 among different parasite strains that render it’s antibodies to be

546

ineffective against heterologous P. falciparum strains (39, 43, 44). Unfortunately due to the

547

problem of inducing allele-specific immunity, AMA-1 has not yielded protection against malaria

548

in vaccine efficacy trials (7, 45).

549

Therefore, it is crucial that potent strain-transcending invasion inhibition is demonstrated

550

with the antibodies against a particular antigen that is being considered for clinical development

551

of a malaria vaccine. This problem has been reported to be overcome by targeting combinations

552

of different conserved parasite antigens or a number of different AMA-1 allelic proteins that

553

produce strong efficacious invasion inhibition in a strain-transcending manner (29, 46).

554

The disappointing results of a number of blood-stage vaccine trials has raised the concern

555

whether the in vitro invasion inhibition assay or GIA assay has any correlation with clinical

556

protection or could predict vaccine efficacy in humans (47). Unfortunately, only a few blood-

557

stage antigens have been tested in field efficacy trials. Due to the problems cited above, more

558

studies with parasite antigens that induce potent strain-transcending invasion inhibition are thus

559

required to validate the correlation of in vitro invasion inhibitory activity with clinical protection

560

in humans. It would thus be beneficial to validate the in vitro invasion inhibitory assay or GIA

561

with in vivo CHMI (controlled human malaria infection) studies using either a sporozoite or

562

blood-stage challenge model (47). However, the in vitro invasion inhibitory assay or GIA 25

563

remains as the only currently available laboratory assay to measure the functionality of

564

antibodies to inhibit erythrocyte invasion by Plasmodium merozoites. In this regard, this assay

565

has been successful in differentiating between invasion inhibitory and non-inhibitory antibodies

566

against a number of P. falciparum antigens as not all antibodies exhibit potent invasion

567

inhibitory activity. Hence, the in vitro invasion inhibition or GIA assay clearly appears to be an

568

informative assay, which in pre-clinical studies can identify and validate novel, efficacious P.

569

falciparum blood-stage targets that elicit strain-transcending invasion-inhibitory antibodies.

570

The past decade has seen a huge body of research conducted on understanding the

571

molecular basis of erythrocyte invasion and characterizing numerous antigens from the large

572

repertoire developed by P. falciparum (2-4, 8, 9). With respect to vaccine development, the goal

573

to search for promising antigens has shifted to the identification of relatively conserved antigens

574

that elicit potent strain-transcending invasion inhibitory antibodies. Among the parasite’s large

575

molecular arsenal, the PfRH proteins have been identified as key determinants of different

576

invasion pathways used by P. falciparum (2-4, 8, 9), of which PfRH5 is the only erythrocyte

577

binding ligand known to be essential for the parasite as its gene is refractory for disruption (22).

578

The importance of PfRH5 has been substantiated by the fact that antibodies against both PfRH5

579

and its erythrocyte receptor, Basigin, potently inhibit erythrocyte invasion by a number of P.

580

falciparum strains from diverse worldwide locations that also exhibit different invasion

581

phenotypes (25-27). The efficacious neutralization of heterologous parasite clones by PfRH5

582

antibodies raised first through a viral vector based prime boost strategy had demonstrated that

583

PfRH5 is a highly promising blood stage vaccine candidate (26).

584

the production of a potent functional wild-type full-length PfRH5 recombinant antigen that could 26

Our goal was to demonstrate

585

be used for the development of a subunit blood-stage malaria vaccine, which would have the

586

potential to be tested individually as well as in combination with other blood stage antigens and

587

possibly even pre-erythrocytic malaria vaccines such as RTS,S (48). RTS,S

588

advanced malaria vaccine currently being tested in a Phase III clinical trial in Africa (49, 50). It

589

is a subunit recombinant vaccine based on the CSP-HbSAg fusion protein that assembles into

590

virus-like particles (48-50).

is the most

591

In this regard, we have produced the full-length, wild type PfRH5 recombinant protein

592

without making any alterations to its sequence in a bacterial organism, E. coli. The recombinant

593

protein binds with erythrocytes with the same specificity as that of the native parasite protein and

594

anti-Basigin monoclonal antibodies specifically blocked the binding of the recombinant protein.

595

This binding was unaffected by other monoclonal antibodies against another major erythrocyte

596

receptor, glycophorin A. All these results strongly suggest that the recombinant PfRH5 was

597

produced with a conformational integrity that mimics the native parasite protein. This work

598

paves the way for a more detailed structure-function analysis of PfRH5 that would not only

599

improve our understanding of its role in the basic biology of the parasite but also help in

600

designing a more efficacious PfRH5 immunogen for vaccine development.

