A genetically attenuated malaria vaccine candidate based on P. falciparum b9/slarp gene-deficient sporozoites

July 9, 2017 | Autor: Geert-jan Van Gemert | Categoría: Humans, Liver, Mice, Animals, Plasmodium falciparum, Elife, Malaria Vaccines, Elife, Malaria Vaccines
Share Embed


Descripción

ACCEPTED MANUSCRIPT

A genetically attenuated malaria vaccine candidate based on P. falciparum b9/slarp gene-deficient sporozoites Ben C L van Schaijk, Ivo H J Ploemen, Takeshi Annoura, Martijn W Vos, Foquet Lander, Geert-Jan van Gemert, Severine Chevalley-Maurel, Marga van de Vegte-Bolmer, Mohammed Sajid, Jean-Francois Franetich, Audrey Lorthiois, Geert Leroux-Roels, Philip Meuleman, Cornelius C Hermsen, Dominique Mazier, Stephen L Hoffman, Chris J Janse, Khan M Shahid, Robert W Sauerwein

DOI: http://dx.doi.org/10.7554/eLife.03582 Cite as: eLife 2014;10.7554/eLife.03582 Received: 4 June 2014 Accepted: 19 November 2014 Published: 19 November 2014

This PDF is the version of the article that was accepted for publication after peer review. Fully formatted HTML, PDF, and XML versions will be made available after technical processing, editing, and proofing. This article is distributed under the terms of the Creative Commons Attribution License permitting unrestricted use and redistribution provided that the original author and source are credited.

Stay current on the latest in life science and biomedical research from eLife. Sign up for alerts at elife.elifesciences.org

A genetically attenuated malaria vaccine candidate based on P. falciparum b9/slarp gene-deficient sporozoites One sentence summary:

Preclinical evaluation of a malaria GAP vaccine candidate

Ben C.L. van Schaijk1†, Ivo H.J. Ploemen1†‡, Takeshi Annoura2,‡‡, Martijn W. Vos1, Lander Foquet3 Geert-Jan van Gemert1, Severine Chevalley-Maurel2, Marga van de Vegte-Bolmer1, Mohammed Sajid2, Jean‐Francois Franetich4,5, Audrey Lorthiois4,5, Geert Leroux-Roels3, Philip Meuleman3, Cornelus C. Hermsen1, Dominique Mazier4,5,6, Stephen L. Hoffman7, Chris J. Janse2, Shahid M. Khan2, Robert W. Sauerwein1* 1

Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands.

2

The Leiden Malaria Research Group, Parasitology, Leiden University Medical Center, Leiden, The Netherlands.

3

Center for Vaccinology, Ghent University and University Hospital, Ghent, Belgium.

4

Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris ; INSERM, U1135, Paris, F-75013 France.

5

Centre d'Immunologie et des Maladies Infectieuses, CIMI-Paris; Université Pierre et Marie Curie-Paris 6,

UMRS CR7, Paris, France. 6 7

AP-HP, Groupe hospitalier Pitié-Salpêtrière, Service Parasitologie-Mycologie, Paris, F-75013 France.

Sanaria Inc. 9800 Medical Center Drive, Suite A209 Rockville, MD, USA.

1

Email addresses: BvS : [email protected] IP: [email protected] TA: [email protected] MV: [email protected] LF : [email protected] GvG: [email protected] SCC: [email protected] MvdV: [email protected] MS: [email protected] JF: [email protected] AL: [email protected] GLR: [email protected] PM: [email protected] ‎CH: [email protected] DM: [email protected]‎ SH: [email protected] CJ: [email protected] SK: [email protected] RS: [email protected]





Authors contributed equally Present address: Institute for Translational Vaccinology, Bilthoven, The Netherlands.

‡‡

Present address: Department of Tropical Medicine, The Jikei University School of Medicine, Tokyo, Japan.

* Corresponding author: Tel: +31 24 3614306; Fax: +31 24 341666 Email address: [email protected] (Robert Sauerwein) Geert Grooteplein 28, 6525 GA, Nijmegen, The Netherlands

2

Abstract 1

A highly efficacious pre-erythrocytic stage vaccine would be an important tool for the control and elimination of

2

malaria but is currently unavailable. High-level protection in humans can be achieved by experimental

3

immunization with Plasmodium falciparum sporozoites attenuated by radiation or under anti-malarial drug

4

coverage. Immunization with genetically attenuated parasites (GAP) would be an attractive alternative approach.

5

Here we present data on safety and protective efficacy using sporozoites with deletions of two genes i.e. the

6

newly identified b9 and slarp, which govern independent and critical processes for successful liver-stage

7

development. In the rodent malaria model, Pb∆b9∆slarpGAP was completely attenuated showing no

8

breakthrough infections while efficiently inducing high level protection. The human Pf∆b9∆slarpGAP generated

9

without drug-resistance markers were infective to human hepatocytes in vitro and to humanized mice engrafted

10

with human hepatocytes in vivo but completely aborted development after infection. These findings support the

11

clinical development of a Pf∆b9∆slarpSPZ vaccine.

12 13 14

Keywords: malaria, vaccine, genetically attenuated parasite (GAP), sporozoite, liver, PfSPZ.

15

3

16

Introduction

17

A vaccine that induces high-level (>90%) sterile protection by inducing immunity that attacks the non-

18

pathologic, asymptomatic pre-erythrocytic stages of Plasmodium falciparum (Pf) will prevent infection, disease,

19

and transmission and could be a powerful instrument to eliminate Pf malaria from geographically defined areas

20

(1, 2). In rodent models, sterile protection can be induced by immunization with live Plasmodium sporozoites

21

attenuated by either irradiation, genetic modification (GAP) or by concomitant anti-parasitic drug treatment (for

22

reviews see(3-6)). In humans, induction of complete sustained protective immunity against a challenge infection

23

has been achieved by previous exposure to the bites of mosquitoes infected with i) live radiation-attenuated

24

Plasmodium sporozoites that invade but then completely arrest in the liver (7, 8) and ii) live sporozoites in

25

volunteers taking chloroquine chemoprophylaxis (CPS) with full parasite liver stage development; once released

26

into the circulation asexual blood stages are killed by chloroquine (9, 10). More recently it has been

27

demonstrated for the first time that sterile immunity can be achieved by intravenous immunization with

28

radiation-attenuated aseptic, purified, cryopreserved Pf sporozoites (SPZ) called PfSPZ Vaccine (11).

29

From a product manufacturing perspective, GAPs have the clear advantage of representing a

30

homogeneous parasite population with a defined genetic identity. The genetic attenuation is an irreversible,

31

intrinsic characteristic of the parasite that does not require additional manufacturing steps like irradiation.

32

Furthermore, in the manufacturing process of GAP-infected mosquitoes, operators are never exposed to Pf

33

parasites that can cause disease. However, clinical development of GAPs has suffered from safety problems

34

related to breakthrough infections during immunization leading to pathological blood stage infections

35

responsible for clinical symptoms and complications. Strains of mice showed differential susceptibility to

36

breakthrough infections after injection of sporozoites of rodent malaria GAPs, demonstrating the need for

37

extensive preclinical rodent screening (12). The P. falciparum GAP Pf∆p52∆p36, is the only GAP so far that has

38

been assessed in humans but the trial in which the Pf sporozoites were administered by mosquito bite had to be

39

terminated, because of breakthrough infections in one volunteer during immunization (13). Our in vitro

40

experiments with Pf∆p52∆p36 confirm that this double gene deletion GAP (i.e. two genes removed from the

41

genome) is not fully attenuated similar to the equivalent rodent GAP, Pb∆p52∆p36 in the P. berghei/ C57BL/6

42

model(12) . Therefore, identification of additional genes critical and uniquely selective for liver stage

43

development has become a major challenge for GAP vaccine development (4, 12, 14). Furthermore, single gene

44

deletion GAPs will most likely not be adequate (14).

4

45

This prompted us to generate and test a GAP with deletions of two independent genes critical for liver

46

stage development. We recently identified a novel P. berghei (Pb) gene deletion mutant, Pb∆b9 lacking the

47

expression of the B9 protein (Pf ortholog: PFC_0750w; PF3D7_0317100)(15). This protein is a newly identified

48

member of the Plasmodium 6-Cys family of proteins. Initial safety evaluation in rodents demonstrated that

49

Pb∆b9 mutants have a stronger attenuation phenotype than mutants lacking the 6-Cys proteins P52 and P36 (15-

50

18). As second target gene for liver stage attenuation, we selected the slarp and sap1 orthologs reported in Pb

51

and P. yoelii (Py) respectively (Pf ortholog: PF11_0480; PF3D7_1147000; hereafter termed slarp). These slarp

52

mutants show an excellent safety profile by full arrest in the liver in mice (19, 20). The SLARP protein is

53

expressed in sporozoites and in early liver stages and is involved in the regulation of transcription (20, 21).

54

Here we report the generation and evaluation of a rodent GAP lacking the genes encoding for B9 and

55

SLARP (Pb∆b9∆slarp) and the generation and evaluation of the equivalent human Pf GAP lacking the Pf

56

ortholog genes. Pf∆b9∆slarp was generated using constructs that allowed for the removal of the drug selectable

57

marker from the genome by FRT/FLPe recombinase methodology (22). The safety and efficacy of Pb∆b9∆slarp

58

and the lack of development of Pf∆b9∆slarp in human hepatocytes, in vitro and in vivo in chimeric mice provide

59

strong support for clinical development of a Pf∆b9∆slarp PfSPZ vaccine.

5

60

Results

61

Arrest of liver stage development and induced protection after P. berghei ∆b9∆slarp GAP

62

Previously, we generated a Pb mutant with disruption of the b9 locus (Pb∆b9) by standard genetic

63

modification using a double cross-over integration event, followed by removal of the drug-selectable marker

64

cassette by negative selection (23). Characterization of the Pb∆b9 phenotype showed that liver stage

65

development was fully abrogated in BALB/c mice and severely compromised in the more stringent C57BL/6

66

murine model for P. berghei(15). Immunization of a single dose of 10K (i.e. 10.000 sporozoites) or‎5K‎Pb∆b9,

67

protected BALB/c mice against a 10K WT-sporozoite challenge, while 80% of mice were still protected after a

68

single 1K immunizing dose (Table 1). In‎C57BL/6‎mice,‎immunization‎with‎50K/20K/20K‎of‎Pb∆b9 resulted in

69

complete protection lasting up to 180 days, reducing to 45% protection when challenged at 1 year post-

70

immunization. However, sporozoite administration occasionally resulted in blood stage infections after

71

administration of high doses thereby compromising the safety profile(15).

