Running title: Model to down-select human malaria vaccines

Similar documents
of Nebraska - Lincoln

Novel ELISA method as exploratory tool to assess immunity induced by radiated attenuated sporozoites to decipher protective immunity

Infecting Anopheles stephensi With Rodent Malaria Parasites Alida Coppi & Photini Sinnis

Gliding Motility Assay for P. berghei Sporozoites

Arrested oocyst maturation in Plasmodium parasites. lacking type II NADH:ubiquinone dehydrogenase

Developmentally Regulated!nfectivity of Malaria Sporozoites for Mosquito Salivary Glands and the Vertebrate Host

PLASMODIUM MODULE 39.1 INTRODUCTION OBJECTIVES 39.2 MALARIAL PARASITE. Notes

A. Effect upon human culture 1. Control of malaria has contributed to world=s population explosion 2. Africans brought to U.S.

PRINCIPAL INVESTIGATOR: Dr. Jetsumon (Sattabongkot) Prachumsri

INVESTIGATING THE MOTILITY OF PLASMODIUM

Supporting Online Material for

CIRCUMSPOROZOITE PROTEINS OF HUMAN MALARIA PARASITES PLASMODIUM FALCIPARUM AND PLASMODIUM VIVA,F*

Quantitative Dynamics of Plasmodium yoelii Sporozoite Transmission by Infected Anopheline Mosquitoes

Enzootic Bovine Leukosis: Milk Screening and Verification ELISA: VF-P02210 & VF-P02220

Sera from 2,500 animals from three different groups were analysed:

Fluoroquinolones ELISA KIT

Blood protozoan: Plasmodium

Received 6 December 2000/Returned for modification 29 January 2001/Accepted 26 March 2001

Diurnal variation in microfilaremia in cats experimentally infected with larvae of

Parasitology Departement Medical Faculty of USU

ENVIRACOR J-5 aids in the control of clinical signs associated with Escherichia coli (E. coli) mastitis

Blood protozoan: Plasmodium

Identification of an AP2-family Protein That Is Critical for Malaria Liver Stage Development

Chimeric Plasmodium falciparum parasites expressing Plasmodium vivax circumsporozoite protein fail to produce salivary gland sporozoites

Control And Preventive Study Of Brucellosis By Using Lipopolysacharide Sub Unit Vaccine Brucella abortus Strain S-19

Visit ABLE on the Web at:

Understanding Epidemics Section 3: Malaria & Modelling

COMMITTEE FOR VETERINARY MEDICINAL PRODUCTS

Evaluation of Different Antigens in Western Blotting Technique for the Diagnosis of Sheep Haemonchosis

Dog vaccination with EgM proteins against Echinococcus granulosus

CelTOS, a novel malarial protein that mediates transmission to mosquito and vertebrate hosts

Neutralization of Micrurus distans distans venom by antivenin (Micrurus fulvius)

FELINE CORONAVIRUS (FCoV) [FIP] ANTIBODY TEST KIT

alaria Parasite Bank Collection sites of P. falciparum isolates PARASITE BIOLOGY

Malaria. This sheet is from both sections recording and includes all slides and diagrams.

Effect of ingested human antibodies induced by RTS, S/AS01 malaria vaccination in children on Plasmodium falciparum

Use of a novel adjuvant to enhance the antibody response to vaccination against Staphylococcus aureus mastitis in dairy heifers.

Epigenetic regulation of Plasmodium falciparum clonally. variant gene expression during development in An. gambiae

Inactivation of Burkholderia mallei in equine serum for laboratory use.

Doug Carithers 1 William Russell Everett 2 Sheila Gross 3 Jordan Crawford 1

EUROPEAN REFERENCE LABORATORY (EU-RL) FOR BOVINE TUBERCULOSIS WORK-PROGRAMME PROPOSAL Version 2 VISAVET. Universidad Complutense de Madrid

Malaria in the Mosquito Dr. Peter Billingsley

SUPPLEMENTARY INFORMATION

BIO Parasitology Spring 2009

Developmental Biology of Sporozoite-Host. Malaria: Implications for Vaccine Design. Javier E. Garcia, Alvaro Puentes and Manuel E.

A Cysteine Protease Inhibitor of Plasmodium berghei Is Essential for Exo-erythrocytic Development

ANNEX I SUMMARY OF PRODUCT CHARACTERISTICS. Medicinal product no longer authorised

VOL. XXIII NO. II THE JOURNAL OF ANTIBIOTICS 559. ANTIBIOTIC 6640.* Ill

COMMITTEE FOR VETERINARY MEDICINAL PRODUCTS

Malaria parasites: virulence and transmission as a basis for intervention strategies

RESULT OF STUDYING SOME ACUTE PHASE PROTEINS AND CORTISOL IN PREGNANT EWES

THE SPOROZOITE ENZYME-LINKED IMMUNOSORBENT ASSAY : APPLICATION IN MALARIA EPIDEMIOLOGY

II. MATERIALS AND METHODS

THE ABUNDANCE AND INFECTION STATUS OF ANOPHELES MOSQUITOES IN LOUDOUN COUNTY, VIRGINIA

Feeding Original XPC TM can help reduce Campylobacter in broilers and turkeys

Efficacies of fenbendazole and albendazole in the treatment of commercial turkeys artificially infected with Ascaridia dissimilis

ANNEX I SUMMARY OF PRODUCT CHARACTERISTICS 1/18

International Journal of Advances in Pharmacy and Biotechnology Vol.3, Issue-2, 2017, 1-7 Research Article Open Access.

