Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
VACCINE COMPRISING AMA1 AND RON2
Document Type and Number:
WIPO Patent Application WO/2015/002959
Kind Code:
A1
Abstract:
Disclosed is a vaccine comprising an immunogenic composition comprising a complex of AMA1 and RON2 (or a fragment thereof), which elicits an immune response to a Plasmodium species in a subject upon administration. The resulting immune response is sufficient to impede or prevent infection by a Plasmodium species.

Inventors:
SRINIVASAN PRAKASH (US)
MILLER LOUIS HOWARD (US)
Application Number:
PCT/US2014/045065
Publication Date:
January 08, 2015
Filing Date:
July 01, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
US HEALTH (US)
International Classes:
A61K39/015; C07K14/445
Domestic Patent References:
WO2002077195A22002-10-03
Foreign References:
EP2520585A12012-11-07
Other References:
MAULD LAMARQUE ET AL: "The RON2-AMA1 Interaction is a Critical Step in Moving Junction-Dependent Invasion by Apicomplexan Parasites", PLOS PATHOGENS, vol. 7, no. 2, 10 February 2011 (2011-02-10), pages e1001276 - e1001276, XP055006947, ISSN: 1553-7366, DOI: 10.1371/journal.ppat.1001276
M. L. TONKIN ET AL: "Host Cell Invasion by Apicomplexan Parasites: Insights from the Co-Structure of AMA1 with a RON2 Peptide", SCIENCE, vol. 333, no. 6041, 22 July 2011 (2011-07-22), pages 463 - 467, XP055006946, ISSN: 0036-8075, DOI: 10.1126/science.1204988
P. SRINIVASAN ET AL: "Immunization with a functional protein complex required for erythrocyte invasion protects against lethal malaria", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 111, no. 28, 15 July 2014 (2014-07-15), pages 10311 - 10316, XP055145125, ISSN: 0027-8424, DOI: 10.1073/pnas.1409928111
PRAKASH SRINIVASAN ET AL: "Disrupting malaria parasite AMA1-RON2 interaction with a small molecule prevents erythrocyte invasion", NATURE COMMUNICATIONS, vol. 4, 2 August 2013 (2013-08-02), XP055145129, DOI: 10.1038/ncomms3261
P. SRINIVASAN ET AL: "Binding of Plasmodium merozoite proteins RON2 and AMA1 triggers commitment to invasion", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 108, no. 32, 25 July 2011 (2011-07-25), pages 13275 - 13280, XP055145950, ISSN: 0027-8424, DOI: 10.1073/pnas.1110303108
CAO J ET AL: "Rhoptry neck protein RON2 forms a complex with microneme protein AMA1 in Plasmodium falciparum merozoites", PARASITOLOGY INTERNATIONAL, ELSEVIER SCIENCE, AMSTERDAM, NL, vol. 58, no. 1, 1 March 2009 (2009-03-01), pages 29 - 35, XP025913923, ISSN: 1383-5769, [retrieved on 20090205], DOI: 10.1016/J.PARINT.2008.09.005
TYLER JESSICA S ET AL: "The C-Terminus of Toxoplasma RON2 Provides the Crucial Link between AMA1 and the Host-Associated Invasion Complex", PLOS PATHOGENS, vol. 7, no. 2, February 2011 (2011-02-01), XP055006903
MAHAMADOU A. THERA ET AL: "A Field Trial to Assess a Blood-Stage Malaria Vaccine", NEW ENGLAND JOURNAL OF MEDICINE, vol. 365, no. 11, 15 September 2011 (2011-09-15), pages 1004 - 1013, XP055145136, ISSN: 0028-4793, DOI: 10.1056/NEJMoa1008115
Attorney, Agent or Firm:
CONNELL, Gary, J. et al. (1560 BroadwaySuite 120, Denver CO, US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. A vaccine comprising: i) an immunogenic composition comprising a complex of AMAl and RON2; and ii) at least one adjuvant and/or at least one physiologically acceptable carrier.

2. A vaccine comprising: i) an immunogenic composition comprising a complex of: a) AMAl; and b) RON2 or a fragment thereof; and ii) at least one adjuvant and/or at least one physiologically acceptable carrier.

3. The vaccine of claim 1 or 2, wherein the composition elicits an immune response to a Plasmodium species in a subject upon administration to the subject.

4. The vaccine of claim 4, wherein the immune response is sufficient to impede or prevent infection by a Plasmodium species.

5. The vaccine of claim 1 or 2, wherein the vaccine is for the treatment of or protection from erythrocytic and/or pre-erythrocytic malaria infection in a subject.