601

While, it was very elegantly demonstrated that PfRH5 binds with Basigin (25), this report

602

did not show any data with regard to the interaction of the native PfRH5 parasite protein and

603

based its inferences on the interaction with a pentamer of the biotinylated recombinant PfRH5-

604

ratCD4(d3+4) fusion protein. In the present study, we have demonstrated that the Basigin

605

monoclonal antibodies at the same concentration at which they blocked erythrocyte invasion also

606

completely inhibited the erythrocyte binding of the native PfRH5 parasite protein obtained from 27

607

culture supernatants. Our data strongly supports the previous study that the native PfRH5

608

parasite protein binds with the erythrocyte surface molecule, Basigin.

609

Further, most importantly, the antibodies raised against recombinant PfRH5 specifically

610

inhibited erythrocyte invasion of heterologous P. falciparum strains with a potent efficiency as

611

high as 85% and 71% at total IgG concentrations of 10 mg/ml and 5 mg/ml, respectively. PfRH5

612

antibodies were observed to be most potent among a pool of antibodies (0.5-10 mg/ml total IgG)

613

tested against six antigens in our portfolio that included four PfRH proteins (PfRH1, PfRH2,

614

PfRH4, PfRH5), PfEBA-175 (PfF2) and PfAARP (29). The normal physiological total IgG

615

concentration in human serum is in the range of 10-14 mg/ml (51). Thus, we have analyzed our

616

antibodies at a maximum total IgG concentration of 10 mg/ml, which is on the lower end of the

617

human physiological concentration. The specific component of the anti-PfRH5 IgG within the

618

total serum IgG (10 mg/ml) would be a much smaller fraction and should be achievable through

619

a human vaccine. In addition, PfRH5 antibodies exhibited the maximum cross-strain neutralizing

620

activity as they were efficacious in blocking a number of P. falciparum clones that display

621

phenotypic variation in their invasion pathways. Our invasion inhibition results are consistent

622

with previous reports of PfRH5 antibodies raised through viral vectors (26) or against the

623

PfRH5-ratCD4(d3+4) fusion protein (27). In our assays, the rPfRH563 antibodies exhibited an

624

EC50 of ~ 3 mg/ml, whereas previous studies with viral vectors and PfRH5-ratCD4(d3+4) fusion

625

protein have shown an EC50 with anti-PfRH5 IgG obtained from different rabbits at a broad

626

concentration range of 0.7-4 mg/ml against multiple P. falciparum strains (26, 27, 36). The

627

difference in invasion inhibitory activity could be attributed to the different nature of the PfRH5

628

immunogen used in the studies as well as the fact that different animals were used in the studies 28

629

to raise the PfRH5 specific antibodies. Outbred animals are known to exhibit large differences in

630

their immune responses and especially when located in different geographical locations.

631

As stated previously, achieving highly potent strain-transcending invasion inhibitory

632

antibodies against conserved functional domains of parasite antigens involved in invasion may

633

be the key to the development of an effective blood stage malaria vaccine. This has been difficult

634

to achieve by targeting single antigens with the exception of PfRH5 as reported here and in

635

previous reports. Earlier potent strain-transcending invasion inhibitory antibodies were

636

demonstrated only by targeting combinations of key merozoite antigens involved in erythrocyte

637

invasion (24, 29, 36, 46, 52).

638

In line with these reports, we feel that targeting single antigens may not be an effective

639

long-term strategy for malaria vaccines as the parasite has the strong ability to develop escape

640

mechanisms to counter the immune pressure. This capability is well displayed in the rapid

641

development of resistance in the parasite under drug pressure that has prompted only

642

combinatorial anti-malarial drug therapies. More so in combination, antibodies neutralize the

643

parasite at lower individual IgG concentrations (3.3 mg/ml total IgG each) that would be easier

644

to achieve using human compatible adjuvants or delivery platforms. PfRH5 antibodies in

645

combination with those against PfRH2, PfAARP and PfF2 produced an additive inhibition of

646

erythrocyte invasion. Our data is consistent with a previous study that has reported a potent

647

synergy in invasion inhibition by adenoviral vector induced antibodies against PfRH5 and other

648

EBA/PfRH antigens (36). We have also demonstrated that these PfRH5 based co-immunized

649

antigen mixtures induce balanced antibody responses against all three antigens with no immune

29

650

interference. Purified total IgG against the antigen mixtures were as potent in inhibiting

651

erythrocyte invasion as the antibodies combined in vitro.

652

Thus, our study establishes a proof of principle for the production of full-length wild-type

653

recombinant PfRH5 in a bacterial expression system that is known to be scalable for mass

654

vaccine production and has the potential to be taken forward for its development as a component

655

of a subunit blood-stage combination malaria vaccine. However, we have demonstrated potent

656

invasion inhibitory antibodies against PfRH5 raised through freund’s formulations that are

657

known to induce very strong immune responses but are not safe for human vaccine applications.