OPTIMIZATION OF PK/PD OF ANTIBIOTICS FOR RESISTANT GRAM-NEGATIVE ORGANISMS

Plasmodium vivax: A Monoclonal Antibody Recognizes a Circumsporozoite Protein Precursor on the Sporozoite Surface

Heartworm Disease in Dogs

THE TRANSMISSION EFFICIENCY OF PLASMODIUM YOELII INFECTED MOSQUITOES

Comparative efficacy of DRAXXIN or Nuflor for the treatment of undifferentiated bovine respiratory disease in feeder cattle

Improving Growth and Yield of Commercial Pheasants Through Diet Alteration and Feeding Program

Testing for antimicrobial activity against multi-resistant Acinetobacter baumannii. For. Forbo Flooring B.V. Final Report. Work Carried Out By

ANNEX I SUMMARY OF PRODUCT CHARACTERISTICS

An experimental study on triclabendazole resistance of Fasciola hepatica in sheep

R-factor mediated trimethoprim resistance: result of two three-month clinical surveys

Diagnosis of Heartworm (Dirofilaria immitis) Infection in Dogs and Cats by Using Western Blot Technique

Neither Mosquito Saliva nor Immunity to Saliva Has a Detectable Effect on the Infectivity of Plasmodium Sporozoites Injected into Mice

Burn Infection & Laboratory Diagnosis

Gye and Cramer (1919) found that the ionizable salts of calcium injected together with the washed spores of Cl. tetani or of certain

Malaria parasite exit from the host erythrocyte: A two-step process requiring extraerythrocytic proteolysis

Period of study: 12 Nov 2002 to 08 Apr 2004 (first subject s first visit to last subject s last visit)

PCR detection of Leptospira in. stray cat and

Study population The target population for the model were hospitalised patients with cellulitis.

Efficacy of Moxidectin 6-Month Injectable and Milbemycin Oxime/Lufenuron Tablets Against Naturally Acquired Toxocara canis Infections in Dogs*

COMMITTEE FOR VETERINARY MEDICINAL PRODUCTS

COMMITTEE FOR VETERINARY MEDICINAL PRODUCTS

SENSITIVE AND -RESISTANT TUBERCLE BACILLI IN LIQUID MEDIUM SENSITIVITY TESTS

Implantation of Tissue Chambers in Turkeys: A Pilot Study

GUIDE TO INFECTION CONTROL IN THE HOSPITAL. Antibiotic Resistance

Bovine Brucellosis Control of indirect ELISA kits

Transmission success of the malaria parasite Plasmodium mexicanum into its vector: role of gametocyte density and sex ratio

Cattle Serologically Positive for Brucella abortus Have Antibodies

Toxocariasis: serological diagnosis by enzyme

Effects of Moxifloxacin PK-PD and drug interactions on its use in the Treatment of Tuberculosis(TB)

Reproductive Vaccination- Deciphering the MLV impact on fertility

Chapter 1 COPYRIGHTED MATERIAL. Introduction to Veterinary Pathology. What is pathology? Who does pathology?


Effects of an Ivermectin Otic Suspension on Egg Hatching of the Cat Ear Mite, Otodectes cynotis, in Vitro*

No-leaching. No-resistance. No-toxicity. >99.999% Introducing BIOGUARD. Best-in-class dressings for your infection control program

Plasmodium 18S rrna of intravenously administered sporozoites does not persist in peripheral blood

ANNEX I SUMMARY OF PRODUCT CHARACTERISTICS

23 Plasmodium coatneyi Eyles, Fong, Warren, Guinn, Sandosham, and Wharton, 1962

Evaluation of a computerized antimicrobial susceptibility system with bacteria isolated from animals

Vaccine. Diagnostic and Vaccine Chapter. J.H. Wolfram a,, S.K. Kokanov b, O.A. Verkhovsky c. article info abstract

AMOXICILLIN AND CLAVULANIC ACID TABLETS Draft proposal for The International Pharmacopoeia (February 2018)

Medical Genetics and Diagnosis Lab #3. Gel electrophoresis

EVALUATION OF THE SENSITIVITY AND SPECIFICITY OF THE EHRLICHIA CANIS DIAGNOSTIC TEST: Anigen Rapid E.canis Ab Test Kit

Transcription:

CVI Accepts, published online ahead of print on 27 March 2013 Clin. Vaccine Immunol. doi:10.1128/cvi.00066-13 Copyright 2013, American Society for Microbiology. All Rights Reserved. 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 Transgenic parasites stably expressing full-length Plasmodium falciparum circumsporozoite protein as a model for vaccine down-selection in mice using sterile protection as endpoint Michael D. Porter 1, Jennifer Nicki 1, Christopher D. Pool 1, Margot DeBot 1, Ratish M. Illam 1, Clara Brando 2, Brooke Bozick 2, Patricia De La Vega 1, Divya Angra 1, Roberta Spaccapelo 3, Andrea Crisanti 4, Jittawadee R. Murphy 2, Jason W. Bennett 1, Robert J. Schwenk 1, Christian F. Ockenhouse 1 and Sheetij Dutta 1*. 1 Malaria Vaccine Development Branch / 2 Entomology Branch, Walter Reed Army Institute of Research, Silver Spring, USA. 3 Università degli Studi di Perugia, Perugia, Italy. 4 Imperial College London, London, UK. *Corresponding Author: Laboratory of Structural Vaccinology, Rm. 3W61, Walter Reed Army Institute of Research, 503 Robert Grant Ave, Silver Spring MD 20910. USA Phone: 301-319-9154; E-mail: Sheetij.dutta@us.army.mil Running title: Model to down-select human malaria vaccines Disclaimer: Research was conducted in compliance with the animal welfare act and other federal statutes and regulations relating to animal experiments and adherent to the principals stated in the Guide for the Care and Use of Laboratory Animals, NRC publication, 1996 edition. The opinions expressed in this publication are those of the authors and are not to be construed as the official position of the United States Department of the Army or Department of Defense. 1

33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 Abstract Circumsporozoite protein (CSP) of Plasmodium falciparum is a protective human malaria vaccine candidate. There is an urgent need for models that can rapidly down-select novel CSP-based vaccine candidates. In the present study, the mouse-mosquito transmission cycle of a transgenic Plasmodium berghei malaria parasite stably expressing a functional full-length P. falciparum CSP was optimized to consistently produce infective sporozoites for protection studies. A minimum sporozoite challenge dose was established, and protection was defined as the absence of blood stage parasites 14 days after intravenous challenge. Specificity of protection was confirmed by vaccinating mice with multiple CSP constructs of differing lengths and compositions. Constructs that induced high NANP repeat ELISA titers were protective and the degree of protection was dependent on the antigen dose. There was a positive correlation between antibody avidity and protection. The antibodies in the protected mice recognized the native CS protein on the parasites and showed sporozoite invasion inhibitory activity. Passive transfer of anti-csp antibodies into naïve mice also induced protection. Thus we have demonstrated the utility of a mouse efficacy model to down-select human CSP based vaccine formulations. 2