6. The vaccine of claim 5, wherein the treatment manifests itself in the subject as the parasitemia being under control and/or the infection being cleared.

7. The vaccine of claim 3 or 4, wherein the Plasmodium species is selected from the group consisting of Plasmodium falciparum, Plasmodium knowlesi, Plasmodium vivax, Plasmodium yoelii, Plasmodium malariae, Plasmodium ovale, Plasmodium brasilianum, Plasmodium cynomulgi, Plasmodium inui, Plasmodium rhodiani, Plasmodium schwetzi, Plasmodium semiovale, and Plasmodium simium.

8. A method of protecting a subject from erythrocytic malaria infection comprising administering the vaccine of claim 1 or 2 to the subject in an amount effective to stimulate an immune response, thus protecting the subject from erythrocytic malaria infection.

9. A method of protecting a subject from pre-erythrocytic malaria infection comprising administering the vaccine of claim 1 or 2 to the subject in an amount effective to stimulate an immune response, thus protecting the subject from pre-erythrocytic malaria infection.

10. A method of protecting a subject from both erythrocytic and pre-erythrocytic malaria infection comprising administering the vaccine of claim 1 or 2 to the subject in an amount effective to stimulate an immune response, thus protecting the subject from both pre-erythrocytic and erythrocytic malaria infection.

11. A method of protecting a subject from infection by a Plasmodium species comprising administering the vaccine of claim 1 or 2 to the subject in an amount effective to stimulate an immune response, thus protecting the subject from infection with the Plasmodium species.

12. The method of any one of claims 8-11, wherein the vaccine is administered orally or parenterally.

13. The method of any one of claims 8-11, wherein the vaccine is administered with another active agent.

14. The method of claim 13, wherein the agent is an antibiotic, antigen, or antibody.

15. A method for vaccinating a subject against a Plasmodium species comprising administering to the subject an effective amount of the vaccine of claim 1 or 2.

16. A method for generating protective antibodies in a subject against a Plasmodium species comprising administering to the subject an effective amount of the vaccine of claim 1 or 2.

17. A method for producing an immune response against a Plasmodium species in a subject comprising administering the vaccine of claim 1 or 2 to the subject in an amount effective to produce an immune response against the Plasmodium species.

18. The method of claim 11, 15, 16, or 17, wherein the Plasmodium species is selected from the group consisting of Plasmodium falciparum, Plasmodium knowlesi, Plasmodium vivax, Plasmodium yoelii, Plasmodium malariae, Plasmodium ovale, Plasmodium brasilianum, Plasmodium cynomulgi, Plasmodium inui, Plasmodium rhodiani, Plasmodium schwetzi, Plasmodium semiovale, and Plasmodium simium.

19. An article of manufacture comprising a closed, pathogen-impermeable container and a sterile vaccine preparation enclosed within said container, wherein said vaccine preparation comprises the vaccine of claim 1 or 2.

Description:
VACCINE COMPRISING AMA1 AND RON2

RELATED APPLICATION DATA

[0001] Each of the applications and patents cited in this text, as well as each document or reference cited in each of the applications and patents (including during the prosecution of each issued patent; "application cited documents"), and each of the PCT and foreign applications or patents corresponding to and/or claiming priority from any of these applications and patents, and each of the documents cited or referenced in each of the application cited documents, are hereby expressly incorporated herein by reference and may be employed in the practice of the invention. More generally, documents or references are cited in this text, either in a Reference List before the claims, or in the text itself; and, each of these documents or references ("herein cited references"), as well as each document or reference cited in each of the herein cited references (including any manufacturer's specifications, instructions, etc.), is hereby expressly incorporated herein by reference.

STATEMENT OF GOVERNMENT INTEREST

[0002] This invention was made with government support; the government has certain rights in the invention.

SEQUENCE LISTING

[0003] Incorporated by reference herein in its entirety is the Sequence Listing entitled

"Sequence_Listing_ST25.txt", created June 30, 2014, size of 107 kilobytes.

BACKGROUND

[0004] Plasmodium is the causative agent of malaria, one of the world's deadliest parasitic diseases. Malaria affects over 300 million people worldwide, causing more than 1 million deaths annually, particularly in young children and pregnant women in sub-Saharan Africa. Currently, there is no vaccine available, and there is widespread resistance to common anti-malarial drugs. RTS,S, a leading vaccine candidate which targets the initial infection of the liver, achieved only partial efficacy (Olotu, et al. 2013 N Engl J Med

368(12): 1111-1120). However, clinical manifestations of the disease are caused by the blood- stage parasites. These disease-causing forms of the parasite primarily reside within the red blood cells (RBC), which, upon maturation (schizont), releases merozoites, the invasive form of Plasmodium that invades new RBCs. Therefore, vaccines targeting the erythrocytic forms of the parasite are desirable for efficient disease control measures.