72

Previously it has been shown by others that Pb∆slarp parasites are completely arrested in liver-stage

73

development with a complete lack of breakthrough blood-stage infections(19, 20). Therefore, we generated a

74

new single gene deletion mutant Pb∆slarp in a parasite line that constitutively expressed a fusion of the reporter

75

proteins GFP and luciferase using a slarp-targeting DNA-construct for deletion by double cross-over

76

homologous integration (Fig. 1- supplement 1). The Pb∆slarp mutant showed blood stage growth and mosquito

77

infections with functional sporozoites similar to wild-type (Supplementary File 1). However, intravenous

78

injection of up to 500k Pb∆slarp sporozoites never led to full development of parasites in the liver as assayed by

79

in vivo imaging (Fig. 1- supplement 1) or analysis of blood stage infection (Table 2). PbΔslarp sporozoites

80

arrested very soon after invasion of cultured Huh7 hepatocytes corroborating the excellent safety findings by

81

Silvie et al (20).

82

Therefore, in order to create a completely attenuated and safe rodent GAP, we additionally disrupted the

83

slarp gene in the Pb∆b9 genome by double cross-over integration (Fig. 1). Asexual growth and sporogonic

84

development/function equaled wild-type (Supplementary File 1). However, Pb∆b9Δslarp sporozoites arrested

85

soon after invasion of cultured Huh7 hepatocytes (Fig. 1) and intravenous injection of 150-200K Pb∆b9∆slarp

86

sporozoites never resulted in breakthrough blood-stage infections in mice (Table 2). Finally, protective efficacy

87

induced by Pb∆b9∆slarp was studied in both BALB/c and C57BL/6 mice. A single immunization dose of 10K,

88

5K or even 1K of Pb∆b9∆slarp sporozoites in BALB/c mice induced full protection against a 10K wild-type

89

sporozoite challenge (Table 1). C57BL/6 mice were 100% protected after 3x10K immunization with

6

90

Pb∆b9∆slarp sporozoites, and the protective efficacy reduced to 60% after a 3x1K immunization dose. A

91

challenge at day 180 post-immunization of a 50/20/20 K dose still resulted in complete protection. The combined

92

data showed that Pb∆b9∆slarp completely arrest during liver-stage development and induce a highly efficient

93

protective immunity in two different strains of mice.

94 95

Generation of a P. falciparum ∆b9∆slarp GAP

96

Considering the desired phenotype as observed in P. berghei, we generated a Pf mutant lacking

97

expression of both B9 (PF3D7_0317100) and SLARP (PF3D7_1147000; sporozoite asparagine-rich protein).

98

These genes are conserved between rodent and human species, both at the level of syntenic location in their

99

respective genomes on chromosomes 3 and 11 respectively, and at the sequence level. Pfb9 shows 37% amino

100

acid sequence identity and 54% sequence similarity with Pbb9(15); Pfslarp shows 28% amino acid sequence

101

identity and 46% sequence similarity with Pbslarp.

102

First, we generated two independent Pf mutants lacking slarp by standard double cross-over integration

103

of a DNA construct and analyzed their phenotype throughout the parasite life cycle (Fig. 2- supplement 1 and

104

2). Blood-stage‎ development‎ of‎ two‎ independently‎ derived‎ Pf∆slarp (i.e.‎ Pf∆slarp-a and –b) parasites was

105

comparable to WT parasites. Pf∆slarp mutants produced WT numbers of gametocytes, oocysts and sporozoites

106

(Fig. 2). Intracellular PfΔslarp-a and -b

107

significantly different in number and morphologically identical to WT parasites at 3 and 24 hours post-infection

108

(hpi) (Fig. 3). However, their number was more than 10-fold reduced at 48 hpi and not detectable from day 3

109

onwards to day 7 post-infection. Parasites lacking b9 in P. falciparum arrested before day 2 post-infection of

110

primary human hepatocytes with the exception of one observed liver schizont at a later timepoint(15). PfΔslarp-

111

a and -b parasites still showed positive HSP70 staining and morphologically normal parasites at 48 hpi in

112

primary human hepatocytes indicating later time point of arrest‎compared‎to‎Pf∆b9 parasites.

parasite development in primary human hepatocytes was not

113

Next, we generated double gene-deletion Pf∆b9∆slarp mutants using the FRT/FLPe recombinase

114

methodology (22). This methodology employs FLPe recombinase to remove a FRT-site flanked drug-resistance

115

marker cassette introduced into the Pf genome when the target gene has been removed by double cross-over

116

homologous recombination as‎shown‎for‎PfΔslarp-b parasites in Fig. 2- supplement 1 and 2. After cloning, this

117

‘marker-free’‎line was subsequently transfected with the Pfb9 gene-targeting construct pHHT-FRT-GFP-b9(15)

118

to delete the b9 locus‎from‎the‎PfΔslarp-b genome (Fig. 2- supplement 1 and 2). Subsequently two ‘marker-

119

free’‎clones, Pf∆b9∆slarp-F7 and Pf∆b9∆slarp-G9 were obtained containing the correct genotype i.e. removal of

7

120

the slarp and b9 gene loci as well as both respective drug-selection cassettes (Fig. 2- supplement 2). In addition,

121

we confirmed loss of expression of both slarp and b9 by RT-PCR analysis by demonstrating the absence of

122

transcripts in mRNA collected from Pf∆b9∆slarp-F7 and Pf∆b9∆slarp-G9 salivary gland sporozoites (Fig. 2-

123

supplement 2). We then examined the phenotype of Pf∆b9∆slarp-F7 and Pf∆b9∆slarp-G9 mutants during blood

124

stage and mosquito development. Asexual blood stage growth of Pf∆b9∆slarp parasites was normal as both

125

clones reached an asexual parasitemia between 0.5 and 5% during cloning within 21 days and Pf∆b9∆slarp

126

clones produced WT-like numbers of gametocytes, oocysts and sporozoites (Fig. 2).

127 128

Developmental arrest of Pf∆b9∆slarp GAPs in human hepatocytes

129

We next analyzed the development of Pf∆b9∆slarp in human hepatocytes using cultured primary

130

hepatocytes and uPA+/+-SCID mice engrafted with human hepatocytes (human liver-uPA-SCID mice)(24).

131

Pf∆b9∆slarp sporozoites showed normal gliding motility, hepatic cell traversal (Fig. 2) as well as invasion of

132

primary human hepatocytes, but parasites were completely absent in two independent experiments at day 2 up to

133

day 7 post-infection following inoculation of primary human hepatocytes with 40.000 Pf∆b9∆slarp F7 or G9

134

sporozoites (Fig. 3). Detailed analyses of 80 individual wells at day 4 post-infection did not result in

135

identification of a single developing parasite. The combined day 2 and day 4 data of‎Pf∆b9∆slarp indicated that

136

the‎ timing‎ of‎ arrest‎ is‎ similar‎ to‎ Pf∆b9 (15) and there had been complete arrest of liver-stage development,

137

similar to Pf∆slarp parasites.

138

In addition, human liver-uPA-SCID mice were intravenously inoculated with 1x106 WT or

139

PfΔb9Δslarp sporozoites. Two heterozygous uPA+/--SCID mice, not engrafted with human hepatocytes, served

140

as controls and were also challenged with P. falciparum sporozoites. Livers were collected either at 24 hpi or 5

141

days post-infection for detection of P. falciparum 18S DNA by quantitative real-time PCR (25). Both mice

142

infected with WT Pf and 1 of the 2 mice infected with PfΔb9Δslarp were positive for Pf 18S DNA at 24 hours

143

post-infection, demonstrating successful sporozoite infection in human hepatocytes (Fig. 3). A lower signal was

144

observed in PfΔb9Δslarp-infected mice at day 1 after infection compared to WT parasites, likely reflecting the

145

early time point of arrest of this GAP. All mice infected with Pf WT (3/3) showed a strong increase in parasite

146

18S DNA at day 5 post-infection, representing successful liver stage development. In contrast, none of the

147

human liver-uPA-SCID mice infected with PfΔb9Δslarp sporozoites showed 18S DNA higher than

148

heterozygous uPA+/--SCID mice, not engrafted with human hepatocytes that had been infected with PfΔb9Δslarp

149

sporozoites (Fig. 3). Although these studies were performed with a limited number of mice, these findings

8

150

indicate that PfΔb9Δslarp parasites can invade but do not develop in livers of humanized mice. Our combined

151

results demonstrate abrogation of development of PfΔb9Δslarp inside human hepatocytes.

9

152

Discussion

153

The Pf GAP PfΔb9Δslarp containing two gene deletions, is proposed as a whole-parasite malaria vaccine

154

candidate. Rationale and arguments are based on in vitro and in vivo experiments and supported by safety and

155

protection data with rodent Pb GAP with deletions of the orthologous genes. The rodent GAP PbΔb9Δslarp

156

completely arrested early in liver stage development in two different mouse strains after injection of very high

157

numbers of sporozoites. In addition, immunizations with PbΔb9Δslarp efficiently induced sterile and long-

158

lasting protective immunity in both BALB/c and C57BL/6 mice. Similarly, the Pf GAP‎ PfΔslarpΔb9,

159

completely aborted development in human hepatocytes one day after invasion, while sporozoites were fully

160

motile and invasive with infectivity comparable to Pf WT sporozoites. Importantly, asexual parasite growth and

161

production of salivary gland sporozoites in the mosquito were unaffected ensuring normal GAP production.

162

Pb∆b9∆slarp is to our knowledge the first completely attenuated rodent mutant in which multiple genes have

163

been deleted that are critical for two independent biological processes during liver stage development, i.e.

164

regulation of parasite genes/transcripts that play a role in early liver stage development stages (20, 21) and the

165

establishment of the PV within the infected hepatocyte(15).