658

Thus, it is imperative for the clinical development of recombinant PfRH5 as a malaria vaccine to

659

identify human compatible adjuvants or delivery platforms that would also elicit similar highly

660

potent neutralizing antibodies. Nevertheless, the production of the wild type full length

661

recombinant PfRH5 protein with no amino acid modifications further enables the structure-

662

function analysis of this highly efficacious and attractive blood-stage malaria vaccine candidate.

663 664 665 666 667 668 669 670 671 672 673 30

674

Acknowledgements

675

We are grateful to Dr Louis Miller (NIH) for providing the P. falciparum clones used in

676

the study and the rPfRH430 expression plasmid. We also wish to thank Dr Lee Hall and Dr Annie

677

Mo from the Parasitology and International Programs Branch (PIPB), NIAID, NIH for providing

678

the recombinant EBA-175 RII protein. The technical assistance of Dr Alka Galav, Rakesh

679

Kumar Singh and Ashok Das from the ICGEB animal facility in performing the animal

680

experiments is deeply appreciated. We wish to thank Dr Inderjeet Kaur at the mass spectrometry

681

facility of the Malaria group, ICGEB for helping us in the LC-MS analysis. We appreciate Ms

682

Surbhi Dabral at the super resolution imaging facility of the Malaria group, ICGEB for technical

683

help in our imaging study.

684

Deepak Gaur is the recipient of the Ramalingaswami Fellowship from the Department of

685

Biotechnology, Government of India. Deepak Gaur is also the recipient of the Grand Challenges

686

Exploration Grant from the Bill and Melinda Gates Foundation. This work was supported by the

687

Bill & Melinda Gates Foundation through the Grand Challenges Explorations Initiative [GCE

688

OPP1007027 to D.G.]; Department of Biotechnology (DBT), Government of India through the

689

Ramalingaswami fellowship program [BT/HRD/35 /02/14/2008 to D.G.], Rapid Grant Scheme

690

for Young Investigators [BT/PR13376/GBD/27/ 260/2009 to D.G.], Vaccine Grand Challenges

691

Program [ND/DBT/12/040 to D.G., C.E.C., V.S.C.]. K.S.R., T.S. are recipients of Senior

692

Research Fellowships of the Council of Scientific and Industrial Research, Government of India;

693

A.K.P is a recipient of a Post-doctoral Research Associateship of DBT; H.S. is the recipient of

694

the Senior Research Fellowship of DBT. A.E. is the recipient of the ICGEB International PhD

31

695

Pre-doctoral fellowship. The funders had no role in study design, data collection and analysis,

696

decision to publish, or preparation of the manuscript.

697

This work has been previously presented at the 24th National Congress of Parasitology in

698

Jabalpur, Madhya Pradesh, India (April 27-29, 2013) and the 2013 Malaria Gordon Research

699

Conference in Tuscany, Italy (August 4-9, 2013).

700 701 702

Conflict of Interest: D.G., V.S.C., C.E.C are named on patent applications relating to PfRH5

703

and/or other malaria vaccines. This does not alter our adherence to all Infection and Immunity

704

policies on sharing data and material.

705 706 707 708 709 710 711 712 713 714 715 716 717 32

718 References: 719 1.

Murray CJ, Rosenfeld LC, Lim SS, Andrews KG, Foreman KJ, Haring D,

720

Fullman N, Naghavi M, Lozano R, Lopez AD. 2012. Global malaria mortality

721

between 1980 and 2010: a systematic analysis. Lancet 379:413–431.

722 2.

Gaur D, Chitnis CE. 2011. Molecular interactions and signaling mechanisms during

723

erythrocyte invasion by malaria parasites. Curr. Opin. Microbiol. 14:422–428.

724 3.

Gaur D, Mayer DC, Miller LH. 2004. Parasite ligand-host receptor interactions during

725

invasion of erythrocytes by Plasmodium merozoites. Int. J. Parasitol. 34:1413–1429.

726 4.

Cowman AF, Crabb BS. 2006. Invasion of red blood cells by malaria parasites. Cell

727

124:755–766.

728 5.

Ogutu BR, Apollo OJ, McKinney D, Okoth W, Siangla J, Dubovsky F, Tucker K,

729

Waitumbi JN, Diggs C, Wittes J., Malkin E, Leach A, Soisson LA, Milman JB,

730

Otieno L, Holland CA, Polhemus M, Remich SA, Ockenhouse CF, Cohen J, Ballou

731

WR, Martin SK, Angov E, Stewart VA, Lyon JA, Heppner DG, Withers MR.

732

2009. Blood stage malaria vaccine eliciting high antigen-specific antibody

733

concentrations confers no protection to young children in Western Kenya. PLoS One

734

4:e4708.