[0005] Apicomplexan parasites have specialized secretory organelles (rhoptries and micronemes) that release their contents during host cell invasion. The micronemes possess the protein apical membrane antigen 1 (AMA1). AMA1 is an essential merozoite surface protein and was previously considered one of the leading blood-stage vaccine candidates (Stowers, et al. 2002 Infect Immun 70(12):6961-6967 and Dutta, et al. (2009) PLoS One 4(12):e8138). Despite the vaccines' ability to elicit high-titred AMA1- specific antibodies, Phase 2 clinical trials showed only weak efficacy

(http://www.ncbi.nlm.nih.gov/pubmed/21916638, Mahamadou, et al. 2011 New England J Med 365(11): 1004-13), even against homologous parasite (Spring, et al. 2009 PLoS One 4(4):e5254, Ouattara, et al. 2010 Malar J 9:175 and Thera, et al. 2011 N Engl J Med

365(11): 1004-1013). Recent efforts to cover the polymorphism in AMA1 demonstrated that combining 4-5 different AMA1 alleles could overcome the strain- specific barrier in vitro (Miura, et al. 2013 Infect Immun 81(5): 1491-1501, Dutta, et al. 2013 PLoS Pathog

9(12):el003840 and Remarque, et al. 2008 Infect Immun 76(6):2660-2670).

[0006] The rhoptry neck sub-compartment possesses a distinct protein repertoire, including rhoptry neck protein (RON2).

SUMMARY

[0007] The discordance between failure to protect humans in vivo and ability to block vaccine-type parasite invasion in vitro (Spring, et al. 2009 PLoS One 4(4):e5254) underscores the need to improve AMA1 vaccine efficacy against homologous parasites. Interaction between AMA1 and another parasite protein RON2 is shown to be essential for successful invasion of RBCs (Lamarque, et al. 2011 PLoS Pathog 7(2):el001276). A small 49-amino acid peptide near the C-terminal of the RON2 protein is sufficient to bind a hydrophobic pocket in AMA1. Small molecules or peptides that block this interaction inhibit merozoite invasion (Srinivasan, et al. 2013 Nat Commun 4:2261 and Richard, et al. 2010 J Biol Chem 285(19): 14815-14822), highlighting the important role of this protein-protein interaction. Crystal structure of the complex revealed that the RON2 peptide (RON2L) binds to a conserved hydrophobic groove in AMA1 resulting in extensive conformational changes in certain loop regions surrounding the groove (Vulliez-Le Normand, et al. 2012 PLoS Pathog 8(6):el002755 and Tonkin, et al. 2011 Science 333(6041):463-467). Antibodies that bind in or near the hydrophobic groove block parasite invasion by inhibiting the binding of RON2 (Dutta, et al. 2013 PLoS Pathog 9(12):el003840 and Srinivasan, et al. 2011 Proc Natl Acad Sci U SA 108(32): 13275-13280). It is further shown that a very high concentration of antibodies against the RON2 peptide is required to inhibit merozoite invasion (Srinivasan, et al. 2011 PNAS 10: 1073). Simply adding anti-AMAl and anti-RON2 peptide antibodies together does not improve inhibition of invasion.

[0008] Described herein is a novel approach based on vaccination with an AMA1-

RON2 complex that provides complete protection against lethal Plasmodium challenge in an animal model. A highly virulent P. yoelli YM (P_ YM) mouse model was used. In marked contrast, animals immunized with the two antigens separately are not protected. Indeed, immunization with a functional complex of RON2 peptide (RON2L) induces antibody- mediated, complete protection against lethal P. yoelli challenge. The data shows that protection of animals is based on antibodies generated against the complex. Significantly, IgG from mice immunized with the complex transferred protection. Furthermore, IgG from P AMA1-RON2 immunized animals have enhanced invasion inhibition compared to IgG elicited by AMAl alone, as passive transfer of IgG but not T cells from AMA1-RON2L vaccinated animals controlled parasitemia.