166

A number of Pb and Py GAPs have previously been reported to arrest at different time points during

167

development in the liver (4, 6). These include GAPs based on genes essential for i) the formation and

168

maintenance of a parasitophorous vacuole (PV) (b9, p52, p36, uis3 and uis4; (15, 16, 26) and ii) type II fatty acid

169

synthesis (i.e. fabb/f, fabz, pdh e1α; (12, 27)), and iii) the regulation of gene expression in the liver stages

170

(sap1/slarp (19-21)). A critical safety requirement for GAPs in order to qualify as vaccine candidate is total

171

absence of blood infections during immunization and therefore the complete abrogation of liver-stage

172

development. Unfortunately many of the above mentioned target genes including p52, p36 and those involved in

173

type II fatty acid synthesis show a leaky phenotype, resulting in blood stage infections after administration of

174

high numbers of sporozoites. Incomplete liver stage arrest obviously disqualifies GAPs for further clinical

175

development for safety reasons.

176

In P. falciparum, GAPs have been generated that lack both the p52 and p36 genes (17, 18). In the Pb

177

rodent model this GAP was not completely attenuated (12). Similarly, this Pf GAP while severely attenuated by

178

the lack of both genes, a low percentage of parasites of this GAP are able to develop into mature liver-stage (12).

179

These observations indicate a partially redundant function for these proteins; indeed a breakthrough blood

180

infection was observed in one out of 6 volunteers after exposure to the bite of mosquitoes infected with

181

sporozoites of a PfΔp52Δp36 GAP (13) .

10

182

Since functional redundancy of related genes has been reported more often in Plasmodium (28-31), we

183

pursued the generation of GAPs from which multiple genes were removed from the genome each governing a

184

critical yet independent cellular process. The selection of those target genes excluded type II fatty acid synthesis

185

(FAS II) because P. falciparum mutants lacking FAS II enzymes fail to generate sporozoites inside the oocyst,

186

indicating that the FAS II pathway is essential for sporogony (32). The gene encoding liver stage antigen 1

187

(LSA-1) may be an attractive candidate but no orthologues are present in rodent or non-human primate

188

Plasmodium species precluding sufficient preclinical testing (33). The reverse is true for two published rodent

189

GAPs with deletions of the genes uis3 or uis4 of which unequivocal orthologues are absent in the P. falciparum

190

genome. Alternatively, genes encoding proteins with a role in late stage parasite liver development could be an

191

attractive target, since induction of protection by late arresting GAPs may be superior to early arresting GAPs (6,

192

34) However, late arresting GAPs are likely more risky and prone to breakthrough infection as shown for GAPs

193

lacking the genes palm or lisp (4).

194

Therefore, we decided to focus on early liver stage arrest and selected the newly identified b9 as a prime

195

candidate. PbΔb9 elicits long-lived protective immune responses in mice and only few breakthrough blood

196

infections occur in mice albeit less than were observed with PbΔp52Δp36 GAP sporozoites (12). The genes p52,

197

p36 and b9, all belong to the recently expanded 6-Cys family of Plasmodium proteins and may share a similar

198

function in formation or maintenance of the PV membrane at the interface of parasite and host cell. Indeed, a

199

triple gene-deletion mutant lacking p52, p36 and b9 is no more attenuated than a mutant lacking b9, suggesting

200

that these genes do not drive independent biological pathways (14-16). To date the early arresting slarp mutant is

201

the only rodent GAP with a Pf orthologue without a record of breakthrough blood infections in mice. Indeed, our

202

data confirm that rodent sporozoites lacking slarp are fully capable of hepatocyte invasion and formation of a PV

203

but completely abort development soon after invasion as previously reported(19-21). Here, we report for the first

204

time that P. falciparum mutants lacking slarp, i.e.‎PfΔslarp completely arrest at day 3 post-infection of primary

205

human hepatocytes while morphologically normal liver stage parasites are still observed at 48hpi.‎ PfΔb9

206

parasites arrest at a point in time before day 2 after hepatocyte invasion, with the exception of a single liver

207

schizont observed at a later time point(15). The multiple attenuated Pb∆b9∆slarp indeed passed our stringent

208

preclinical safety screen and no breakthrough blood infections were observed in all conditions tested. In

209

addition, we showed‎that‎immunization‎with‎Pb∆b9∆slarp sporozoites induced strong and sustained protective

210

immunity in BALB/c and C57BL/6 mice with similar efficacy as reported for mutant sporozoites lacking P52 (or

211

P52 and P36) or γ-radiated sporozoites (16, 35-37).

11

212

Live vaccine strains (attenuated by natural selection or genetic engineering) may be potentially released

213

into the environment. Therefore, safety issues concerning the medical as well as environmental aspects must be

214

considered including the absence of heterologous DNA sequences (in particular drug resistance genes) from the

215

genome of GAPs (38, 39). Thus, a Pf∆b9∆slarp GAP was generated free of a drug-resistance marker using

216

FRT/FLPe-recombinase methodology. This approach permits the removal of drug-resistance markers that were

217

introduced to generate the mutant and results in an altered genome that retains only two 34 nucleotide FRT

218

sequences. The removal of the drug resistance marker has the additional advantage that these parasites are easily

219

amenable to further genetic modification (22).

220

The Pf∆b9∆slarp GAP aborted early development in cultured primary human hepatocytes with a

221

phenotype‎and‎timing‎similar‎to‎Pf∆b9 and studies performed in a limited number of chimeric mice engrafted

222

with human hepatocytes confirm this arrest phenotype. From the combined Pb and Pf data one can conclude that

223

Δb9 attenuation phenotype induces highly effective protection, although it may at a low frequency produce a

224

breakthrough blood infection. Therefore the additional deletion of slarp in these mutants provides these parasites

225

with complete attenuation that is essential in order to proceed with human trials.

226

An important prerequisite for further downstream clinical development and manufacturing(11) is to

227

show that production of Pf∆b9∆slarp sporozoites is unabated and similar to WT parasites. We have shown that

228

the‎Pf∆b9∆slarp GAP produces WT numbers of sporozoites that are fully capable of infecting hepatocytes. In

229

addition we have produced aseptic, purified, cryopreserved PfΔb9Δslarp sporozoites (data not shown).

230

Preliminary data from a 6 day attenuation assay in HC-04 cells showed that like irradiated PfSPZ (5, 40), none of

231

the PfΔb9Δslarp sporozoites developed to mature liver stage parasites expressing PfMSP-1 (data not shown), as

232

do aseptic, purified, cryopreserved WT sporozoites(41).

233

In conclusion, we have generated a multiply attenuated Pf∆b9∆slarp GAP, free of any drug-resistance

234

gene, and demonstrated that Pf∆b9∆slarp sporozoites invade hepatocytes comparably to WT sporozoites and are

235

completely attenuated. These findings provide a solid foundation for clinical development and testing of a

236

PfSPZ∆b9∆slarp vaccine.

12

237

Material and methods

238 239

P. berghei reference parasite lines

240

The following reference lines of the ANKA strain of P. berghei were used: line cl15cy1(42, 43) and line

241

676m1cl1 (PbGFP-Luccon; see RMgm-29 in www.pberghei.eu). PbGFP-Luccon expresses a fusion protein of GFP

242

and Luciferase from the eef1a promoter (42, 44).

243 244

P. falciparum parasites and culture

245

For transfections, the parasite used was directly from a characterized good manufacturing process

246

(GMP) produced working cell bank (WCB) of the P. falciparum NF54 wild type strain (45), produced by

247

Sanaria Inc, identical to that described previously (5, 40, 41). Blood stages of wt,‎ PfΔslarp-a, PfΔslarp-b,

248

PfΔb9Δslarp-F7 and PfΔb9Δslarp-G9 were cultured in a semi-automated culture system using standard in vitro

249

culture conditions for P. falciparum and induction of gametocyte production in these cultures was performed as

250

previously described (46-48). Fresh human red blood cells and serum were obtained from Dutch National blood

251

bank (Sanquin Nijmegen, NL; permission granted from donors for the use of blood products for malaria

252

research). Cloning of transgenic parasites was performed by the method of limiting dilution in 96-well plates as

253

described (49). Parasites of the positive wells were transferred to the semi-automated culture system and cultured

254

for further phenotype and genotype analyses (see below).

255 256

Experimental animals

257

For P. berghei infections, female C57BL/6J and BALB/c (12 weeks old; Janvier France) and Swiss OF1

258

(8 weeks old Charles River) were used. All animal experiments with rodent parasites performed at the LUMC

259

(Netherlands) were approved by the Animal Experiments Committee of the Leiden University Medical Center

260

(DEC 07171; DEC 10099) and at the RUNMC (Netherlands) by the Radboud University Experimental Animal

261

Ethical Committee (RUDEC 2008-123, RUDEC 2008-148, RUDEC 2010-250, RUDEC 2011-022, RUDEC

262

2011-208). The Dutch Experiments on Animal Act is established under European guidelines (EU directive

263

86/609/CEE) regarding the Protection of Animals used for Experimental and Other Scientific Purposes.

264

Human liver-uPA-SCID mice (chimeric mice) were produced as described before (24). The study

265

protocol for infecting these mice with P. falciparum sporozoites was approved by the animal ethics committee of

266

the Faculty of Medicine and Health Sciences of the Ghent University.

267 13

268

Generation and genotyping of P. berghei mutants

269

To disrupt the P. berghei slarp gene (PBANKA_090210) a construct was generated using the adapted

270

‘Anchor-tagging’‎ PCR-based method as described (12) (Fig. 1 supplement 1). The two targeting fragments

271

(1195 bp and 823 bp) of slarp were amplified using genomic DNA (parasite line cl15cy1) as template with the

272

primer‎pairs‎5960/5961‎(5’target‎sequence)‎and‎5962/5963‎(3’target‎sequence).‎See‎Supplementary File 2a for

273

the sequence of the primers. Using this PCR-based targeting construct (pL1740) the mutant Pb∆slarp-a

274

(1839cl3) was generated in the PbGFP-Luccon reference line using standard methods of transfection and positive

275

selection with pyrimethamine (Fig. 1 supplement 1). The generation of the drug-selectable marker-free mutant

276

Pb∆b9∆sm (1309cl1m0cl2; RMgmDB no. 934) has been described by(15). This mutant, which contains a

277

disrupted b9 gene and is drug-selectable marker free, was used for deleting the slarp gene (PBANKA_090210).