735 6.

Spring MD, Cummings JF, Ockenhouse CF, Dutta S, Reidler R, Angov E,

736

Bergmann-Leitner E, Stewart VA, Bittner S, Juompan L, Kortepeter MG,

737

Nielsen R, Krzych U, Tierney E, Ware LA, Dowler M, Hermsen CC, Sauerwein

738

RW, de Vlas SJ, Ofori-Anyinam O, Lanar DE, Williams JL, Kester KE, Tucker

739

K, Shi M, Malkin E, Long C, Diggs CL, Soisson L, Dubois MC, Ballou WR, 33

740

Cohen J, Heppner DG Jr. 2009. Phase 1/2a study of the malaria vaccine candidate

741

apical membrane antigen-1 (AMA-1) administered in adjuvant system AS01B or

742

AS02A. PLoS One 4:e5254.

743 7.

Thera MA, Doumbo OK, Coulibaly D, Laurens MB, Ouattara A, Kone AK,

744

Guindo AB, Traore K, Traore I, Kouriba B, Diallo DA, Diarra I, Daou M, Dolo A,

745

Tolo Y, Sissoko MS, Niangaly A, Sissoko M, Takala-Harrison S, Lyke KE, Wu Y,

746

Blackwelder WC, Godeaux O, Vekemans J, Dubois MC, Ballou WR, Cohen J,

747

Thompson D, Dube T, Soisson L, Diggs CL, House B, Lanar DE, Dutta S, Heppner

748

DG Jr., Plowe CV. 2011. A field trial to assess a blood-stage malaria vaccine. N. Engl.

749

J. Med. 365:1004–1013.

750 8.

Tham WH, Healer J, Cowman AF. 2012. Erythrocyte and reticulocyte binding-like

751

proteins of Plasmodium falciparum. Trends Parasitol. 28(1):23-30.

752 9.

Cowman AF, Berry D, Baum J. 2012. The cellular and molecular basis for malaria

753

parasite invasion of the human red blood cell. J Cell Biol. 198(6):961-971.

754 10. Rayner JC, Vargas-Serrato E, Huber CS, Galinski MR, Barnwell JW. 2001. A 755

Plasmodium falciparum homologue of Plasmodium vivax reticulocyte binding protein

756

(PvRBP1) defines a trypsin-resistant erythrocyte invasion pathway. J. Exp. Med.

757

194:1571–1581.

758 11. Triglia T, Duraisingh MT, Good RT, Cowman AF. 2005. Reticulocyte-binding 759

protein homologue 1 is required for sialic acid-dependent invasion into human

760

erythrocytes by Plasmodium falciparum. Mol. Microbiol. 55:162–174.

34

761 12. Rayner JC, Galinski MR, Ingravallo P, Barnwell JW. 2000. Two Plasmodium 762

falciparum genes express merozoite proteins that are related to Plasmodium vivax and

763

Plasmodium yoelii adhesive proteins involved in host cell selection and invasion. Proc.

764

Natl. Acad. Sci. USA. 97: 9648–9653.

765 13. Duraisingh MT, Triglia T, Ralph SA, Rayner JC, Barnwell JW, McFadden 766

GI, Cowman AF. 2003. Phenotypic variation of Plasmodium falciparum merozoite

767

proteins directs receptor targeting for invasion of human erythrocytes. EMBO J.

768

22:1047–1057.

769 14. Kaneko O, Mu J, Tsuboi T, Su X, Torii M. 2002. Gene structure and expression of a 770

Plasmodium falciparum 220-kDa protein homologous to the Plasmodium vivax

771

reticulocyte binding proteins. Mol. Biochem. Parasitol. 121:275–278.

772 15. Gao X, Yeo KP, Aw SS, Kuss C, Iyer JK, Genesan S, Rajamanonmani R, Lescar 773

J, Bozdech Z, Preiser PR. 2008. Antibodies targeting the PfRH1 binding domain

774

inhibit invasion of Plasmodium falciparum merozoites. PLoS Pathog. 4:e1000104.

775 16. Sahar T, Reddy KS, Bharadwaj M, Pandey AK, Singh S, Chitnis CE, Gaur D. 776

2011. Plasmodium falciparum reticulocyte binding-like homologue protein 2 (PfRH2)

777

is a key adhesive molecule involved in erythrocyte invasion. PLOS One 6:e17102.

778 17. Triglia T, Chen L, Lopaticki S, Dekiwadia C, Riglar DT, Hodder AN, Ralph SA, 779

Baum J, Cowman AF. 2011. Plasmodium falciparum merozoite invasion is inhibited

780

by antibodies that target the PfRh2a and b binding domains. PLoS Pathog. 7:e1002075.