[0009] Protection may be mediated by antibodies recognizing new inhibitory epitopes of AMAl and/or the AMA1/RON2 peptide complex exposed by the binding of RON2 to AMAl. Interestingly, the qualitative increase in efficacy appears to be, in part, due to a switch in the proportion of antibodies targeting the RON2 binding site in AMAl. This indicates that that the complex functions as an antigen exposing unique, inhibitory epitopes that may be distinctive from the two antigens by themselves. Because AMAl and RON2 are critical for the Plasmodium parasite's invasion of the host cell, targeting them by raising a host immune response to a complex of the two proteins should block infection. These results suggest that a multi-allele AMAl (to overcome polymorphisms) in complex with RON2L should be effective in protecting against all P. falciparum parasites.

[0010] It is demonstrated herein that the human parasite P 3D7 AM A 1 -RON2L complex induces qualitatively higher growth inhibitory antibodies than AMAl alone in in vitro assays. Surprisingly, the results indicate that the increase in inhibitory antibodies generated by the complex may, in part, be due to a switch in the proportion of antibodies against the loops surrounding the hydrophobic groove with which RON2 interacts. The data suggest that a vaccine comprising a multi-allele AMA1 in complex with RON2L may be more efficacious compared to AMA1 alone in targeting both homologous and heterologous parasites.

[0011] The invention encompasses treatment and/or prophylaxis of Plasmodium infection/malaria (used interchangeably herein) by administration of vaccines disclosed herein. In some embodiments, such vaccines are administered to a subject suffering from malaria or susceptible to Plasmodium infection. In some embodiments, a subject is considered to be suffering from malaria, if the subject is displaying one or more symptoms commonly associated with therewith. In some embodiments, the subject is known or believed to have been exposed to at least one Plasmodium species. In some embodiments, a subject is considered to be susceptible to Plasmodium infection, if the subject is known or believed to have been exposed to a Plasmodium species. In some embodiments, a subject is known or believed to have been exposed to a Plasmodium species, if the subject has been in contact with other individuals known or suspected to have been infected with the same, and/or if the subject is or has been present in a location in which malaria is known or thought to be prevalent. Vaccines disclosed herein may be administered prior to or after development of one or more symptoms of Plasmodium infection/malaria.

[0012] Thus, in one aspect, the invention provides a vaccine comprising: i) an immunogenic composition comprising a complex of AMA1 and RON2; and ii) at least one adjuvant and/or at least one physiologically acceptable carrier.

[0013] In another aspect, the invention provides a vaccine comprising: i) an immunogenic composition comprising a complex of: a) AMA1 or a fragment thereof; and b) RON2 or a fragment thereof; and ii) at least one adjuvant and/or at least one physiologically acceptable carrier. The full-length RON2 protein is 2189 amino acids (aa). Identified herein is one 49 amino acid region sufficient to bind AMA1 and elicit a protective immune response. A shorter fragment within that 49 aa region or another fragment of the full-length protein may likewise exhibit AMA1 binding capability and elicit a protective immune response. Thus, full-length RON2, RON2 peptide, peptide derived from RON2, RON2L, and a fragment of RON2 (the latter four "terms/phrases" being used interchangeably) are contemplated in the vaccines, methods, and articles of manufacture disclosed herein.

[0014] In still another aspect, the invention provides a vaccine comprising: i) an immunogenic composition comprising a fusion protein complex of AMA1 and RON2 (or a fragment thereof); and ii) at least one adjuvant and/or at least one physiologically acceptable carrier. In the instant aspect, it would not be necessary to prepare the individual components and then mix them to form a complex. Rather, a fusion protein is created by joining the genes coding for the separate proteins. Translation of the fusion gene then results in a single polypeptide.

[0015] In one embodiment of a vaccine according to the invention, the immunogenic composition elicits an immune response to a Plasmodium species in a subject upon administration to the subject. In another embodiment, the immune response is sufficient to impede or prevent infection by a Plasmodium species.

[0016] In another embodiment of a vaccine according to the invention, the vaccine is for the treatment of or protection from erythrocytic (blood stage) and/or pre-erythrocytic (liver stage) malaria infection in a subject. In another embodiment of a vaccine according to the invention, the vaccine is for the treatment of or protection from erythrocytic malaria infection in a subject. In another embodiment of a vaccine according to the invention, the vaccine is for the treatment of or protection from pre-erythrocytic malaria infection in a subject. In still another embodiment, the treatment manifests itself in the subject as the parasitemia being under control and/or the infection being cleared.

[0017] In one embodiment of a vaccine according to the invention, the Plasmodium species is selected from the group consisting of Plasmodium falciparum, Plasmodium knowlesi, Plasmodium vivax, Plasmodium yoelii, Plasmodium malariae, Plasmodium ovale, Plasmodium brasilianum, Plasmodium cynomulgi, Plasmodium inui, Plasmodium rhodiani, Plasmodium schwetzi, Plasmodium semiovale, and Plasmodium simium.