278

To delete the slarp gene the gene-deletion construct pL1740 was used as described above. Using this construct

279

the mutant PbΔb9Δslarp (line 1844cl1) was generated in‎ the‎ Pb∆b9∆sm line using standard methods of

280

transfection and positive selection with pyrimethamine (Fig. 1).

281

Correct integration of the constructs into the genome of mutant parasites was analysed by diagnostic

282

PCR-analysis and Southern analysis of PFG-separated chromosomes as shown in Fig. 1 and Fig. 1 supplement

283

1. PFG-separated chromosomes were hybridized with a probe recognizing hdhfr or‎ the‎ 3’-UTR dhfr/ts of P.

284

berghei (43).

285 286

Generation and genotyping of P. falciparum mutants

287

The slarp gene (PF3D7_1147000) in P. falciparum WT parasites, (NF54wcb) was deleted using a

288

modified construct based on plasmid pHHT-FRT-(GFP)-Pf52 (22) (Fig. 2- supplement 1). Targeting regions

289

were generated by PCR using primers BVS179 and BVS180‎for‎the‎5’‎target‎region‎and‎primers‎BVS182‎and‎

290

BVS184‎for‎the‎3’‎target‎region‎(see‎Supplementary File 2b for‎primer‎sequences).‎The‎5’and‎3’‎target‎regions‎

291

were cloned into pHHT-FRT-(GFP)-Pf52 digested with BsiWI, BssHII and NcoI, XmaI, respectively, resulting in

292

the plasmid pHHT-FRT-GFP-slarp. The b9 gene (PF3D7_0317100) of PfΔslarp-b P. falciparum parasites was

293

deleted using a modified construct based on plasmid pHHT-FRT-(GFP)-Pf52 (22) (Fig. 2- supplement 1).

294

Targeting regions were generated by PCR using primers BVS84 and BVS85‎for‎the‎5’‎target‎region‎and‎primers‎

295

BVS88‎ and‎ BVS89‎ for‎ the‎ 3’‎ target‎ region.‎ The‎ 5’and‎ 3’‎ target‎ regions‎ were‎ cloned‎ into‎ pHHT-FRT-(GFP)-

296

Pf52 digested with NcoI, XmaI and MluI, BssHII resulting in the plasmid pHHT-FRT-GFP- b9. All DNA

297

fragments were amplified by PCR amplification (Phusion, Finnzymes) from genomic P. falciparum DNA (NF54

14

298

strain) and all PCR fragments were sequenced after TOPO TA (Invitrogen) sub-cloning. Transfection of WT

299

(NF54wcb) parasites with the plasmid pHHT-FRT-GFP-slarp and selection of mutant parasites was performed

300

as described (22) resulting in the selection of the parasite line PfΔslarp -a. The second PfΔslarp parasite line,

301

originating from an independent transfection, was subsequently transfected with pMV-FLPe to remove the drug-

302

selectable marker cassette using FLPe as described (22) and cloned resulting in the parasite clone PfΔslarp -b.

303

Subsequent transfection of PfΔslarp-b parasites with the plasmid pHHT-FRT-GFP-b9 and selection were

304

performed as described above resulting in the parasite line PfΔb9Δslarp. The parasite line PfΔb9Δslarp was

305

subsequently transfected with pMV-FLPe to remove the drug- selectable marker cassette using FLPe and cloned

306

as described above resulting in the cloned parasite lines PfΔb9Δslarp-F7 and PfΔb9Δslarp-G9 that are free of

307

drug-resistance markers.

308

Genotype analysis of PfΔslarp and PfΔb9Δslarp parasites was performed by Expand Long range

309

dNTPack (Roche) diagnostic, long-range, PCR (LR-PCR) and Southern blot analysis (Fig. 2- supplement 2).

310

Genomic DNA of blood stages of WT or mutant parasites was isolated and analyzed by LR-PCR using primer

311

pair p1, p2 (slarp) and p3, p4 (b9) (see Supplementary File 2b for primer sequences) for correct integration of

312

the constructs in the respective slarp and b9 loci by double cross-over homologous recombination. The LR-PCR

313

program has an annealing step of 48°C for 30 seconds and an elongation step of 62°C for 10-15 minutes. All

314

other‎ PCR‎ settings‎ were‎ according‎ to‎ manufacturer’s‎ instructions. PCR products were directly analyzed by

315

standard agarose gel electrophoresis or first digested with restriction enzymes for further confirmation of the

316

genotype and removal of resistance markers was confirmed by sequencing. For Southern blot analysis, genomic

317

DNA was digested with TaqI or RcaI restriction enzymes for analysis of integration into the slarp and b9 loci

318

respectively. Southern blot was generated by capillary transfer as described (50) and DNA was hybridized to

319

radioactive probes specific for the targeting regions used for the generation of the mutants and generated by PCR

320

(see above).

321

The presence or absence of slarp and b9 transcripts in WT and mutant sporozoites was analyzed by

322

reverse transcriptase-PCR (Fig. 2- supplement 2). Total RNA was isolated using the RNeasy mini Kit (Qiagen)

323

from 106 salivary gland sporozoites collected by dissection of mosquitoes 16 days after feeding with WT,

324

PfΔslarp-a, PfΔslarp-b, PfΔb9Δslarp-F7 and PfΔb9Δslarp -G9 parasites. Remaining DNA was degraded using

325

DNAseI (Invitrogen). cDNA was synthesized using the First Strand cDNA synthesis Kit for RT-PCR AMV

326

(Roche). As a negative control for the presence of genomic DNA, reactions were performed without reverse

327

transcriptase (RT-). PCR amplification was performed for regions of slarp using primers BVS290, BVS292 and

15

328

for regions of b9 using primers BVS286 and BVS288. Positive control was performed by PCR of 18S rRNA

329

using primers 18Sf and 18Sr.

330 331

Phenotype analyses of blood stages of P. berghei and P. falciparum mutants

332

Asexual multiplication rate and gametocyte production of P. berghei blood stages were determined as

333

described (12). The P. berghei mutants were maintained in Swiss mice. The multiplication rate of blood stages

334

and gametocyte production were determined during the cloning procedure (43) and were not different from

335

parasites of the reference ANKA lines. P. falciparum blood stage development and gametocyte production was

336

analyzed as described(22).

337 338

Analysis of P. berghei and P. falciparum sporozoite production and in vitro motility, hepatocyte traversal and

339

infectivity of sporozoites

340

Feeding of A. stephensi mosquitoes with P. berghei and P. falciparum, determination of oocyst

341

production and sporozoite collection as well as P. berghei gliding motility were performed as described (12). P.

342

falciparum gliding motility of sporozoites was determined as described(17, 51). P. falciparum cell traversal and

343

invasion of hepatocytes was determined in Huh7 cells and primary human hepatocytes respectively as

344

described(17). Infectivity of P. berghei sporozoites and development was determined in cultures of Huh7 cells as

345

described(15). For analysis of liver stage development by immunofluorescence, parasites were stained with the

346

following primary antibodies: anti-PbEXP1 (PBANKA_092670; raised in chicken(52)) and anti-PbHSP70

347

(PBANKA_081890; raised in mouse (53)). Infectivity of P. falciparum sporozoites and development was

348

analysed in primary human hepatocytes as described(17). Briefly for analysis of development by

349

immunofluorescence,

350

(PF3D7_0930300 (54)); anti-CSP (PF3D7_0304600; 3SP2) using double labeling. Anti-mouse secondary

351

antibodies, conjugated to Alexa-488 or Alexa-594 (Invitrogen) were used for visualization. Primary human

352

hepatocytes were isolated from healthy parts of human liver fragments which were collected during unrelated

353

surgery in agreement with French national ethical regulations(55) and after oral informed consent from adult

354

patients undergoing partial hepatectomy as part of their medical treatment (Service de Chirurgie Digestive,

355

Hépato-Bilio-Pancréatique et Transplantation Hépatique, Hôpital Pitié-Salpêtrière, Paris, France). The collection

356

and use of this material for the purposes of the study presented here were undertaken in accordance with French

357

national ethical guidelines under Article L. 1121-1‎of‎ the‎‘‘Code‎ de‎ la‎ Santé‎ Publique’’.‎Given‎ that‎ the‎ tissue‎

parasites

were

stained

with

the

following

primary

antibodies:

anti-HSP70

16

358

samples‎are‎classed‎as‎surgical‎waste,‎that‎they‎were‎used‎anonymously‎(the‎patient’s‎identity‎is‎inaccessible to

359

the researchers), and that they were not in any way genetically manipulated, article L. 1211-2 stipulates that their

360

use for research purposes is allowed provided that the patient does not express any opposition to the surgeon

361

prior to surgery and after being informed of the nature of the research in which they might be potentially

362

employed. Within this framework, the collection and use of this material was furthermore approved by the

363

Institutional Review Board (Comité de Protection des Personnes) of the Centre Hospitalo-Universitaire Pitié-

364

Salpêtrière, Assistance Publique-Hôpitaux de Paris, France.

365 366

Analysis of P. berghei sporozoite infectivity in mice and in vivo imaging of liver stage development

367

C57BL/6 or BALB/c mice were inoculated with sporozoites by intravenous injection of different

368

sporozoite numbers, ranging from 1x104–5x105. Blood stage infections were monitored by analysis of Giemsa-

369

stained thin smears of tail blood collected on day 4–14 after inoculation of sporozoites. The prepatent period

370

(measured in days post sporozoite infection) is defined as the day when a blood stage infection with a

371

parasitemia of 0.5–2% is observed. Liver stage development in live mice was monitored by real time in vivo

372

imaging of liver stages as described (56). Liver stages were visualized by measuring luciferase activity of

373

parasites (expressing luciferase under the eef1a promoter) in whole bodies of mice (57).

374 375

Immunizations of mice with P. berghei sporozoites

376

Prior to immunization, P. berghei sporozoites were collected at day 21-27 after mosquito infection by

377

hand-dissection. Salivary glands were collected in DMEM (Dulbecco's Modified Eagle Medium from GIBCO)

378

and homogenized in a homemade glass grinder. The number of sporozoites was determined by counting in

379

triplicate in a Bürker-Türk counting chamber using phase-contrast microscopy. BALB/c and C57BL/6 mice were

380

immunized by intravenous injection using different numbers of mutant sporozoites. BALB/c mice received one

381

immunization and C57BL/6 mice received three immunizations with two 7 day intervals. Immunized mice were

382

monitored for blood infections by analysis of Giemsa stained films of tail blood at day 4‐16 after immunization.