35

781 18. Stubbs J, Simpson KM, Triglia T, Plouffe D, Tonkin CJ, Duraisingh MT, Maier 782

AG, Winzeler EA, Cowman AF. 2005. Molecular mechanism for switching of P.

783

falciparum invasion pathways into human erythrocytes. Science 309:1384–1387.

784 19. Gaur D, Furuya T, Mu J, Jiang LB, Su XZ, Miller LH. 2006. Upregulation of 785

expression of the reticulocyte homology gene 4 in the Plasmodium falciparum clone

786

Dd2 is associated with a switch in the erythrocyte invasion pathway. Mol. Biochem.

787

Parasitol. 145:205–215.

788 20. Gaur D, Singh S, Singh S, Jiang L, Diouf A, Miller LH. 2007. Recombinant 789

Plasmodium falciparum reticulocyte homology protein 4 binds to erythrocytes and

790

blocks invasion. Proc. Natl. Acad. Sci. USA. 104:17789–17794.

791 21. Hayton K, Gaur D, Liu A,Henschen B, Singh S, Lambert L, Furuya T, Bouttenot 792

R, Doll M, Nawaz F, Mu J, Jiang L, Miller LH, Wellems TE. 2008. Erythrocyte

793

binding protein PfRH5 polymorphisms determine species-specific pathways of

794

Plasmodium falciparum invasion. Cell Host Microbe 4:40–51.

795 22. Baum J, Chen L, Healer J, Lopaticki S, Boyle M, Triglia T, Ehlgen F, Ralph 796

SA, Beeson JG, Cowman AF. 2009. Reticulocyte-binding protein homologue 5 – an

797

essential adhesin involved in invasion of human erythrocytes by Plasmodium

798

falciparum. Int. J. Parasitol. 39:371–380.

799 23. Hayton K, Dumoulin P, Henschen B, Liu A, Papakrivos J, Wellems TE. 2012. 800

Various PfRH5 polymorphisms can support Plasmodium falciparum invasion into the

801

erythrocytes of owl monkeys and rats. Mol. Biochem. Parasitol. 187(2):103-110.

36

802 24. Chen L, Lopaticki S, Riglar DT, Dekiwadia C, Uboldi AD, Tham WH, O'Neill MT, 803

Richard D, Baum J, Ralph SA, Cowman AF. 2011. An EGF-like Protein Forms a

804

Complex with PfRh5 and Is Required for Invasion of Human Erythrocytes

805

by Plasmodium falciparum. PLoS Pathog. 7:e1002199.

806 25. Crosnier C, Bustamante LY, Bartholdson SJ, Bei AK, Theron M, Uchikawa M, 807

Mboup S, Ndir O, Kwiatkowski DP, Duraisingh MT, Rayner JC, Wright GJ. 2011.

808

Basigin is a receptor essential for erythrocyte invasion by Plasmodium falciparum.

809

Nature 480:534–537.

810 26. Douglas AD, Williams AR, Illingworth JJ, Kamuyu G, Biswas S, Goodman AL, 811

Wyllie DH, Crosnier C, Miura K, Wright GJ, Long CA, Osier FH, Marsh K,

812

Turner AV, Hill AV, Draper SJ. 2011. The blood-stage malaria antigen PfRH5 is

813

susceptible to vaccine-inducible cross-strain neutralizing antibody. Nat. Commun.

814

2:601.

815 27. Bustamante LY, Bartholdson SJ, Crosnier C, Campos MG, Wanaguru M, Nguon 816

C,

817

recombinant Plasmodium falciparum PfRH5 protein induces inhibitory antibodies that

818

are effective across common PfRH5 genetic variants. Vaccine. 31(2):373-379.

Kwiatkowski

DP,

Wright

GJ,

Rayner

JC.

2012.

A

full-length

819 28. Rodriguez M, Lustigman S, Montero E, Oksov Y, Lobo CA. 2008. PfRH5: a novel 820

reticulocyte-binding family homolog of Plasmodium falciparum that binds to the

821

erythrocyte, and an investigation of its receptor. PLoS One. 3(10):e3300.

822 29. Pandey AK, Reddy KS, Sahar T, Gupta S, Singh H, Reddy EJ, Asad M, Siddiqui 823

FA, Gupta P, Singh B, More KR, Mohmmed A, Chitnis CE, Chauhan VS, Gaur D. 37

824

2013. Identification of a potent combination of key Plasmodium falciparum merozoite

825

antigens that elicit strain-transcending parasite-neutralizing antibodies. Infect Immun.

826

81(2):441-451.

827 30. Siddiqui FA, Dhawan S, Singh S, Singh B, Gupta P, Pandey A, Mohmmed A, Gaur 828

D, Chitnis CE. 2013. A thrombospondin structural repeat containing rhoptry protein

829

from Plasmodium falciparum mediates erythrocyte invasion. Cell Microbiol.