[0018] In another embodiment of a vaccine according to the invention, the vaccine further comprises an active agent. The active agent may, without limitation, be an antibiotic, and antigen, or an antibody.

[0019] In another aspect, the invention provides a method of protecting a subject from erythrocytic (blood stage) malaria infection comprising administering a vaccine as disclosed herein to the subject in an amount effective to stimulate an immune response, thus protecting the subject from erythrocytic (blood stage) malaria infection.

[0020] In another aspect, the invention provides a method of protecting a subject from pre-erythrocytic (liver stage) malaria infection comprising administering a vaccine as disclosed herein to the subject in an amount effective to stimulate an immune response, thus protecting the subject from pre-erythrocytic (liver stage) malaria infection.

[0021] In another aspect, the invention provides a method of protecting a subject from both erythrocytic and pre-erythrocytic malaria infection comprising administering a vaccine as disclosed herein to the subject in an amount effective to stimulate an immune response, thus protecting the subject from both pre-erythrocytic and erythrocytic malaria infection. The degree of protection from each may vary.

[0022] In another aspect, the invention provides a method of protecting a subject from infection by a Plasmodium species comprising administering a vaccine as disclosed herein to the subject in an amount effective to stimulate an immune response, thus protecting the subject from infection with the Plasmodium species.

[0023] In one embodiment of a method according to the invention, the vaccine is administered orally or parenterally.

[0024] In another embodiment of a method according to the invention, the vaccine is administered with another active agent. In yet another embodiment, the active agent is an antibiotic, antigen, or antibody.

[0025] In another aspect, the invention provides a method for vaccinating a subject against a Plasmodium species comprising administering to the subject an effective amount of a vaccine as disclosed herein.

[0026] In yet another aspect, the invention provides a method for generating protective antibodies in a subject against a Plasmodium species comprising administering to the subject an effective amount of a vaccine as disclosed herein.

[0027] In still another aspect, the invention provides a method for producing an immune response against a Plasmodium species in a subject comprising administering a vaccine as disclosed herein to the subject in an amount effective to produce an immune response against the Plasmodium species.

[0028] In one embodiment of a method according to the invention, the Plasmodium species is selected from the group consisting of Plasmodium falciparum, Plasmodium knowlesi, Plasmodium vivax, Plasmodium yoelii, Plasmodium malariae, Plasmodium ovale, Plasmodium brasilianum, Plasmodium cynomulgi, Plasmodium inui, Plasmodium rhodiani, Plasmodium schwetzi, Plasmodium semiovale, and Plasmodium simium.

[0029] In one aspect, the invention provides an article of manufacture comprising a closed, pathogen-impermeable container and a sterile vaccine preparation enclosed within said container, wherein said vaccine preparation comprises a vaccine as disclosed herein.

[0030] Other aspects of the invention are described in or are obvious from the following disclosure and are within the ambit of the invention. BRIEF DESCRIPTION OF THE DRAWINGS

[0031] The following Detailed Description, given by way of Examples, but not intended to limit the invention to specific embodiments described, may be understood in conjunction with the accompanying figures, in which:

[0032] Figure 1 depicts a graph illustrating the potential of the AMA1-RON2 peptide complex in protecting against a lethal parasite challenge. Mice immunized with buffer (Ctrl), the individual antigens (P_yAMAl, P_yR2L, respectively), or when mice are immunized with the two antigens separately (not as complex), the animals are not able to control parasitemia (% parasitemia over time increases). On the other hand, mice immunized with AMA1-RON2 complex (P_yAMAl/P_yR2L) are able to control parasitemia and clear the infection.

[0033] Figure 2 depicts a graph illustrating the potential of the AMA1-RON2 peptide complex in protecting against a lethal parasite challenge. Mice immunized with buffer (Ctrl), the individual antigens (P_ AMAl, P_ R2L, respectively), or when mice are immunized with the two antigens separately (not as complex), the animals succumb to the disease (% survival over time decreases). On the other hand, mice immunized with AMA1-RON2 complex (P_ AMAl/P_ R2L) survive the disease.