56 57 Introduction 58 59 60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 A malaria parasite infected mosquito injects approximately 10-200 sporozoites into the vertebrate host s skin during a blood meal (1). These sporozoites travel to the liver where each successful invasion of a liver hepatocyte yields approximately 30,000 blood-stage merozoites (2). Hence immune interventions that block sporozoite invasion are currently thought to be the most effective way to protect against malaria. The most abundant P. falciparum sporozoite surface protein is the 397 amino acid circumsporozoite protein (CSP). Genetic analysis of CSP from multiple Plasmodium species reveals a highly conserved structure (3). The central region of CSP is comprised of species-specific repeats that are flanked by an amino-terminal (N-) region containing a conserved five amino acid sequence, region I, and a carboxyl-terminal (C-) region, region II, that contains a conserved cell-adhesive motif similar to that observed in the mammalian thrombospondin protein (4). Malaria still causes extensive morbidity and mortality, and the development of a vaccine against this parasite is an urgent research priority. Because of its abundance and exposed location on the sporozoite surface, CSP has been widely investigated as a candidate malaria vaccine antigen. RTS,S, the most advanced human malaria vaccine candidate to date, contains the central repeats and the cysteine-rich C-terminal region of Plasmodium falciparum CSP (PfCSP). Vaccination with RTS,S induces sterile protection against experimental sporozoite challenge in about 50% of the vaccinees (5); this vaccine is now 3

79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100 101 undergoing advanced Phase III trials at multiple centers in Africa. Although RTS,S marked a critical tipping point as a proof-of-concept recombinant protein vaccine against malaria, the efficacy and duration of RTS,S-based protection is not sufficient to eradicate the disease. It is hypothesized that a full-length CSPbased vaccine might confer improved protection by eliciting immune responses to the N-terminal region of CSP, antibodies against which have been associated with protection from disease (6). Although non-human primates are considered the best model to predict human protection against malaria, these models are costly and often require splenectomy of the monkey. Moreover, the few P. falciparum strains shown to infect new world monkeys do not match the commonly used vaccine strain, 3D7 (7-9). Although transgenic monkey parasites expressing a full length P. falciparum CSP would be ideal, none are currently available for use. Several reports suggest that transgenic rodent parasites expressing the P. falciparum CSP gene are viable and infective in mice. One such parasite in which the central repeat region of P.berghei CSP was exchanged with that of P. falciparum has recently been used to evaluate the protective efficacy of P. falciparum CSP vaccines in mice (10, 11). However, such parasites provide no information on the protective role of the N- and C-terminal epitopes of CSP. Furthermore, the outcome of these murine challenge studies may be clouded by measuring the reduction of parasite-specific RNA rather than directly assessing protective efficacy. To address these issues, we have optimized a vaccine 4

102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 evaluation model based on a previously described transgenic parasite in which the full-length P. berghei CSP gene was replaced with P. falciparum CSP (4). This parasite line was originally produced to study the structure-function relationship of CSP during salivary gland and hepatocyte invasion. While the full-length CSP transgenic sporozoites showed reduced salivary gland invasion, the mouse infectivity of these parasites was similar to that of wild-type P. berghei sporozoites (4). Our data indicate that this full-length transgenic parasite can be used for the rapid down-selection of recombinant P. falciparum CSP-based vaccines using sterile protection as an endpoint. Materials and Methods Recombinant CSP vaccination. The genes for the CSP constructs used to immunize mice were codon-optimized for high-level expression in E. coli using the 3D7 strain CSP sequence (accession number XM_001351086.1). The histidine-tagged proteins from the soluble fraction were purified to homogeneity using chromatography columns (purification process to be presented elsewhere). The endotoxin content of the vaccine proteins was less than 5 EU/μg as measured by the LAL Endotoxin Assay (Associates of Cape Cod, East Falmouth, MA). The antigens were mixed with Montanide ISA720 adjuvant (Seppic Inc., Paris, France) in a 3:7 antigen:adjuvant v/v ratio, and the formulation was emulsified by vigorous vortexing for 10-15 min. One hundred microliters of vaccine was administered to C57BL/6 mice (The Jackson Laboratory, Bar Harbor, ME) intra-peritoneally, and three immunizations were given at 2 wk 5

125 126 127 128 129 130 131 132 133 134 135 136 137 138 139 140 141 142 143 144 145 146 147 intervals. Mice were bled 2 wk after each vaccination. IgG used in the passive transfer experiment was pooled from three rabbits that were vaccinated three times with 100 μg of N-(NANP) 19 -C protein adjuvanted with Montanide ISA720 at 4 wk intervals. Adaptation of Tr-Pb in the rodent and mosquito host. Transgenic P. berghei parasites (Tr-Pb), previously described by Tewari et al. (4), were obtained and injected intra-peritoneally into three outbred mice. Six days following inoculation, the presence of blood-stage parasitemia was confirmed by Giemsastained blood smear examination. Blood from one infected mouse was collected, diluted 1:1 with PBS, and 0.2 ml was injected into three naive mice to start the first blood passage cycle (BP-1) (Fig. 1A). Three days post-infection, 200-300 female Anopheles stephensi mosquitoes were allowed to feed on two of the three BP-1 infected mice while the third mouse was used to generate a cohort of BP-2 infected mice via infected blood transfer. This process was repeated until the fourth blood passage, at which point three naïve mice were inoculated via infectious sporozoite bite, thus generating a new set for which the four blood passage cycle was repeated. Oocyst and sporozoite rating. The oocyst and sporozoite data from 10 cycles, each consisting of four blood passages, were collected and statistically analyzed to determine infectivity differences between the wild-type and transgenic parasites. At 8-11 days after feeding on parasitemic mice, 10 mosquitoes were randomly sampled from each carton, and midguts were removed and stained with 6