830

15(8):1341-1356.

831 31. Sim BK, Chitnis CE, Wasniowska K, Hadley TJ, Miller LH. 1994. Receptor and 832

ligand domains for invasion of erythrocytes by Plasmodium falciparum. Science 264:

833

1941–1944.

834 32. Pandey KC, Singh S, Pattnaik P, Pillai CR, Pillai U, Lynn A, Jain SK, Chitnis CE. 835

2002. Bacterially expressed and refolded receptor binding domain of Plasmodium

836

falciparum EBA-175 elicits invasion inhibitory antibodies. Mol. Biochem. Parasitol.

837

123:23–33.

838 33. Jiang L, Gaur D, Mu J, Zhou H, Long CA, Miller LH. 2011. Evidence for EBA- 175 839

as a component of a ligand blocking blood stage malaria vaccine. Proc Natl Acad Sci

840

USA 108: 7553–7558.

841 34. Wickramarachchi T, Devi YS, Mohmmed A, Chauhan VS. 2008. Identification and 842

characterization of a novel Plasmodium falciparum merozoite apical protein involved in

843

erythrocyte binding and invasion. PLoS One 3:e1732.

38

844 35. Gaur D, Storry JR, Reid ME, Barnwell JW, Miller LH. 2003. Plasmodium 845

falciparum is able to invade erythrocytes through a trypsin-resistant pathway

846

independent of glycophorin B. Infect Immun 71(12): 6742-6746.

847 36. Williams AR, Douglas AD, Miura K, Illingworth JJ, Choudhary P, Murungi LM, 848

Furze JM, Diouf A, Miotto O, Crosnier C, Wright GJ, Kwiatkowski DP, Fairhurst

849

RM, Long CA, Draper SJ. 2012. Enhancing blockade of Plasmodium falciparum

850

erythrocyte invasion: assessing combinations of antibodies against PfRH5 and other

851

merozoite antigens. PLoS Pathog. 8(11):e1002991.

852 37. Crompton PD, Miura K, Traore B, Kayentao K, Ongoiba A, Weiss G, Doumbo S, 853

Doumtabe D, Kone Y, Huang CY, Doumbo OK, Miller LH, Long CA, Pierce SK.

854

2010. In vitro growth-inhibitory activity and malaria risk in a cohort study in Mali.

855

Infect. Immun. 78:737–745.

856 38. Rono J, Färnert A, Olsson D, Osier F, Rooth I, Persson KE. 2012. Plasmodium 857

falciparum line-dependent association of in vitro growth-inhibitory activity and risk of

858

malaria. Infect. Immun. 80:1900–1908.

859 39. Kennedy MC, Wang J, Zhang Y, Miles AP, Chitsaz F, Saul A, Long CA, Miller 860

LH, Stowers AW. 2002. In vitro studies with recombinant Plasmodium falciparum

861

apical membrane antigen 1 (AMA1): production and activity of an AMA1 vaccine and

862

generation of a multiallelic response. Infect Immun. 70(12):6948-6960.

863 40. Duan J, Mu J, Thera MA, Joy D, Kosakovsky Pond SL, Diemert D, Long C, Zhou 864

H, Miura K, Ouattara A, Dolo A, Doumbo O, Su XZ, Miller L. 2008. Population

865

structure of the genes encoding the polymorphic Plasmodium falciparum apical 39

866

membrane antigen 1: implications for vaccine design. Proc. Natl. Acad. Sci. USA.

867

105:7857–7862.

868 41. Srinivasan P, Beatty WL, Diouf A, Herrera R, Ambroggio X, Moch JK, Tyler JS, 869

Narum DL, Pierce SK, Boothroyd JC, Haynes JD, Miller LH. 2011. Binding of

870

Plasmodium merozoite proteins RON2 and AMA1 triggers commitment to invasion.

871

Proc Natl Acad Sci USA. 108(32):13275-13280.

872 42. Lamarque M, Besteiro S, Papoin J, Roques M, Vulliez-Le Normand B, Morlon873

Guyot J, Dubremetz JF, Fauquenoy S, Tomavo S, Faber BW, Kocken CH, Thomas

874

AW, Boulanger MJ, Bentley GA, Lebrun M. 2011. The RON2-AMA1 interaction is

875

a critical step in moving junction-dependent invasion by apicomplexan parasites. PLoS

876

Pathog. 10;7(2):e1001276.

877 43. Healer J, Murphy V, Hodder AN, Masciantonio R, Gemmill AW, Anders 878

RF, Cowman AF, Batchelor A. 2004. Allelic polymorphisms in apical membrane

879

antigen 1 are responsible for evasion of antibody mediated inhibition in Plasmodium

880

falciparum. Mol Microbiol 52: 159 168.