[0034] Figure 3 (i.e., Figures 3A-3I, as follows) demonstrates that immunization with

AMA1-RON2L complex, but not AMAl alone, protects mice against lethal P. yoelii YM challenge. (Figure 3 A) Five mice per group were immunized with AMAl, RON2L-KLH or AMA1-RON2L complex and challenged with 10 4 infected RBCs (iRBCs) intravenously. Five mice immunized with buffer in Freund's adjuvant were used as controls. Error bars indicate mean + sem. (Figure 3B) Kaplan-Meir curve of the overall survival of animals in Fig. 3 A. (Figure 3C) ELISA titers of antibody response against AMAl and RON2L from sera of mice from Fig. 3a. Sera were used at 1:8000 and 1:2000 dilutions for AMAl and RON2L, respectively. Error bars indicate mean + sem at O.D405. (Figure 3D) In silico homology model of P_ AMAl-P_ RON2L complex based on P AMA1-P RON2 complex structure. Arrows indicate the two cysteines in the PyRON2L peptide (Figure 3E) Mutation of Cysl856 and 1868 to Ala abolishes RON2L binding to P AMAl. (Figure 3F) Mutating the two cysteines to alanines (c/a) in the RON2L peptide required for binding to AMAl abrogates complex driven protection in mice. Five mice per group were challenged with 10 5 iRBCs. Error bars indicate mean + sem. (Figure 3G) Protection requires vaccination with the AMA1- RON2L complex as immunizing animals with the two antigens in separate sites (AMAl + RON2L) does not protect. Five mice per group for control and AMA1-RON2L and four mice for AMA1+RON2L were challenged with 10 5 iRBCs. Error bars indicate mean + sem.

(Figure 3H) Passive transfer of IgG from mice immunized with the AMA1-RON2L complex controls parasitemia. 400 μg of IgG from either control (PBS-Freund's adjuvant immunized mice) or AMA1-RON2L immunized mice were passively transferred on day -1, 0 and +1 and challenged on day 0 with 10 5 iRBCs using 5 mice per group. Error bars indicate mean + sem. (Figure 31) Kaplan-Meir curve of the overall survival of animals in Fig. 3H.

[0035] Figure 4 {i.e., Figures 4A-4E, as follows) demonstrates that Ρ/ΆΜΑ 1 -RON2L complex generates better quality P. falciparum invasion inhibitory antibodies than P/AMAl. (Figure 4A) IgG purified from rats immunized with P/3D7AMA1-RON2L complex induces higher growth inhibition compared to IgG from P/AMAl immunized rats (n=4). 2 mg/mL IgG was used in the inhibition assay and results are mean + sem of pooled data from two independent experiments. (Figure 4B) P/AMAl and P/AMA1-RON2L complex induces similar levels of anti-AMAl antibodies. ELISA units represent the AMAl -specific antibody titer in purified IgG (2 mg/mL) and serum from four immunized rats used in Fig. 4A. Error bars indicate mean + sem. (Figure 4C) GIA was measured using increasing concentrations of IgG from AMAl, RON2L and AMA1-RON2L groups. Data shown are the mean parasite inhibition from an assay performed in duplicate. The contribution of anti-RON2L antibody towards the increased GIA observed in the AMA1-RON2L group was analyzed by mixing 1 mg/mL each of anti-RON2L IgG and anti-AMAl IgG (AMAl + RON2L). Data shown are mean + sem for four rats. (Figure 4D) Inhibition of invasion is reversed by the addition of recombinant P/3D7 AMAl to IgG from P AMAl (blue) and P AMAl -RON2L (red) groups. Data shown are the mean + sem parasite inhibition from two independent experiments performed in duplicate. All four data points are plotted. (Figure 4E) Binding of biotinylated RON2L peptide to immobilized P/3D7 AMAl inhibited by serial dilution of IgG against P AMAl (blue) and P/AMAl -RON2L complex (red). The X-axis indicates the amount of total AMAl-specific EU present at each of the dilutions. EC 50 (50% inhibition of RON2L binding) was measured by plotting a nonlinear fit of the individual data points (*, p=0.018).

[0036] Figure 5 {i.e., Figures 5A-5D, as follows) demonstrates the proportion of anti-

AMAl and anti-AMAl -RON2L complex IgG to loop regions surrounding the AMAl hydrophobic groove. (Figure 5A) View of the loops surrounding the hydrophobic groove in the absence of RON2L. (Figure 5B) View of the loops in the RON2L-bound form. (Figure 5C) Overlay of AMAl loop structures surrounding the hydrophobic groove in the presence (colored) and absence of RON2L (grey). (Figure 5D) Binding of IgG from AMAl (blue) and AMA1-RON2L complex (red) to biotinylated peptides immobilized on streptavidin plates. The X-axis indicates the amount of total AMA1- specific EU present at each of the dilutions. Data are mean + sem (n=4). A representative of 3 independent experiments is shown.