148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 methylene blue. Oocyst ratings were defined as: 0 = 0 oocysts; 1 = 1-25; 2 = 26-50; 3 = 51-100; and 4 = 100+ oocysts. Eighteen days after the infectious blood meal, salivary glands dissected from 10 mosquitoes were examined for the presence and estimated quantity of sporozoites. Sporozoite ratings were defined as: 0 = 0 sporozoites; 1 = 1-100 sporozoites per mosquito; 2 = 100-1000; 3 = 1000-10,000; and 4 = 10,000+ sporozoites. Isolation of sporozoites for challenge and passive transfer. Fresh naïve mouse serum was isolated from whole blood using serum separator tubes (Becton Dickinson and Co., Franklin Lakes, NJ) and was used to supplement RPMI-1640 (BioWhittaker, Walkersfield, MD) to a final concentration of 5% v/v (RPMI/serum). Sporozoites were collected using the Ozaki method (12). Briefly, a hole was punctured at the bottom of a 0.5 ml siliconized eppendorf tube and plugged with a small amount of glass wool, and the tube was placed inside a larger, 1.5 ml siliconized eppendorf collection tube. Mosquitoes were killed by immersing in 70% ethanol for approximately 1 min, rinsed with tap water, and bathed in a small volume of RPMI/serum. Dissections were performed in a drop of RPMI/serum under a dissecting microscope. Heads plus salivary glands were gently separated from the thorax and up to 50 heads were added per Ozaki tube. One hundred microliters RPMI/serum was added followed by centrifugation at 9000xg for 2 min. The sporozoite pellet was re-suspended in the eluate and transferred into a separate eppendorf tube. Another 100 µl of RPMI/serum was added to the original tube which was re-spun at 9000xg for 2 min, and the pellet was combined with the initial recovery. The sporozoites were 7

171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 stored on ice after isolation. Sporozoites were counted on a hemocytometer, diluted to 25,000 per ml in RPMI/serum, and 100 μl of this suspension was injected intravenously into the lateral tail vein of each mouse. In the passive transfer experiment, mice were administered intraperitoneally 1 mg of Protein-G (GE Healthcare, Pittsburgh, PA) purified rabbit anti-pfcsp IgG or 1 mg of rabbit anti-pfama1 control IgG. A second dose of IgG was given the next morning, and mice were challenged with 2500 sporozoites 5 h later. Monitoring mouse infection. Levels of parasitemia in the mice were monitored daily using thin blood smears from day 5 up to day 14 post-challenge. Blood smears were fixed with methanol and stained with Giemsa. Positive infection in mice was defined as the appearance of two parasites in 25 high powered fields (100X magnification). Mice found to be infected with blood stages of the parasite (not protected) were sacrificed and mice that did not develop blood stage parasitemia by day 14 were reported as protected. ELISA. Immulon 2HB TM plates (Thermo Scientific, Rochester, NY) were coated overnight at 4 o C with either 50 ng/well recombinant CSP [N-(NANP) 19 -C] or 20 ng/well (NANP) 6 peptide. Plates were washed with PBS containing 0.05% Tween- 20 (PBS/T) and blocked with PBS containing 1% casein for 1 h. Fifty microliters of diluted primary antibody was added to the wells in duplicate for 2 h at 22 o C, plates were washed 3x with PBS/T, and 50 μl/well of a 1:15,000 dilution of peroxidase-conjugated anti-mouse IgG (Southern Biotech, Birmingham, AL) was added. After a 1 h incubation, plates were washed 4x with PBS/T and developed 8

194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 using 50 μl/well ABTS peroxidase substrate system (KPL, Gaithersburg, MD) for 1 h. OD 415 was measured using a Synergy4 microplate reader (Biotek, Highland Park, VT) and endpoint titer, defined as the serum dilution that resulted in OD 415 of 1.0, was calculated using Gen5 TM software (Biotek). The avidity ELISA was performed similarly, with 75 μl of a 1:1000 serum dilution added to the blocked wells for 1 h. After washing, 100 μl of either 6M urea or PBS was incubated in the wells for 10 min. Plates were washed and the remaining ELISA technique was performed as described above. Avidity index was defined as the ratio of OD 415 obtained in the presence and absence of 6M urea. Indirect immunofluorescence assay (IFA). Sporozoites were placed on an IFA slide, air-dried, and fixed with chilled methanol for 1 min. Wells were blocked with PBS containing 5% BSA, and serial dilutions of the test antibodies were added and incubated for 2 h. Slides were washed 3x with PBS and incubated with a 1:100 dilution of fluorescein-labeled anti-mouse IgG (Southern Biotech) for 1 h. The slides were washed again and anti-fade solution (Molecular Probes Inc., Eugene, OR) was applied. Fluorescence was observed under a UV microscope (200X magnification). An adjuvant control serum pool diluted 1:100 was used as a negative control. The mouse PbCSP-specific mab, 4B10, and PfCSP-specific mab, 49-1B2, were used in the study. 214 9

215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 Sporozoite invasion inhibition assay. Mouse serum was tested for its ability to inhibit sporozoite entry into hepatocytes (13). Briefly, wells of a LabTeK glass chamber slide were coated with ECL attachment matrix (Millipore, Billerica, MA), 45,000 HepG2-A16 cells were added, and the slide was incubated overnight at 37 C and 5% CO 2. Fifty microliters of the appropriate serum dilution, along with 50 µl of P. falciparum NF54 sporozoite suspension (25,000 sporozoites), was added per well. The chamber slides were then incubated for 3 h at 37 C and 5% CO 2. Slides were washed with PBS and fixed with chilled methanol. Sporozoites that had invaded hepatocytes were visualized by staining with a P. falciparum CSP-specific mab (49-1B2) followed by the addition of a 1:200 dilution of HRP conjugated goat anti-mouse IgG secondary antibody (KPL, Gaithersburg, MD). Color was developed using DAB reagent (KPL) and slides were mounted with Permount TM mounting medium (Fisher Scientific, Hampton, NH). The number of intracellular sporozoites per well was counted with a phase-contrast microscope at 200X magnification. The percent inhibition of sporozoite invasion was calculated as (invasion events in the medium control invasion events in the test serum)/invasion events in the medium control x 100. Native CS western blot. Proteins from 50,000 sporozoites per well were separated by SDS-PAGE, electrophoretically transferred to a nitrocellulose membrane, and blocked with PBS/C for 1 h. The blot was incubated with a 1:1000 dilution of immune serum, washed with PBS/T, and incubated with a 10