383

Immunized mice were challenged at different time points after immunization by intravenous injection of 1x104

384

sporozoites from the P. berghei ANKA reference line cl15cy1. In each experiment, age matched naive mice

385

were included to verify infectivity of the sporozoites used for challenge. After challenge, mice were monitored

386

for blood infections by analysis of Giemsa stained films of tail blood at day 4‐21.

387

17

388

Development of Pf Δb9Δslarp GAP in chimeric mice engrafted with human hepatocytes

389

Human liver-uPA-SCID mice were produced as described before (24). Briefly, within two weeks after

390

birth homozygous uPA+/+-SCID mice (25) were transplanted with approximately 106 cryopreserved primary

391

human hepatocytes obtained from a single donor (BD Biosciences, Erembodegem, Belgium). To evaluate

392

successful engraftment, human albumin was quantified in mouse plasma with an in-house ELISA (Bethyl

393

Laboratories Inc., Montgomery, TX). The study protocol was approved by the animal ethics committee of the

394

Faculty of Medicine and Health Sciences of the Ghent University. Human liver-uPA-SCID mice (n=10) and

395

non-chimeric heterozygous uPA+/--SCID mice (control, n=2) were intravenously injected with 106 fresh isolated

396

PfΔb9Δslarp -G9 or as a control WT sporozoites. One and 5 days post-infection livers were removed and each

397

liver was cut into 12 standardized sections and stored in RNALater (Sigma) at 4 °C until analysis as

398

described(25). From each part DNA was extracted to assess the parasite load by Pf18S qPCR and to assess the

399

number of human and mouse hepatocytes by Multiplex qPCR PTGER2 analysis(25).

400

18

401

References

402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452

1.

2. 3. 4. 5.

6. 7.

8.

9.

10.

11.

12.

13.

C. V. Plowe, P. Alonso, S. L. Hoffman, The potential role of vaccines in the elimination of falciparum malaria and the eventual eradication of malaria. The Journal of infectious diseases 200, 1646-1649 (2009). MalERA. (2010). N. S. Butler, A. M. Vaughan, J. T. Harty, S. H. Kappe, Whole parasite vaccination approaches for prevention of malaria infection. Trends Immunol 33, 247-254 (2012). S. M. Khan, C. J. Janse, S. H. Kappe, S. A. Mikolajczak, Genetic engineering of attenuated malaria parasites for vaccination. Current opinion in biotechnology 23, 908-916 (2012). S. L. Hoffman, P. F. Billingsley, E. James, A. Richman, M. Loyevsky, T. Li, S. Chakravarty, A. Gunasekera, R. Chattopadhyay, M. Li, R. Stafford, A. Ahumada, J. E. Epstein, M. Sedegah, S. Reyes, T. L. Richie, K. E. Lyke, R. Edelman, M. B. Laurens, C. V. Plowe, B. K. Sim, Development of a metabolically active, non-replicating sporozoite vaccine to prevent Plasmodium falciparum malaria. Human vaccines 6, 97-106 (2010). K. Nganou-Makamdop, R. W. Sauerwein, Liver or blood-stage arrest during malaria sporozoite immunization: the later the better? Trends in parasitology 29, 304-310 (2013). D. F. Clyde, H. Most, V. C. McCarthy, J. P. Vanderberg, Immunization of man against sporozite-induced falciparum malaria. The American journal of the medical sciences 266, 169177 (1973). S. L. Hoffman, L. M. Goh, T. C. Luke, I. Schneider, T. P. Le, D. L. Doolan, J. Sacci, P. de la Vega, M. Dowler, C. Paul, D. M. Gordon, J. A. Stoute, L. W. Church, M. Sedegah, D. G. Heppner, W. R. Ballou, T. L. Richie, Protection of humans against malaria by immunization with radiation-attenuated Plasmodium falciparum sporozoites. The Journal of infectious diseases 185, 1155-1164 (2002). M. Roestenberg, M. McCall, J. Hopman, J. Wiersma, A. J. Luty, G. J. van Gemert, M. van de Vegte-Bolmer, B. van Schaijk, K. Teelen, T. Arens, L. Spaarman, Q. de Mast, W. Roeffen, G. Snounou, L. Renia, A. van der Ven, C. C. Hermsen, R. Sauerwein, Protection against a malaria challenge by sporozoite inoculation. The New England journal of medicine 361, 468477 (2009). M. Roestenberg, A. C. Teirlinck, M. B. McCall, K. Teelen, K. N. Makamdop, J. Wiersma, T. Arens, P. Beckers, G. van Gemert, M. van de Vegte-Bolmer, A. J. van der Ven, A. J. Luty, C. C. Hermsen, R. W. Sauerwein, Long-term protection against malaria after experimental sporozoite inoculation: an open-label follow-up study. Lancet 377, 1770-1776 (2011). R. A. Seder, L. J. Chang, M. E. Enama, K. L. Zephir, U. N. Sarwar, I. J. Gordon, L. A. Holman, E. R. James, P. F. Billingsley, A. Gunasekera, A. Richman, S. Chakravarty, A. Manoj, S. Velmurugan, M. Li, A. J. Ruben, T. Li, A. G. Eappen, R. E. Stafford, S. H. Plummer, C. S. Hendel, L. Novik, P. J. Costner, F. H. Mendoza, J. G. Saunders, M. C. Nason, J. H. Richardson, J. Murphy, S. A. Davidson, T. L. Richie, M. Sedegah, A. Sutamihardja, G. A. Fahle, K. E. Lyke, M. B. Laurens, M. Roederer, K. Tewari, J. E. Epstein, B. K. Sim, J. E. Ledgerwood, B. S. Graham, S. L. Hoffman, Protection against malaria by intravenous immunization with a nonreplicating sporozoite vaccine. Science 341, 1359-1365 (2013). T. Annoura, I. H. Ploemen, B. C. van Schaijk, M. Sajid, M. W. Vos, G. J. van Gemert, S. Chevalley-Maurel, B. M. Franke-Fayard, C. C. Hermsen, A. Gego, J. F. Franetich, D. Mazier, S. L. Hoffman, C. J. Janse, R. W. Sauerwein, S. M. Khan, Assessing the adequacy of attenuation of genetically modified malaria parasite vaccine candidates. Vaccine, (2012). M. Spring, J. Murphy, R. Nielsen, M. Dowler, J. W. Bennett, S. Zarling, J. Williams, P. de la Vega, L. Ware, J. Komisar, M. Polhemus, T. L. Richie, J. Epstein, C. Tamminga, I. Chuang, N. Richie, M. O'Neil, D. G. Heppner, J. Healer, M. O'Neill, H. Smithers, O. C. Finney, S. A. Mikolajczak, R. Wang, A. Cowman, C. Ockenhouse, U. Krzych, S. H. Kappe, First-in-human evaluation of genetically attenuated Plasmodium falciparum sporozoites administered by bite of Anopheles mosquitoes to adult volunteers. Vaccine 31, 4975-4983 (2013).

19

453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507

14.

15.

16.

17.

18.

19.

20. 21.

22.

23.

24.

25.

26.

27.

28.

I. H. Ploemen, H. J. Croes, G. J. van Gemert, M. Wijers-Rouw, C. C. Hermsen, R. W. Sauerwein, Plasmodium berghei Deltap52&p36 Parasites Develop Independent of a Parasitophorous Vacuole Membrane in Huh-7 Liver Cells. PloS one 7, e50772 (2012). T. Annoura, B. C. van Schaijk, I. H. Ploemen, M. Sajid, J. W. Lin, M. W. Vos, A. G. Dinmohamed, D. K. Inaoka, S. R. Rijpma, G. J. van Gemert, S. Chevalley-Maurel, S. M. Kielbasa, F. Scheltinga, B. Franke-Fayard, O. Klop, C. C. Hermsen, K. Kita, A. Gego, J. F. Franetich, D. Mazier, S. L. Hoffman, C. J. Janse, R. W. Sauerwein, S. M. Khan, Two Plasmodium 6-Cys family-related proteins have distinct and critical roles in liver-stage development. Faseb J, (2014). M. R. van Dijk, B. Douradinha, B. Franke-Fayard, V. Heussler, M. W. van Dooren, B. van Schaijk, G. J. van Gemert, R. W. Sauerwein, M. M. Mota, A. P. Waters, C. J. Janse, Genetically attenuated, P36p-deficient malarial sporozoites induce protective immunity and apoptosis of infected liver cells. Proc Natl Acad Sci U S A 102, 12194-12199 (2005). B. C. van Schaijk, C. J. Janse, G. J. van Gemert, M. R. van Dijk, A. Gego, J. F. Franetich, M. van de Vegte-Bolmer, S. Yalaoui, O. Silvie, S. L. Hoffman, A. P. Waters, D. Mazier, R. W. Sauerwein, S. M. Khan, Gene disruption of Plasmodium falciparum p52 results in attenuation of malaria liver stage development in cultured primary human hepatocytes. PloS one 3, e3549 (2008). K. M. VanBuskirk, M. T. O'Neill, P. De La Vega, A. G. Maier, U. Krzych, J. Williams, M. G. Dowler, J. B. Sacci, Jr., N. Kangwanrangsan, T. Tsuboi, N. M. Kneteman, D. G. Heppner, Jr., B. A. Murdock, S. A. Mikolajczak, A. S. Aly, A. F. Cowman, S. H. Kappe, Preerythrocytic, live-attenuated Plasmodium falciparum vaccine candidates by design. Proc Natl Acad Sci U S A 106, 13004-13009 (2009). A. S. Aly, S. A. Mikolajczak, H. S. Rivera, N. Camargo, V. Jacobs-Lorena, M. Labaied, I. Coppens, S. H. Kappe, Targeted deletion of SAP1 abolishes the expression of infectivity factors necessary for successful malaria parasite liver infection. Mol Microbiol 69, 152-163 (2008). O. Silvie, K. Goetz, K. Matuschewski, A sporozoite asparagine-rich protein controls initiation of Plasmodium liver stage development. PLoS pathogens 4, e1000086 (2008). A. S. Aly, S. E. Lindner, D. C. MacKellar, X. Peng, S. H. Kappe, SAP1 is a critical posttranscriptional regulator of infectivity in malaria parasite sporozoite stages. Mol Microbiol 79, 929-939 (2011). B. C. van Schaijk, M. W. Vos, C. J. Janse, R. W. Sauerwein, S. M. Khan, Removal of Heterologous Sequences from Plasmodium falciparum Mutants Using FLPe-Recombinase. PloS one 5, e15121 (2010). J. W. Lin, T. Annoura, M. Sajid, S. Chevalley-Maurel, J. Ramesar, O. Klop, B. M. FrankeFayard, C. J. Janse, S. M. Khan, A novel 'gene insertion/marker out' (GIMO) method for transgene expression and gene complementation in rodent malaria parasites. PloS one 6, e29289 (2011). P. Meuleman, L. Libbrecht, R. De Vos, B. de Hemptinne, K. Gevaert, J. Vandekerckhove, T. Roskams, G. Leroux-Roels, Morphological and biochemical characterization of a human liver in a uPA-SCID mouse chimera. Hepatology 41, 847-856 (2005). L. Foquet, C. C. Hermsen, G. J. van Gemert, L. Libbrecht, R. Sauerwein, P. Meuleman, G. Leroux-Roels, Molecular detection and quantification of Plasmodium falciparum-infected human hepatocytes in chimeric immune-deficient mice. Malaria journal 12, 430 (2013). K. A. Kumar, P. Baxter, A. S. Tarun, S. H. Kappe, V. Nussenzweig, Conserved protective mechanisms in radiation and genetically attenuated uis3(-) and uis4(-) Plasmodium sporozoites. PloS one 4, e4480 (2009). A. M. Vaughan, M. T. O'Neill, A. S. Tarun, N. Camargo, T. M. Phuong, A. S. Aly, A. F. Cowman, S. H. Kappe, Type II fatty acid synthesis is essential only for malaria parasite late liver stage development. Cellular microbiology 11, 506-520 (2009). M. R. van Dijk, B. C. van Schaijk, S. M. Khan, M. W. van Dooren, J. Ramesar, S. Kaczanowski, G. J. van Gemert, H. Kroeze, H. G. Stunnenberg, W. M. Eling, R. W. Sauerwein, A. P. Waters, C. J. Janse, Three members of the 6-cys protein family of Plasmodium play a role in gamete fertility. PLoS pathogens 6, e1000853 (2010). 20