[0037] Figure 6 (i.e., Figures 6A-6C, as follows) demonstrates: (Figure 6A) Kaplan-

Meir curve of the overall survival of animals in Fig. 3F. (Figure 6B) Kaplan-Meir curve of the overall survival of animals in Fig. 3G. (Figure 6C) T cell transfer does not protect against P_yYM challenge. Kaplan-Meir curve of the overall survival of animals after passive transfer of 2 x 10 6 T cells from mice immunized with PBS-adjuvant (blue) or mice immunized with P_yAMAl-RON2L complex (red) on days -1, 0 and +1. Mice that received no cells were used as infection controls (black). Five mice per group were used and were challenged

intravenously withl0 5 P_yYM parasites on day 0.

[0038] Figure 7 demonstrates: anti-PfRON2L antibody titers induced by P/RON2L-

KLH and Ρ ΆΜΑ 1 -RON2L complex in rats. ELISA units represent the RON2L- specific antibody titer in either purified IgG (2 mg/mL) or serum from four immunized rats used in Fig. 4A. Error bars indicate mean + sem.

[0039] Figure 8 demonstrates: sequence alignment showing polymorphisms in the individual domain I loops surrounding the hydrophobic groove across multiple P. falciparum parasite strains. From the top of the alignment to the bottom, the SEQ ID NOs for the respective sequences are as follows: SEQ ID NO: 18, SEQ ID NO: 19, SEQ ID NO:20, SEQ ID NO:21, SEQ ID NO:22, SEQ ID NO:23, SEQ ID NO:24, SEQ ID NO:25, SEQ ID NO:26, and SEQ ID NO:27.

[0040] Figure 9 (i.e., Figures 9A-9D, as follows) demonstrates biochemical and biophysical characterization of recombinant EcPyAMAl. Analysis of purified recombinant EcPyAMAl by (Figure 9A) Coomassie blue stained SDS-PAGE gel and (Figure 9B) Western blot with a PyAMAl specific monoclonal antibody (mAb) 45B1 under non-reduced conditions. Molecular mass markers are shown alongside. (Figure 9C) Reversed-phase- HPLC analysis showing single peak along with the acetonitrile gradient elution. (Figure 9D) Analytical size-exclusion chromatography showing single monomeric peak.

[0041] Figure 10 shows: peptide sequences corresponding to P_yRON2L, P RON2L and P AMAl domain I and domain II loop regions used herein. Disulfide bridged cysteine residues in the peptides are underlined. All peptides were amidated (NH 2 ) at the C-terminus. From the top sequence to the bottom, the SEQ ID NOs for the respective sequences are as follows: SEQ ID NO:28, SEQ ID NO:29, SEQ ID NO:30, SEQ ID NO:31, SEQ ID NO:32, SEQ ID NO:33, SEQ ID NO:34, SEQ ID NO:35, SEQ ID NO:36, SEQ ID NO:37, SEQ ID NO:38, and SEQ ID NO:39. [0042] Figure 11 (i.e., Figures 11A-11F, as follows) demonstrates: non-human primate challenge using virulent human malaria parasite P. falciparum. Figures 11 A- 11C: parasitemia of control (Figure 11 A), AMAl (Figure 11B), and AMA1-RON2 (Figure 11C) - vaccinated animals. All control (n=6) and AMAl (n=8) became infected, but 4/8 animals immunized with AMA1-RON2 complex were sterile-protected, and an additional ¾ animals had a significantly delayed parasitemia. (Figure 1 ID) Time to patency after challenge. (Figure 1 IE) IgG purified from the vaccinated animals were tested in an in vitro growth inhibition assay (GIA). AM A 1-RON2L- induced IgG inhibited significantly higher than IgG from AMAl alone at 2.5 mg/mL. (Figure 1 IF) Correlation analysis of in vivo protection and in vitro inhibition. AMAl-RON2-immunized animals that were protected correlated well with increased GIA.

DETAILED DESCRIPTION

[0043] Reference will now be made in detail to representative embodiments of the invention. While the invention will be described in conjunction with the enumerated embodiments, it will be understood that the invention is not intended to be limited to those embodiments. On the contrary, the invention is intended to cover all alternatives, modifications, and equivalents that may be included within the scope of the present invention as defined by the claims.

[0044] One skilled in the art will recognize many methods and materials similar or equivalent to those described herein, which could be used in and are within the scope of the practice of the present invention. The present invention is in no way limited to the methods and materials described.

Definitions

[0045] Unless defined otherwise, technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods, devices, and materials similar or equivalent to those described herein can be used in the practice or testing of the invention, the preferred methods, devices, and materials are now described.