881 44. Dutta S, Lee SY, Batchelor AH, Lanar DE. 2007. Structural basis of antigenic escape 882

of a malaria vaccine candidate. Proc Natl Acad Sci U S A. 104(30):12488-12493.

883 45. Ouattara A, Takala-Harrison S, Thera MA, Coulibaly D, Niangaly A, Saye R, Tolo 884

Y, Dutta S, Heppner DG, Soisson L, Diggs CL, Vekemans J, Cohen J, Blackwelder

885

WC, Dube T, Laurens MB, Doumbo OK, Plowe CV. 2013 Molecular basis of allele-

886

specific efficacy of a blood-stage malaria vaccine: vaccine development implications. J

887

Infect Dis. 207(3):511-519. 40

888 46. Miura K, Herrera R, Diouf A, Zhou H, Mu J, Hu Z, MacDonald NJ, Reiter K, 889

Nguyen V, Shimp RL Jr, Singh K, Narum DL, Long CA, Miller LH. 2013.

890

Overcoming allelic specificity by immunization with five allelic forms of Plasmodium

891

falciparum apical membrane antigen 1. Infect Immun. 81(5):1491-1501.

892 47. Duncan CJ, Hill AV, Ellis RD. 2012. Can growth inhibition assays (GIA) predict 893

blood-stage malaria vaccine efficacy? Hum Vaccin Immunother. 8(6):706-714.

894 48. Regules JA, Cummings JF, Ockenhouse CF. 2011. The RTS,S vaccine candidate for 895

malaria. Expert Rev Vaccines. 10(5):589-599.

896 49. White NJ. 2011. A vaccine for malaria. N Engl J Med. 365(20):1926-1927. 897 50. Daily JP. 2012. Malaria vaccine trials--beyond efficacy end points. N Engl J Med. 898

367(24):2349-2351.

899 51. Stoop JW, Zegers BJ, Sander PC, Ballieux RE. 1969. Serum immunoglobulin levels 900

in healthy children and adults. Clin. Exp. Immunol. 4:101–112.

901 52. Lopaticki S, Maier AG, Thompson J, Wilson DW, Tham WH, Triglia T, Gout A, 902

Speed TP, Beeson JG, Healer J, Cowman AF. 2011. Reticulocyte and erythrocyte

903

binding-like proteins function cooperatively in invasion of human erythrocytes by

904

malaria parasites. Infect. Immun. 79:1107–1117.

905 906 907 908 909 910 911 912 41

913

Figure Legends

914 915

Figure 1: Production of the recombinant rPfRH563 protein. (A) Schematic

916

representation of the PfRH5 parasite protein. Region in black denotes the signal peptide

917

(residues 1-26) and region in green (residues 27-526) was expressed in E. coli. (B)

918

Purified rPfRH563 protein analyzed on 12% SDS-PAGE, stained with coomassie blue.

919

2µg of rPfRH563 has been loaded in each well. (C) Recombinant rPfRH563 protein

920

detected in immunoblots using the anti-His tag antibody. R and NR represents -reducing

921

and non-reducing conditions, respectively.

922 923

Figure 2: Erythrocyte binding activity of the native PfRH5 parasite protein and

924

recombinant rPfRH563 protein. Anti-PfRH5 antibodies in immunoblots detected both

925

native PfRH5 and rPfRH563 among the proteins that bound with the erythrocyte surface

926

and got eluted off by 1.5 M NaCl. (A) Native PfRH5 from the 3D7 culture supernatant

927

bound untreated (U) and all three enzymatically treated human erythrocytes

928

[neuraminidase (N), trypsin (T), chymotrypsin (C)] but failed to bind with proteinase K

929

(PK) treated erythrocytes. Thus, PfRH5 binds erythrocytes in a sialic acid independent,

930

trypsin/chymotrypsin resistant, proteinase K sensitive manner. (B) rPfRH563 bound

931

human erythrocytes with the same specificity as that of native PfRH5. (C) Native PfEBA-

932

175, as well as (D) PfF2, (recombinant receptor binding domain of PfEBA-175) were

933

analyzed as a control with the same set of enzymatically treated erythrocytes. Both native

934

PfEBA-175 and PfF2 bound erythrocytes in neuraminidase/trypsin/proteinase K 42

935

sensitive, chymotrypsin resistant manner. PBS denotes Phosphate buffer saline, pH 7.4

936

that contained no protein. No specific protein was detected in the eluate fractions with the

937

PBS control suggesting that no non-specific erythrocyte protein was being detected in the

938

assay. (* U= Untreated; N= Neuraminidase; T= Trypsin; and C= Chymotrypsin; PK =

939

Proteinase K)

940 941

Figure 3: Antibodies against recombinant rPfRH563 blocked erythrocyte binding of

942

both the native PfRH5 and rPfRH563 proteins. Purified total rabbit IgG against

943

rPfRH563 blocked binding of (A) native PfRH5 and (B) recombinant rPfRH563 in a dose

944

dependent manner. The PfRH5 antibodies had no effect on the erythrocyte binding of (C)

945

native EBA-175 or (D) recombinant PfF2 even at the maximum IgG concentration of 800

946

µg/ml.