238 239 240 1:5000 dilution of alkaline phosphatase conjugated anti-mouse secondary antibody (Southern Biotech) for 1h. The blots were washed again and developed with NBT-BCIP substrate tablets (Roche, Nutley, New Jersey). 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 Statistical analysis. Data were plotted using Excel TM (Microsoft, Redmond, WA) and Prism Graphpad TM (Graphpad Software Inc., La Jolla, CA). Unpaired groups of data were compared for significant differences utilizing a Mann- Whitney non-parametric test. P-values <0.05 were used to determine statistical significance. Results Adapting the Tr-Pb parasite in mouse-mosquito hosts. Data from 10 independent mosquito-mouse transmission cycles showed no significant difference in the percentage of mosquitoes infected with oocysts and oocyst ratings between transgenic (Tr-Pb) and wild-type (WT-Pb) P. berghei infected mosquitoes (Fig. 1B, C). However, the percentage of mosquitoes infected with salivary gland sporozoites and the mean sporozoite ratings (on a 0-4 scale) were significantly lower in Tr-Pb infected mosquitoes (Mann Whitney, p<0.05 for both comparisons). Initial batches obtained by feeding mosquitoes on mice that were infected by blood stages typically yielded <500 sporozoites per mosquito as compared to >10,000 sporozoites for the WT-Pb infected mosquitoes. The 11

261 262 263 264 265 266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 transmission cycle was then modified, eliminating the blood stage transmission, and mosquitoes were fed only on mice that had been infected directly by mosquito bite. This modification selected for a line that was better adapted to produce salivary gland sporozoites, increasing the average yield to ~2,000 sporozoites per mosquito (Fig. 1D). However, no change in the rodent infectivity of the adapted parasites was notable. The genetic replacement of the PbCSP gene with PfCSP was confirmed by IFA using a Pf-specific monoclonal antibody (mab), 49-1B2. WT-Pb sporozoites reacted with a PbCSP-specific mab, 4B10, but only weak reactivity was observed with the PfCSP mab, 49-1B2 (Fig. 1E). In contrast, the Tr-Pb sporozoites reacted with only the PfCSP mab. Challenge route and dose optimization. To reproduce the natural mode of transmission used for experimental human challenges, either 5 or 10 Tr-Pb sporozoite-infected mosquitoes were fed on individual C57BL/6 mice. This challenge route yielded variable infectivity data that were highly dependent upon the sporozoite load of individual mosquitoes in the carton. A subcutaneous challenge using isolated sporozoites also showed low infectivity despite a high sporozoite inoculum (Table 1, Expts. I and II). The challenge route was then modified to an intravenous (IV) administration of defined numbers of isolated sporozoites. IV injection of 2500 sporozoites successfully infected all inoculated mice (Expt. II), with patency typically observed by six days postchallenge (Expt. III, IV). As compared to C57BL/6 mice, the BALB/c strain was more resistant to Tr-Pb parasites and required a 4-5 times higher sporozoite 12

284 285 286 dose than the C57BL/6 strain. Based on these data, all ensuing challenge experiments were performed with C57BL/6 mice using an IV dose of 2500 Tr-Pb sporozoites. 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 Utility and validity of the Tr-Pb model. A major goal of this study was to establish the effectiveness of the Tr-Pb sporozoite model for the down-selection of different CSP-based vaccine antigens. To accomplish this the following four closely related PfCSP constructs were prepared and tested: Construct N- (NANP) 19 -C contained the N-terminal region, 19 repeats and C-terminal cysteine rich region; construct N-(NANP) 5 -C contained the N-terminal, 5 repeats and C-terminal; construct (NANP) 18 -C contained 18 repeats and C- terminal; and construct Cterm contained only the C-terminal region of PfCSP (Fig. 2A, 2B). Groups of nine mice received three intra-peritoneal vaccinations of 2.5 μg antigen formulated in Montanide ISA720 at 2 wk intervals. At 2 weeks post third vaccination, mice were challenged with 2500 Tr-Pb sporozoites. Antibody titers were measured against a repeat peptide (NANP) 6 or the near full-length construct N-(NANP) 19 -C protein coated onto ELISA plates. As expected, the five NANP repeat-containing construct induced lower repeat specific antibody titers than constructs that contained either 19 or 18 repeats (Fig. 2C, 2D). Interestingly, the (NANP) 18 -C construct lacking the N-terminus exhibited faster kinetics of repeat-specific antibody acquisition after the second vaccination, as compared to constructs (N-(NANP) 19 -C and N-(NANP) 5 -C) that contained the N-terminal region (Fig. 2C). Similarly, after the third vaccination, 13

307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 the magnitude of repeat-specific antibodies in the (NANP) 18 -C-immunized group was higher than that observed for the N-(NANP) 19 -C and N-(NANP) 5 -C groups (p=0.02 and 0.0001 respectively) (Fig. 2D). The presence of the N-terminal region, however, did not globally reduce the immunogenicity of N-(NANP) 19 -C and N-(NANP) 5 -C constructs as their group titers were similar to the (NANP) 18 - C-induced titers when measured against the near full-length CSP and the Cterm plate antigens (p>0.2 for all comparisons) (Fig. 2D). Upon challenge with Tr- Pb parasites none of the adjuvant control, N-(NANP) 5 -C, or Cterm vaccinated mice were protected (open triangles in Fig. 2D). In contrast, 4 of 9 N-(NANP) 19 - C and 4 of 9 (NANP) 18 -C vaccinated mice were protected. Titration of vaccine-induced protection. Groups of mice received, at 2 wk intervals, three intraperitoneal injections of incremental doses of N-(NANP) 19 -C protein formulated in Montanide ISA720. The geometric mean antibody titer against all three plate antigens at 2 wk post third vaccination increased with increasing antigen dose (Fig. 3A). Mice were challenged with 2500 sporozoites 2 wk after the third vaccination. While no protection was observed in the adjuvant control group, 1 of 6 mice in the 1 μg dose group; 5 of 7 in the 2.5 μg group; 3 of 7 in the 5 μg group; and 7 of 7 mice in the 10 μg group were protected. Hence the degree of protection increased with escalating antigen dose. 327 328 329 Association between antibody titer and protection against Tr-Pb. IgG subclasses and antibody avidity were measured using the sera of individual mice from the various immunization regimens. The protection status of a mouse 14