508 509 510 511 512 513 514 515 516 517 518 519 520 521 522 523 524 525 526 527 528 529 530 531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562

29.

30.

31.

32.

33.

34.

35. 36.

37.

38. 39. 40.

41.

42.

43.

J. W. Lin, P. Meireles, M. Prudencio, S. Engelmann, T. Annoura, M. Sajid, S. ChevalleyMaurel, J. Ramesar, C. Nahar, C. M. Avramut, A. J. Koster, K. Matuschewski, A. P. Waters, C. J. Janse, G. R. Mair, S. M. Khan, Loss-of-function analyses defines vital and redundant functions of the Plasmodium rhomboid protease family. Molecular microbiology 88, 318-338 (2013). K. Heiss, H. Nie, S. Kumar, T. M. Daly, L. W. Bergman, K. Matuschewski, Functional characterization of a redundant Plasmodium TRAP family invasin, TRAP-like protein, by aldolase binding and a genetic complementation test. Eukaryotic cell 7, 1062-1070 (2008). J. Liu, E. S. Istvan, I. Y. Gluzman, J. Gross, D. E. Goldberg, Plasmodium falciparum ensures its amino acid supply with multiple acquisition pathways and redundant proteolytic enzyme systems. Proceedings of the National Academy of Sciences of the United States of America 103, 8840-8845 (2006). B. C. van Schaijk, T. R. Kumar, M. W. Vos, A. Richman, G. J. van Gemert, T. Li, A. G. Eappen, K. C. Williamson, B. J. Morahan, M. Fishbaugher, M. Kennedy, N. Camargo, S. M. Khan, C. J. Janse, K. L. Sim, S. L. Hoffman, S. H. Kappe, R. W. Sauerwein, D. A. Fidock, A. M. Vaughan, Type II fatty acid biosynthesis is essential for Plasmodium falciparum sporozoite development in the midgut of Anopheles mosquitoes. Eukaryotic cell, (2013). S. A. Mikolajczak, J. B. Sacci, Jr., P. De La Vega, N. Camargo, K. VanBuskirk, U. Krzych, J. Cao, M. Jacobs-Lorena, A. F. Cowman, S. H. Kappe, Disruption of the Plasmodium falciparum liver-stage antigen-1 locus causes a differentiation defect in late liver-stage parasites. Cell Microbiol 13, 1250-1260 (2011). N. S. Butler, N. W. Schmidt, A. M. Vaughan, A. S. Aly, S. H. Kappe, J. T. Harty, Superior antimalarial immunity after vaccination with late liver stage-arresting genetically attenuated parasites. Cell host & microbe 9, 451-462 (2011). R. S. Nussenzweig, J. Vanderberg, H. Most, C. Orton, Protective immunity produced by the injection of x-irradiated sporozoites of plasmodium berghei. Nature 216, 160-162 (1967). B. Douradinha, M. R. van Dijk, R. Ataide, G. J. van Gemert, J. Thompson, J. F. Franetich, D. Mazier, A. J. Luty, R. Sauerwein, C. J. Janse, A. P. Waters, M. M. Mota, Genetically attenuated P36p-deficient Plasmodium berghei sporozoites confer long-lasting and partial cross-species protection. Int J Parasitol 37, 1511-1519 (2007). M. Labaied, A. Harupa, R. F. Dumpit, I. Coppens, S. A. Mikolajczak, S. H. Kappe, Plasmodium yoelii sporozoites with simultaneous deletion of P52 and P36 are completely attenuated and confer sterile immunity against infection. Infect Immun 75, 3758-3768 (2007). EMEA, C. f. m. p. f. h. u. (CHMP), Ed. (2006), chap. 4.2. J. Frey, Biological safety concepts of genetically modified live bacterial vaccines. Vaccine 25, 5598-5605 (2007). J. E. Epstein, K. Tewari, K. E. Lyke, B. K. Sim, P. F. Billingsley, M. B. Laurens, A. Gunasekera, S. Chakravarty, E. R. James, M. Sedegah, A. Richman, S. Velmurugan, S. Reyes, M. Li, K. Tucker, A. Ahumada, A. J. Ruben, T. Li, R. Stafford, A. G. Eappen, C. Tamminga, J. W. Bennett, C. F. Ockenhouse, J. R. Murphy, J. Komisar, N. Thomas, M. Loyevsky, A. Birkett, C. V. Plowe, C. Loucq, R. Edelman, T. L. Richie, R. A. Seder, S. L. Hoffman, Live attenuated malaria vaccine designed to protect through hepatic CD8(+) T cell immunity. Science 334, 475-480 (2011). M. Roestenberg, E. M. Bijker, B. K. Sim, P. F. Billingsley, E. R. James, G. J. Bastiaens, A. C. Teirlinck, A. Scholzen, K. Teelen, T. Arens, A. J. van der Ven, A. Gunasekera, S. Chakravarty, S. Velmurugan, C. C. Hermsen, R. W. Sauerwein, S. L. Hoffman, Controlled human malaria infections by intradermal injection of cryopreserved Plasmodium falciparum sporozoites. The American journal of tropical medicine and hygiene 88, 5-13 (2013). C. J. Janse, B. Franke-Fayard, G. R. Mair, J. Ramesar, C. Thiel, S. Engelmann, K. Matuschewski, G. J. van Gemert, R. W. Sauerwein, A. P. Waters, High efficiency transfection of Plasmodium berghei facilitates novel selection procedures. Mol Biochem Parasitol 145, 6070 (2006). C. J. Janse, J. Ramesar, A. P. Waters, High-efficiency transfection and drug selection of genetically transformed blood stages of the rodent malaria parasite Plasmodium berghei. Nature protocols 1, 346-356 (2006). 21

563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 597 598 599 600 601 602 603 604 605

44.

45.

46. 47.

48.

49.

50. 51. 52. 53.

54.

55.

56.

57.

B. Franke-Fayard, H. Trueman, J. Ramesar, J. Mendoza, M. van der Keur, R. van der Linden, R. E. Sinden, A. P. Waters, C. J. Janse, A Plasmodium berghei reference line that constitutively expresses GFP at a high level throughout the complete life cycle. Mol Biochem Parasitol 137, 23-33 (2004). T. Ponnudurai, A. D. Leeuwenberg, J. H. Meuwissen, Chloroquine sensitivity of isolates of Plasmodium falciparum adapted to in vitro culture. Tropical and geographical medicine 33, 50-54 (1981). T. Ifediba, J. P. Vanderberg, Complete in vitro maturation of Plasmodium falciparum gametocytes. Nature 294, 364-366 (1981). T. Ponnudurai, A. H. Lensen, A. D. Leeuwenberg, J. H. Meuwissen, Cultivation of fertile Plasmodium falciparum gametocytes in semi-automated systems. 1. Static cultures. Trans R Soc Trop Med Hyg 76, 812-818 (1982). T. Ponnudurai, A. H. Lensen, G. J. Van Gemert, M. P. Bensink, M. Bolmer, J. H. Meuwissen, Infectivity of cultured Plasmodium falciparum gametocytes to mosquitoes. Parasitology 98 Pt 2, 165-173 (1989). S. Thaithong, in Application of genetic engineering to research on tropical disease pathogens with special reference to Plasmodia, S. Panyim, P. Wilairat, Y. Yuthavong, Eds. (Bangkok, 1985), pp. 379-387. J. Sambrook, W. D. Russel, Molecular Cloning: a laboratory manual. (Cold Spring Harbor Laboratory press, Cold Spring Harbor, ed. 3rd, 2001). M. J. Stewart, J. P. Vanderberg, Malaria sporozoites leave behind trails of circumsporozoite protein during gliding motility. J Protozool 35, 389-393 (1988). A. Sturm, V. Heussler, Live and let die: manipulation of host hepatocytes by exoerythrocytic Plasmodium parasites. Med Microbiol Immunol 196, 127-133 (2007). A. K. Mueller, N. Camargo, K. Kaiser, C. Andorfer, U. Frevert, K. Matuschewski, S. H. Kappe, Plasmodium liver stage developmental arrest by depletion of a protein at the parasitehost interface. Proc Natl Acad Sci U S A 102, 3022-3027 (2005). L. Renia, D. Mattei, J. Goma, S. Pied, P. Dubois, F. Miltgen, A. Nussler, H. Matile, F. Menegaux, M. Gentilini, et al., A malaria heat-shock-like determinant expressed on the infected hepatocyte surface is the target of antibody-dependent cell-mediated cytotoxic mechanisms by nonparenchymal liver cells. Eur J Immunol 20, 1445-1449 (1990). L. C. Gouagna, M. van der Kolk, W. Roeffen, J. P. Verhave, W. Eling, R. Sauerwein, C. Boudin, Role of heat-labile serum factor or host complement in the inhibition of Plasmodium falciparum sporogonic stages in Anopheles stephensi by gametocyte carriers' serological factors. Parasitology 134, 1315-1327 (2007). I. H. Ploemen, S. Chakravarty, G. J. van Gemert, T. Annoura, S. M. Khan, C. J. Janse, C. C. Hermsen, S. L. Hoffman, R. W. Sauerwein, Plasmodium liver load following parenteral sporozoite administration in rodents. Vaccine, (2012). I. H. Ploemen, M. Prudencio, B. G. Douradinha, J. Ramesar, J. Fonager, G. J. van Gemert, A. J. Luty, C. C. Hermsen, R. W. Sauerwein, F. G. Baptista, M. M. Mota, A. P. Waters, I. Que, C. W. Lowik, S. M. Khan, C. J. Janse, B. M. Franke-Fayard, Visualisation and quantitative analysis of the rodent malaria liver stage by real time imaging. PloS one 4, e7881 (2009).