947 948

Figure 4: Anti-Basigin monoclonal antibodies blocked the erythrocyte binding of

949

native and recombinant PfRH5. (A) Anti-Basigin TRA-1-85 monoclonal antibodies

950

potently inhibited invasion of human erythrocytes by the P. falciparum clone 3D7, with

951

complete blockade of invasion observed at 5µg/ml. At the same invasion inhibitory IgG

952

concentrations, the TRA-1-85 monoclonal antibodies potently blocked the erythrocyte

953

binding of both the (B) native PfRH5 protein and (C) recombinant rPfRH563. Anti-

954

glycophorin A monoclonal antibodies had no effect on the binding of both (B) native and

955

(C) recombinant PfRH5.

956 43

957

Figure 5: Invasion inhibitory activity of anti-PfRH5 rabbit antibodies. (A) Invasion

958

inhibitory activity of purified rabbit total IgG (0.5-10 mg/ml) against rPfRH563 and the

959

receptor binding domains of other key merozoite ligands (PfRH1, PfRH2, PfRH4,

960

PfAARP, PfF2) against the P. falciparum clone 3D7. AMA-1 IgG (5 mg/ml) was used as

961

a positive control. (B) Strain-transcending parasite neutralization activity of anti-PfRH5

962

total rabbit IgG (0.5-10 mg/ml) against five diverse P. falciparum clones. The control

963

anti-HPR total IgG failed to exhibit any invasion inhibition at the maximum

964

concentration of 10 mg/ml. The results represent the average of three independent

965

experiments performed in duplicate. The error bars represent the standard error of the

966

mean.

967 968

Figure 6: Invasion inhibitory efficacy of PfRH5 based antibody combinations

969

against P. falciparum clones (3D7, Dd2). Purified rabbit total IgG against the six

970

individual antigens (PfRH1, PfRH2, PfRH4, PfRH5, PfAARP, PfF2) were evaluated

971

individually (3.3 mg/ml) as well as in all ten possible PfRH5 based triple antibody

972

combinations (3.3 mg/ml each; total 10 mg/ml) against (A) the sialic acid independent

973

clone 3D7 and (B) the sialic acid dependent clone Dd2. The control anti-HPR total IgG

974

failed to exhibit any invasion inhibition at the maximum concentration of 10 mg/ml. The

975

results represent the average of three independent experiments performed in duplicate.

976

The error bars represent the standard error of the mean.

977 978 44

979

Figure 7: Strain-transcending parasite neutralization of PfRH5 based antibody

980

combinations. Purified rabbit total IgG against the six individual antigens (PfRH1,

981

PfRH2, PfRH4, PfRH5, PfAARP, PfF2) were evaluated individually and in the six most

982

potent PfRH5 based antibody combinations (identified from Fig. 6) against five diverse

983

P. falciparum clones. The invasion inhibitory activity of the antibody combination,

984

AARP+RH2+PfF2, was also analyzed in the assay. The control anti-HPR total IgG failed

985

to exhibit any invasion inhibition at the maximum concentration of 10 mg/ml. Three

986

independent assays were performed in duplicate. The error bars represent the standard

987

error of the mean.

988 989

Figure 8: Invasion inhibitory activity of antibodies raised against the two co-

990

immunized antigen formulations: Total IgG purified from mice sera raised against the

991

immunogens (RH5, RH2, PfF2, AARP, PfF2+RH5+AARP, RH2+RH5+AARP) were

992

evaluated for its invasion inhibitory activity (at concentrations of 1, 3.3, 5 and 10 mg/ml)

993

against the (A) sialic acid independent clone 3D7 and the (B) sialic acid dependent clone

994

Dd2. The control anti-HPR total IgG failed to exhibit any invasion inhibition at the

995

maximum concentration of 10 mg/ml. Two independent assays were performed in

996

duplicate. The error bars show the standard error of the mean. * denotes statistical

997

significance between the invasion inhibition produced by the antibodies against the two

998

antigen mixtures (PfF2+RH5+AARP, RH2+RH5+AARP) with respect to the individual

999

PfRH5 antibodies at the same concentrations of purified total IgG, 3.3 mg/ml and 5 mg/ml

1000

(p values < 0.05). 45

Lihat lebih banyak...

Comentarios

Copyright © 2017 DATOSPDF Inc.