330 331 332 333 334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 correlated with antibody titer against the near full-length protein and the repeat peptide. As seen in Fig. 3B, 70% of the mice that had ELISA titers >10,000 against both full-length and repeat antigens were protected, in contrast to only 28% protection in mice with low repeat titers and 21% protection in mice with low antibody titers to both full-length and repeat antigens. Mice in the three highest dose groups (10, 5, and 2.5 µg CSP) were tested for antibody avidity. Avidity index was associated with protection as 100% of mice with >80% avidity index and >10,000 full-length ELISA titer were protected (Fig. 3C). Low levels of IgG2 induced by Montanide ISA720 formulations precluded any correlation of subclasses with protection. Biological activity of CSP antibodies. Antibodies against the N-(NANP) 19 -C protein showed positive reactivity with fixed sporozoites by indirect immunofluorescence assay (IFA), and pooled sera showed increasing IFA titers with escalating antigen dose (Fig. 4A, 4B). Antibodies against N-(NANP) 19 -C immunization showed positive reactivity with a ~60 kda band on a western blot against an extract of sporozoite infected mosquito salivary glands (Fig. 4C). In a sporozoite invasion inhibition assay, anti-n-(nanp) 19 -C sera (1:100 dilution) caused ~90% inhibition of sporozoite invasion of HepG2-A16 cells as compared to ~20% inhibition by normal mouse serum (Fig. 4D). Inhibition of sporozoite invasion activity increased with antigen dose and ~90% inhibition was observed using a serum pool from the 10 μg N-(NANP) 19 -C group in which all mice had been protected (Fig. 4E). 15

352 353 354 355 356 357 358 359 360 361 362 363 364 365 366 367 368 369 370 371 372 373 374 Passive transfer of PfCSP-specific antibodies confers protection in the Tr- Pb challenge model. Mice were administered purified rabbit IgG against the N- (NANP) 19 -C vaccine and challenged with 2500 Tr-Pb sporozoites. All five anti- AMA1 IgG recipient mice developed parasitemia by day 5, while 3 of 5 mice that received anti-csp IgG were protected through day 14 post-challenge. In another experiment, Tr-Pb sporozoites were pre-incubated with a 1:4 dilution of either mouse serum against N-(NANP) 19 -C or control normal mouse serum. After a 5 min incubation at 37 o C, 3000 sporozoites were injected intravenously into each of three naïve C57BL/6 mice. Two of three anti-csp group mice were protected through day 14, while all three control mice became positive by day 6 post-challenge. Discussion There is substantial evidence that antibodies and T-cell mediated immune responses to CSP can protect against the pre-erythrocytic stage of malaria (14). Owing to the escalating costs and regulatory constraints associated with human vaccine trials, and due to the limitations of primate models of P. falciparum sporozoite challenge (7-9), there is an urgent need to develop rodent models for the routine down-selection of second-generation vaccine formulations of PfCSP vaccines. Here we present studies that led to the optimization of one such rodent challenge model based on transgenic P. berghei parasites that express fulllength PfCSP (4). Our data showed that this model can distinguish between the 16

375 376 377 protective efficacies of several closely related CSP antigens using a highly stringent endpoint of sterile protection following intravenous injection of sporozoites. 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 A notable area of concern with parasites that are transgenic for full-length CSP is reduced sporozoite salivary gland invasion. Rodent parasites expressing either Plasmodium falciparum or the avian Plasmodium gallinaceum CSP develop normally in the midgut yet show significantly reduced salivary gland burdens in the mosquito. In such studies, the N-terminal and repeat regions of CSP were specifically implicated in migration to the salivary glands (15, 16). We found in our studies that switching from blood-stage passages to a more natural vectorborne mosquito to mouse passage yielded an adapted line of Tr-Pb that routinely produced sufficient (although still relatively low) numbers of infective sporozoites for challenge experiments. Other criteria for a successful challenge included sporozoite yields >1000 per mosquito, addition of 5% freshly collected mouse serum to the dissection medium, minimizing the dissection time to under one hour, and storing dissected sporozoites on ice until injection. The observation that constructs containing the N-terminus induced lower levels of repeat antibodies than those lacking the N-terminus requires further investigation as it may reflect the shielding of CSP domains by the N-terminal region that is known to occur during sporozoite passage from the mosquito to the liver (17). Although the relative immunogenicity of different regions of CSP may be different between humans and mice, it is notable that the only successful 17

398 399 400 401 402 403 404 405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 CSP-based malaria vaccine, RTS,S, also lacks the N-terminal region. Thus these observations in mice could have implications for improving CSP vaccine design (18). Although the C-terminal region of P. berghei CSP has been associated with protection in mice (19, 20), our C-terminal construct of PfCSP did not confer protection against a Tr-Pb challenge. This result was reminiscent of a human trial in which a repeatless CSP construct did not protect the vaccinees (21). Doud et al. have recently shown that key epitopes on the C-terminal region involve non-contiguous residues of the molecule (22). It is possible that the isolated C-terminal protein used for immunization in our study did not adopt the same conformation as the C-terminal region in the intact CSP molecule. Additionally, it is noteworthy that several mice with low repeat-specific antibody titers were protected in the present study, as is often observed with RTS,S immunized humans. Although cellular immune responses to CSP were not measured, T-cell epitopes in the N- and C-terminal regions of CSP could have contributed to protection (6, 19), as has been observed by others using CSP transgenic parasite models (11, 23). In conclusion, we report sterile protection by recombinant PfCSP vaccines against the full-length PfCSP transgenic mouse parasite challenge. The protection was dependent on antigen dose and the nature of the CSP immunogen. In support of our observations, it is noteworthy that during the course of our study, Kaba et al. also reported using the same Tr-Pb parasite line to monitor protection induced in mice following immunization with a nanoparticle vaccine 18