22

606

Acknowledgments

607

We would like to thank the following people from RUMC (Nijmegen) for technical support: Claudia Lagarde,

608

Alex Ignacio, Daniëlle Janssen, Rianne Siebelink-Stoter, Wouter Graumans, Jolanda Klaassen, Laura Pelser-

609

Posthumus, Astrid Pouwelsen and Jacqueline Kuhnen; and the following people from LUMC (Leiden) for

610

technical support: Jai Ramesar, Jing-wen Lin and Hans Kroeze. We acknowledge the Sanaria Manufacturing

611

Team for the GMP produced working cell bank of PfNF54.

612

613

Author Contributions

614

Conceived and designed the experiments: BVS IP TA LF CJ SK RS. Performed the experiments: BVS IP TA

615

MV LF GJVG SCM MVDVB MS JFF AL. Analyzed the data: BVS IP TA MV CJ DM SK CCH. Contributed

616

Reagents/materials/analysis tools: SH GLR PM. Wrote the paper: BVS IP CJ SK RS. All authors commented

617

and corrected the manuscript.

618 619

Competing Interests

620

The authors have declared that no competing interests exist. However, SLH, is CEO of Sanaria Inc., a

621

biotechnology company focused on whole sporozoite malaria vaccines.

622

Funding: This study was performed within the framework of Top Institute Pharma (Netherlands) project: T4-

623

102. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of

624

the manuscript.

625 626

Data and Materials availability

627

The materials described in this study must be acquired through a material transfer agreement.

628

23

629

Table 1: Protection of mice after immunization with P. berghei Pb∆b9 or Pb∆b9∆slarp sporozoites Mouse Strain

Pb Mutant

Day of Challenge a

Immunization regimes No. protected/ No challenged 10k b 5k

BALB/c Pb∆b9 Pb∆b9∆slarp

10 10

C57Bl6

1k

10/10c 20/20

18/20 10/10

8/10 20/20

50/20/20k d

10/10/10k

1/1/1k

Pb∆b9

10 90 180 365

4/4 5/5 9/9 e 5/11

nd

nd

Pb∆b9∆slarp

10 180

nd 6/6

10/10 nd

6/10 nd

630 631

a

Number of days post last immunization;104 wild type sporozoites were injected by IV route.

632

b

Immunization dose: number of sporozoites x1000.

633

c

Protected/total # of immunized mice (%); protection was 0/15 in naive control BALB/c and 0/10 in C57BL/6

634

mice.

635

d

Immunization dose with 7 day intervals between immunizations.

636

e

Immunization dose 50/10/20k with 7 day intervals between immunizations.

637

nd= not done.

638 639

Table 2: Breakthrough blood-stage infections after intravenous injection of Pb∆slarp and

640

Pb∆b9∆slarp sporozoites. Infectiona Spz x 103

Breakthrough blood infection/total # mice

Pre-patent periodc (days)

WTb

10

5/5

4-5

Pb∆slarp Pb∆slarp Pb∆b9∆slarp

50 25 25

0/5 0/10 0/10

WTb

10

5/5

Pb∆slarp Pb∆slarp Pb∆b9∆slarp Pb∆b9∆slarp

500 200 200 150

0/5 0/10 0/10 0/5

Mouse Strain

Mutant

BALB/c

C57BL/6

4-5

641 642

a

Inoculation dose of sporozoites administered IV

643

b

P. berghei ANKA strain: line cl15cy1

644

c

Day with parasitemia of 0.5–2%

24

645 646

LEGENDS TO FIGURES

647 648

Fig. 1: Generation and genotype analyses of P. berghei mutant PbΔb9Δslarp

649

A. Generation‎of‎mutant‎PbΔb9Δslarp.‎For‎PbΔb9Δslarp the DNA-construct pL1740 was generated containing

650

the positive/negative selectable marker cassette hdhfr/yfcy. This construct was subsequently used to generate the

651

mutant‎PbΔb9Δslarp in‎the‎PbΔb9Δsm mutant. See Supplementary File 2a for the sequence of the primers.

652

B. Diagnostic PCR and Southern analysis of Pulse Field Gel (PFG)-separated chromosomes of mutant

653

PbΔb9Δslarp confirming correct disruption of the slarp and the b9 locus. See Supplementary File 2a for the

654

sequence‎of‎the‎primers‎used‎for‎the‎selectable‎marker‎gene‎(SM);‎5’-integration‎event‎(5’);‎3’-integration event

655

(3’)‎and‎the‎slarp and the b9 ORF. For Southern analysis, PFG-separated chromosomes were hybridized using a

656

3’UTR‎pbdhfr probe that recognizes the construct integrated into P. berghei slarp locus on chromosome 9, the

657

endogenous locus of dhfr/ts on‎chromosome‎7‎and‎a‎3’UTR‎pbdhfr probe that recognizes the construct integrated

658

into P. berghei b9 locus on chromosome 8. In addition, the chromosomes were hybridized with the hdhfr probe

659

recognizing the integrated construct into the slarp locus on chromosome 9.

660

C. Development of liver-stages in cultured hepatocytes as visualized by staining with antibodies recognizing the

661

parasitophorous vacuole membrane (anti-EXP1; green) and the parasite cytoplasm (anti-HSP70; red). Nuclei are

662

stained with Hoechst-33342. Hpi: hours post infection. Scale bar represents 10µm.

663 664

Fig. 1- supplement 1: Generation and genotype analyses of P. berghei mutant PbΔslarp-a

665

A. Generation‎of‎mutant‎PbΔslarp-a.‎For‎PbΔslarp-a the DNA-construct pL1740 was generated containing the

666

positive/negative selectable marker cassette hdhfr/yfcy. This construct was subsequently used to generate the

667

mutant‎PbΔslarp-a (1839cl3) in the PbGFP-Luccon reference line. See Supplementary File 2a for the sequence

668

of the primers.

669

B. Diagnostic PCR and Southern analysis of Pulse Field Gel (PFG)-separated‎chromosomes‎of‎mutant‎Δslarp-a

670

confirming correct disruption of the slarp-locus. See Supplementary File 2afor the sequence of the primers

671

used‎for‎the‎selectable‎marker‎gene‎(SM);‎5’-integration‎event‎(5’);‎3’-integration‎event‎(3’)‎and‎the‎slarp ORF.

672

Mutant‎PbΔslarp-a has been generated in the reference P. berghei ANKA line PbGFP-Luccon which has a gfp-

673

luciferase gene integrated into the silent 230p locus (PBANKA_030600) on chromosome 3. For Southern

674

analysis, PFG-separated‎ chromosomes‎ were‎ hybridized‎ using‎ a‎ 3’UTR‎ pbdhfr probe that recognizes the

675

construct integrated into P. berghei slarp locus on chromosome 9, the endogenous locus of dhfr/ts on

676

chromosome 7 and the gfp-luciferase gene integrated into chromosome 3. In addition, the chromosomes were

677

hybridized with the hdhfr probe recognizing the integrated construct into the slarp locus on chromosome 9.

678

C. Real time in vivo imaging of ∆slarp luciferase-expressing liver-stage parasites in C57BL/6 mice at 24, 35 and

679

45 hours post infection. C57BL/6 mice were IV injected with either 5x104 Pb-GFPLuccon sporozoites (n=5)

680

resulting in a full liver infection (upper panel: representative image of WT infected mice), or with 5 x10 5

681

Pb∆slarp-a sporozoites (n=5) (lower panel: representative image of Pb∆slarp-luc infected mice).

682 683

Fig. 2. Phenotypes of P. falciparum Pf∆slarp and Pf∆b9∆slarp parasites

25

684

A. Gametocyte, oocyst and sporozoite production. Gametocyte numbers (stage II and IV-V) per 1000

685

erythrocytes at day 8 and day 14 after the start of gametocyte cultures. Exflagellation (Exfl) of male gametocytes

686

in stimulated samples from day 14 cultures (++ score = >10 exflagellation centers per microscope field at 400x

687

magnification). Median number of oocysts at day 7, IQR is the inter quartile range and sporozoite (day 21)

688

production (x1000) in A. stephensi mosquitoes.

689

B. Gliding motility of P. falciparum WT‎ (cytochalasin‎ D‎ treated‎ and‎ untreated),‎ PfΔslarp-b,‎ PfΔb9Δslarp-F7

690

and PfΔb9Δslarp-G9 parasites. Gliding motility was quantified by determining the mean percentage ± standard

691

deviation of parasites that exhibited gliding motility by producing‎ characteristic‎ CSP‎ trails‎ (≥‎ 1‎ circles)‎ or‎

692

parasites that did not produce CSP trails (0 circles).