421 422 423 424 425 426 427 428 429 430 431 expressing B and CD8 T-cell epitopes from the PfCSP (23). Although the biological relevance of this mouse protection model, vis-à-vis human vaccine development, remains to be confirmed, sterile protection as the efficacy endpoint can provide a means to rapidly evaluate PfCSP-based vaccines for future human trials. Acknowledgements The funding for this work was provided by the USAID Malaria Vaccine Development Program. We thank Dr. Lorraine Soisson and Dr. Carter Diggs for their support and advice. Downloaded from http://cvi.asm.org/ on September 24, 2018 by guest 19

432 433 Table 1: 434 435 Expt. I II III IV Mean Spz. Dose Route n Infected Day of patency 5000 SQ 7 1 8 7500 SQ 7 1 5 10000 SQ 7 5 6.3 2500 IV 7 7 5.5 2500 IV 7 7 5 1000 IV 7 7 5 2500 IV 10 10 5.2 1000 IV 10 9 5.4 Downloaded from http://cvi.asm.org/ on September 24, 2018 by guest 20

436 References 437 438 439 440 441 442 443 444 445 446 447 448 449 450 451 452 453 454 455 456 457 458 459 460 461 462 463 464 465 466 467 468 469 470 471 472 473 474 475 476 477 1. Jin Y, Kebaier C, Vanderberg J. 2007. Direct microscopic quantification of dynamics of Plasmodium berghei sporozoite transmission from mosquitoes to mice. Infection and immunity 75:5532-5539. 2. Meis JF, Verhave JP. 1988. Exoerythrocytic development of malarial parasites. Advances in parasitology 27:1-61. 3. Dame JB, Williams JL, McCutchan TF, Weber JL, Wirtz RA, Hockmeyer WT, Maloy WL, Haynes JD, Schneider I, Roberts D, Sanders GS, Reddy P, Diggs CL, Miller LH. 1984. Structure of the gene encoding the immunodominant surface antigen on the sporozoite of the human malaria parasite Plasmodium falciparum. Science 225:593-599. 4. Tewari R, Spaccapelo R, Bistoni F, Holder AA, Crisanti A. 2002. Function of region I and II adhesive motifs of Plasmodium falciparum circumsporozoite protein in sporozoite motility and infectivity. The Journal of biological chemistry 277:47613-47618. 5. Vekemans J, Leach A, Cohen J. 2009. Development of the RTS,S/AS malaria candidate vaccine. Vaccine 27 Suppl 6:G67-71. 6. Bongfen SE, Ntsama PM, Offner S, Smith T, Felger I, Tanner M, Alonso P, Nebie I, Romero JF, Silvie O, Torgler R, Corradin G. 2009. The N-terminal domain of Plasmodium falciparum circumsporozoite protein represents a target of protective immunity. Vaccine 27:328-335. 7. Collins WE, Sullivan JS, Williams A, Galland GG, Nace D, Williams T, Barnwell JW. 2009. The Santa Lucia strain of Plasmodium falciparum in Aotus monkeys. The American journal of tropical medicine and hygiene 80:536-540. 8. Hayes DE, Ward RA. 1977. Sporozoite transmission of falciparum malaria (Burma-Thau. strain) from man to Aotus monkey. The American journal of tropical medicine and hygiene 26:184-185. 9. Collins WE, Sullivan JS, Morris CL, Galland GG, Richardson BB, Roberts JM. 1997. The Malayan IV strain of Plasmodium falciparum in Aotus monkeys. The American journal of tropical medicine and hygiene 56:49-56. 10. Persson C, Oliveira GA, Sultan AA, Bhanot P, Nussenzweig V, Nardin E. 2002. Cutting edge: a new tool to evaluate human pre-erythrocytic malaria vaccines: rodent parasites bearing a hybrid Plasmodium falciparum circumsporozoite protein. Journal of immunology 169:6681-6685. 11. Kastenmuller K, Espinosa DA, Trager L, Stoyanov C, Salazar AM, Pokalwar S, Singh S, Dutta S, Ockenhouse CF, Zavala F, Seder RA. 2012. Full-length P. Falciparum Circumsporozoite Protein Administered with Poly-ICLC or GLA/SE Elicits Potent Antibody and CD4+ T Cell Immunity and Protection in Mice. Infection and immunity. 21

478 479 480 481 482 483 484 485 486 487 488 489 490 491 492 493 494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512 513 514 515 516 517 518 519 520 12. Ozaki LS, Gwadz RW, Godson GN. 1984. Simple centrifugation method for rapid separation of sporozoites from mosquitoes. The Journal of parasitology 70:831-833. 13. Hollingdale MR, Nardin EH, Tharavanij S, Schwartz AL, Nussenzweig RS. 1984. Inhibition of entry of Plasmodium falciparum and P. vivax sporozoites into cultured cells; an in vitro assay of protective antibodies. Journal of immunology 132:909-913. 14. Schwenk RJ, Richie TL. 2011. Protective immunity to pre-erythrocytic stage malaria. Trends in parasitology 27:306-314. 15. Myung JM, Marshall P, Sinnis P. 2004. The Plasmodium circumsporozoite protein is involved in mosquito salivary gland invasion by sporozoites. Molecular and biochemical parasitology 133:53-59. 16. Aldrich C, Magini A, Emiliani C, Dottorini T, Bistoni F, Crisanti A, Spaccapelo R. 2012. Roles of the amino terminal region and repeat region of the Plasmodium berghei circumsporozoite protein in parasite infectivity. PloS one 7:e32524. 17. Coppi A, Pinzon-Ortiz C, Hutter C, Sinnis P. 2005. The Plasmodium circumsporozoite protein is proteolytically processed during cell invasion. The Journal of experimental medicine 201:27-33. 18. Kester KE, Cummings JF, Ofori-Anyinam O, Ockenhouse CF, Krzych U, Moris P, Schwenk R, Nielsen RA, Debebe Z, Pinelis E, Juompan L, Williams J, Dowler M, Stewart VA, Wirtz RA, Dubois MC, Lievens M, Cohen J, Ballou WR, Heppner DG, Jr., Rts SVEG. 2009. Randomized, double-blind, phase 2a trial of falciparum malaria vaccines RTS,S/AS01B and RTS,S/AS02A in malaria-naive adults: safety, efficacy, and immunologic associates of protection. The Journal of infectious diseases 200:337-346. 19. Roggero MA, Meraldi V, Lopez JA, Eberl G, Romero JC, Matile H, Betschart B, Corradin G, Renggli J. 2000. The synthetic, oxidized C- terminal fragment of the Plasmodium berghei circumsporozoite protein elicits a high protective response. European journal of immunology 30:2679-2685. 20. Bergmann-Leitner ES, Scheiblhofer S, Weiss R, Duncan EH, Leitner WW, Chen D, Angov E, Khan F, Williams JL, Winter DB, Thalhamer J, Lyon JA, Tsokos GC. 2005. C3d binding to the circumsporozoite protein carboxyterminus deviates immunity against malaria. International immunology 17:245-255. 21. Heppner DG, Gordon DM, Gross M, Wellde B, Leitner W, Krzych U, Schneider I, Wirtz RA, Richards RL, Trofa A, Hall T, Sadoff JC, Boerger P, Alving CR, Sylvester DR, Porter TG, Ballou WR. 1996. Safety, immunogenicity, and efficacy of Plasmodium falciparum repeatless circumsporozoite protein vaccine encapsulated in liposomes. The Journal of infectious diseases 174:361-366. 22

521 522 523 524 525 526 527 528 529 530 531 532 22. Doud MB, Koksal AC, Mi LZ, Song G, Lu C, Springer TA. 2012. Unexpected fold in the circumsporozoite protein target of malaria vaccines. Proceedings of the National Academy of Sciences of the United States of America 109:7817-7822. 23. Kaba SA, McCoy ME, Doll TA, Brando C, Guo Q, Dasgupta D, Yang Y, Mittelholzer C, Spaccapelo R, Crisanti A, Burkhard P, Lanar DE. 2012. Protective Antibody and CD8(+) T-Cell Responses to the Plasmodium falciparum Circumsporozoite Protein Induced by a Nanoparticle Vaccine. PloS one 7:e48304. Downloaded from http://cvi.asm.org/ on September 24, 2018 by guest 23

533 Figure Legends 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 Table 1: Optimization of the challenge dose in C57BL/6 mice. Sporozoite dose required for 100% infectivity in mice was established by subcutaneous (SQ) or intravenous (IV) injection of sporozoites. Shown are the number of mice challenged (n), the number of infected mice as determined by patent parasitemia in the blood, and mean day of patency for each group. Fig. 1: Tr-Pb optimization in the mosquito-mouse model. (A) Passage cycles of transgenic P. berghei parasite (Tr-Pb) from mouse to mosquito. Data from 10 mosquito-mouse cycles, each consisting of 4 blood passages, were collected. (B) Percentage of mosquitoes infected by oocysts and sporozoites of Tr-Pb (clear bars) or wild type (black) parasites. Mean (+s.e.m) and Mann-Whitney p-values are shown. (C) Oocyst and sporozoite ratings (mean+s.e.m). Oocyst ratings were defined as: 0 = 0 oocysts per midgut; 1 = 1-25; 2 = 26-50; 3 = 51-100; and 4 = 100+ oocysts. Sporozoite load ratings were defined as: 0 = 0 sporozoites per mosquito; 1 = 1-100; 2 = 100-1000; 3 = 1000-10,000; and 4 = 10,000+ sporozoites. (D) Sporozoite yield per mosquito using mouse-mouse blood transmission or mouse-mosquito (vector-borne) transmission. (E) Reactivity of anti-p. berghei CSP mab 4B10 and anti-p. falciparum CSP mab 49-1B2 against Tr-Pb and WT P. berghei sporozoites. 24

555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576 577 Fig. 2: Protection data on various PfCSP constructs. (A) Four constructs were expressed in E. coli, two of which included the N- and C- terminal region and either 19 or 5 of the 38 NANP repeats of 3D7 P. falciparum CSP. A third construct contained 18 NANP repeats and the C- terminal region and a fourth construct (Cterm) contained only the C- terminal region. (B) Coomassie blue-stained gel of purified proteins on a non-reduced SDS-PAGE. Lane 1, N-(NANP) 19 -C; lane 2, (NANP) 18 -C; lane 3, N-(NANP) 5 -C; lane 4, Cterm CS protein. (C) Mean ELISA endpoint titers measured 2 wk after each vaccination against the repeat and near full-length construct. (D) Titers of individual mice in each group measured against the (NANP) 6 peptide, the near full-length protein, or the Cterm protein coated on plates. Geometric mean titers (±95%CI) are shown. Mice protected against Tr-Pb challenge (open triangles) and nonprotected (closed circles) are shown. Fig. 3: Protection data with incremental doses of PfCSP. (A) Endpoint titers of mice vaccinated with increasing doses of N-(NANP) 19 -C protein measured at 2 wk post third dose against the (NANP) 6 repeat peptide, near full-length N-(NANP) 19 -C protein, or the Cterm protein coated on ELISA plates. Geometric means (±95% CI) are shown. Protected mice are shown as open triangles. (B) Endpoint titers of mice from the construct and dosetitration experiments at 2 wk post third vaccination. Full-length CSP ELISA titers were plotted against the repeat peptide ELISA titers. 25

578 579 580 581 582 583 584 585 586 587 588 589 590 591 592 593 594 595 596 Protected mice ( P, red open triangles or diamonds) and non-protected mice ( NP, closed symbols) are indicated. Inset shows the percentage of mice protected in each quadrant. (C) Near full-length ELISA titer of N- (NANP) 19 -C immunized mice, from the construct and dose titration study, plotted against their respective avidity indicies. Fig. 4: Biological activity of mouse antibodies against N-(NANP) 19 -C construct. (A) Recognition of CSP on the surface of fixed sporozoites; shown is a typical phase-contrast and fluorescence image. (B) IFA titers are from a single experiment using pooled sera from N-(NANP) 19 -C vaccinated dose-titration mice. (C) Western blot reactivity at a 1:1000 serum dilution against salivary gland preparation of P. falciparum sporozoite infected (inf) or uninfected (un) mosquitoes. (D) Dose response assay showing inhibition of sporozoite invasion using sera from N-(NANP) 19 -C vaccinated mice. (E) Pooled sera at a 1:100 dilution from the N-(NANP) 19 -C dose titration experiment tested in an inhibition of sporozoite invasion assay. Shown are the mean (+s.e.m) of three replicate wells. 26