693

C. Cell traversal ability of P. falciparum NF54,‎PfΔslarp-b and‎PfΔb9Δslarp-F7 sporozoites as determined by

694

FACS counting of Dextran positive Huh7 cells. Shown is the mean percentage ± standard deviation of FITC

695

positive cells. Dextran control (control): hepatocytes cultured in the presence of Dextran but without the addition

696

of sporozoites.

697 698

Fig. 2- supplement 1: Consecutive gene deletion of slarp and b9 in P. falciparum

699

Schematic representation of the genomic loci of A. slarp (PF11_0480; PF3D7_1147000) on chromosome 11

700

(Chr. 11) and B. b9 (PFC_0750w; PF3D7_0317100) on chromosome 3 (Chr. 3) of wild-type (wt; NF54wcb),

701

PfΔslarp and‎ PfΔb9Δslarp gene deletion mutants‎ before‎ (PfΔslarp a and PfΔb9Δslarp) and after the FLPe

702

mediated removal of the hdhfr::gfp resistance‎marker‎(PfΔslarp b and PfΔb9Δslarp clones F7/G9) respectively.

703

The constructs for the targeted deletion of slarp (pHHT-FRT-

704

GFP slarp) and b9 (pHHT-FRT-GFP-B9) contain two FRT sequences (red triangles) that are recognized by

705

FLPe. P1, P2 and P3, P4 primer pairs for LR-PCR analysis of slarp and b9 loci respectively; T (TaqI) and R

706

(RcaI): restriction sites used for Southern blot analysis and sizes of restriction fragments are indicated; cam:

707

calmodulin; hrp: histidine rich protein; hsp: heatshock protein; fcu: cytosine deaminase/uracil phosphoribosyl-

708

transferase; hdhfr::gfp: human dihydrofolate reductase fusion with green fluorescent protein; pbdt: P.berghei

709

dhfr terminator.

710 711

Fig. 2- supplement 2: Genotype analysis of the generated Pf∆slarp and Pf∆b9∆slarp parasites

712

A. Long range PCR analysis of genomic DNA from WT, PfΔslarp and PfΔb9Δslarp asexual parasites confirms

713

the slarp gene deletion and consecutive gene deletions of both slarp and b9 respectively and subsequent removal

714

of the hdhfr::gfp resistance marker. The PCR products are generated using primers P1,P2 for slarp and P3,P4 for

715

b9 (see A and B respectively; for primer sequences see primer table in Supplementary File 2b) and PCR

716

products are also digested with restriction enzymes x (XmaI) and kx (KpnI/XcmI) respectively for confirmation

717

(i.e. slarp LR-PCR‎ product‎ sizes:‎ WT,‎ 12kb,‎ is‎ undigested;‎ Δslarp-a, 5.4kb is digested into 1.3kb and 4.0kb

718

fragments,‎Δslarp-b, 2.4kb is digested into 1.3kb and 1.1kb fragments. b9 LR-PCR product sizes: WT, 5.5kb, is

26

719

digested‎ into‎ 756bp,‎ 793bp‎ and‎ 4.0kb‎ fragments;‎ Δb9-b, 2.6kb is digested into 756bp, 793bp and 1.1kb

720

fragments).

721

B. Southern‎ analysis‎ of‎ restricted‎ genomic‎ DNA‎ from‎ WT,‎ PfΔslarp-a, PfΔslarp-b,‎ PfΔb9Δslarp-F7 and

722

PfΔb9Δslarp-G9 asexual parasites. DNA was digested with restriction enzyme (E: TaqI) and probed with the

723

5’slarp targeting‎region‎(P:‎5’ slarp-T; see A) on the left side of the slarp Southern‎or‎probed‎with‎the‎3’slarp

724

targeting‎ region‎ (P:‎ 3’ slarp-T; see A) on the right side of the slarp panel. For analysis of the b9 integration

725

DNA was digested with restriction enzymes (E: RcaI) and probed with‎the‎5’b9 targeting‎region‎(P:‎5’ b9-T; see

726

A) on the right panel. The expected fragment sizes are indicated in panel A.

727

C. RT-PCR analysis showing absence of b9 and slarp transcripts in P. falciparum PfΔslarp-a, PfΔslarp-b,

728

PfΔb9Δslarp-F7 and PfΔb9Δslarp-G9 mutant sporozoites. PCR amplification using purified sporozoite RNA

729

was performed either in the presence or absence of reverse transcriptase (RT+ or RT-, respectively) and

730

generated the expected 506bp and 580bp fragments for slarp and b9 respectively, the positive control was

731

performed by PCR of 18S rRNA using primers 18Sf/18Sr (for primer sequences see Supplementary File 2b)

732

and generated the expected 130bp fragment.

733 734

Fig. 3. Development of P. falciparum Pf∆slarp and Pf∆b9∆slarp parasites in human primary hepatocytes

735

A. In vitro invasion of P. falciparum wt,‎ PfΔslarp-a,‎ PfΔslarp-b,‎ PfΔb9Δslarp-F7 and PfΔb9Δslarp-G9

736

sporozoites in primary human hepatocytes. Invasion is represented as the mean ratio ± standard deviation of

737

extra- and intracellular sporozoites by double staining at 3 and 24 hours post infection, determined after 3 wash

738

steps to remove sporozoites in suspension.

739

B. Immunofluorescence assay of PfΔslarp-b parasites in human primary hepatocytes at 3 and 24 hours post

740

infection. Parasites are visualized by staining with anti-PfCSP antibodies (green; Alexa-488) and parasite and

741

hepatocyte nuclei are stained with DAPI (blue). Images were photographed on an Olympus FV1000 confocal

742

microscope. Scale bar represents 5μm.

743

C. Development of P. falciparum wt,‎PfΔslarp-a PfΔslarp-b‎(top‎panel),‎PfΔb9Δslarp-F7 and PfΔb9Δslarp-G9

744

(bottom panel) liver-stages in primary human hepatocytes following inoculation with 40.000 sporozoites. From

745

day 2 to 7 the mean number ± standard deviation of parasites per 96-well was determined by counting parasites

746

stained with anti-P. falciparum HSP70 antibodies. The bottom panel represents experiments performed in

747

primary human hepatocytes from 2 different donors. No parasites present (NP).

748

D. Development‎of‎liver‎stages‎of‎PfΔb9Δslarp GAP in chimeric mice engrafted with human hepatocytes. Mice

749

were infected with 106 wt‎or‎PfΔb9Δslarp-G9 sporozoites by intravenous inoculation. At 24 hours or at 5 days

750

after sporozoite infection, livers were collected from the mice and the presence of parasites determined by qPCR

751

of the parasite-specific 18S DNA. uPA HuHEP; chimeric homozygous uPA+/+-SCID mice engrafted with human

27

752

hepatocytes. As controls, uPA mice; heterozygous uPA+/--SCID mice not engrafted with human hepatocytes

753

were used.

28

Figure 1

A 5’ target

SM (hdhfr/yfcu) 875 bp 6128 6127 slarp

5’ target

DNA construct pL1740

3’ target

slarp locus (PBANKA_090210) in ȴb9ȴsm genome (1309cl1m0cl2)

3’ target

1631 bp 1075 bp 1287 bp 6347 4771 6125 6349 6346 6126 5’ target

SM (hdhfr/yfcu)

3’ target

1225 bp 4438 4437 b9

B

Wďѐb9ѐslarp

Disrupted locus (1844cl1)

Wild-type locus (PBANKA_080810)

Wďѐb9ѐslarp

WT

SM 5’ 3’ O B9 SM 5’ 3’ O B9 kb 1.5 1

Chr 9 8

7 3’UTR hdhfr dhfr

ɲyWϭ WT (24 hpi)

Wďѐb9ѐslarp (24 hpi)

ɲ,^WϳϬ

,ŽĞĐŚƐƚ

Merge

Figure 2

A Phenotypes of P. falciparum WĨѐslarp and WĨѐď9ѐslarp parasites. Parasite

WT WĨȴslarp-a WĨȴslarp-b WĨȴb9ȴslarp-F7 WĨȴb9ȴslarp-G9

Gametocyte Gametocyte stage IV-V (range) stage II (range) 6,6 (1-13)

51 (29-61)

8,8 (3-15) 9,1 (2-27) 8,2 (1-140 8,6 (4-12)

58 (41-65) 49 (27-70) 39 (22-47) 49 (27-65)

džĨů.

% Infected Oocyst Mean no. of mosquitoes production sporozoites (Infected/dissected) (IQR) (std)

++ ++ ++ ++ ++

27 (12-42)

95% (104/110)

48 (23-95)

23 (8-59) 36 (17-59) 34 (20-54) 33 (13-64)

93% (37-40) 96% (106/110) 96% (77/80) 94% (75/80)

50 (22-97) 77 (22-174) 81 (33-106) 62 (22-105)

C

B Gliding motility

,ĞƉĂƚŽĐLJƚĞƚƌĂǀĞƌƐĂů

Figure 3

A

wt WĨȴslarp-a WĨȴslarp-b WĨȴb9ȴslarp-F7 WĨȴb9ȴslarp-G9

B

ɲ^W

C

DAPI

ĞǀĞůŽƉŵĞŶƚŽĨWĨȴslarp parasites wt Pfȴslarp-a Pfȴslarp-b

EŽ͘ůŝǀĞƌƐƚĂŐĞƉĂƌĂƐŝƚĞƐ

WT (3 hpi)

WĨѐslarp-b (3 hpi)

WT (24 hpi)

Day 2

NP

NP

NP

NP

NP

3

4

5

6

7

ĞǀĞůŽƉŵĞŶƚŽĨWĨȴb9ȴslarp parasites wt Pfȴslarp-b Pfȴb9ȴslarp-F7 Pfȴb9ȴslarp -G9

WĨѐslarp-b (24 hpi) NP

Day 2

D

NP

NP

NP

NP

3

4

5

6

Total Pf/106 ,Ƶ,ĞƉ

wt WĨȴb9ȴslarp-G9

Day

1

1

5 uPA ,Ƶ,W

5

5 uPA

NP

7

Lihat lebih banyak...

Comentarios

Copyright © 2017 DATOSPDF Inc.