[0046] As used in this application, including the appended claims, the singular forms

"a," "an," and "the" include plural references, unless the content clearly dictates otherwise, and are used interchangeably with "at least one" and "one or more." Thus, reference to "a polynucleotide" includes a plurality of polynucleotides or genes, and the like. [0047] As used herein, the term "about" represents an insignificant modification or variation of the numerical value, such that the basic function of the item to which the numerical value relates is unchanged.

[0048] As used herein, the terms "comprises," "comprising," "includes," "including,"

"contains," "containing," and any variations thereof, are intended to cover a non-exclusive inclusion, such that a process, method, product-by-process, or composition of matter that comprises, includes, or contains an element or list of elements does not include only those elements but may include other elements not expressly listed or inherent to such process, method, product-by-process, or composition of matter.

[0049] The terms "subject", "patient", and "individual", as used herein,

interchangeably refer to a multicellular animal (including mammals (e.g. , humans, non- human primates, murines, ovines, bovines, ruminants, lagomorphs, porcines, caprines, equines, canines, felines, ayes, etc.), avians (e.g. , chicken), amphibians (e.g. Xenopus), reptiles, and insects (e.g. Drosophila). "Animal" includes guinea pig, hamster, ferret, chinchilla, mouse and cotton rat.

[0050] Reference herein to any numerical range (for example, a dosage range) expressly includes each numerical value (including fractional numbers and whole numbers) encompassed by that range. For example, reference herein to a range of "less than x"

(wherein x is a specific number) includes whole numbers x- 1, x-2, x-3, x-4, x-5, x-6, etc., and fractional numbers x-0.1, x-0.2, x-0.3, x-0.4, x-0.5, x-0.6, etc. In yet another illustration, reference herein to a range of from "x to y" (wherein x is a specific number, and y is a specific number) includes each whole number of x, x+1, x+2...to y-2, y-1, y, as well as each fractional number, such as x+0.1, x+0.2, x+0.3...to y-0.2, y-0.1. In another example, the term "at least 95%" includes each numerical value (including fractional numbers and whole numbers) from 95% to 100%, including, for example, 95%, 96%, 97%, 98%, 99% and 100%. - the latter will only be kept in and filled in with appropriate numbers if we use any greater than or less than numerical characterization...

[0051] The terms "antigen," "immunogen," "antigenic," "immunogenic,"

"antigenically active," "immunologic", and "immunologically active", as used herein, refer to any substance (including a molecule with one or more epitopes) that is capable of inducing a specific immune response (including eliciting a soluble antibody response) and/or cell- mediated immune response (including eliciting a cytotoxic T-lymphocyte (CTL) response), for example, to a malarial pathogen. The antigen can, in certain embodiments, be a complete protein, portions of proteins, peptides, fusion proteins, glycosylated proteins, and combinations thereof. In the context of the vaccines disclosed herein, one or more antigens (for example, AMA1/RON2 complex) may be provided directly or as part of a recombinant nucleic acid expression system to provide the antigenic complex to trigger an immune response in a subject. The antigen may also be a DNA molecule that produces the antigenic complex in the subject.

[0052] In general, "RON2", as used herein, refers to the protein, while "RON2L", as used herein, refers to the peptide sequence (49 amino acids). However, "AMA1/RON2" and "AMA1/RON2L" are used interchangeably herein when referring to the protein complex.

[0053] As used herein, the term "protein complex" or "complex" refers to the association of at least AMA1 and RON2. The term "protein complex" or "complex" includes a fusion protein complex. The proteins of the complex may be associated by a variety (including combination) of methods, including, without limitation, functional,

stereochemical, conformational, biochemical, or electrostatic association. In another embodiment, a multi-allele AMA1 (to overcome polymorphisms) in complex with RON2L is effective in protecting against both homologous and heterologous parasites.

[0054] An "immunogenic composition", as used herein, refers to a composition capable of eliciting an immune response to at least AMA1 or RON2 or AMA1/RON2 complex, when the composition is administered to a subject. The immune response elicited by such a composition comprised in a vaccine according to the invention affects the ability of a Plasmodium species to infect a subject immunized with the composition. Preferably, the ability of a Plasmodium species to infect an immunized subject is impeded or prevented.

[0055] The term "immune response", as used herein, refers to the development in a subject of a secretory and/or humoral and/or cellular immunological response to an antigen. "Humoral" immune response refers to the production of antibodies; "cellular" immune response refers to the activation of T-lymphocytes. An immune response elicited by an immunogenic composition comprised in a vaccine according to the invention may be a protective immune response. In such an embodiment, a protective response is generated in the vaccinated subject. Preferably, a protective immune response protects against subsequent infection by a Plasmodium species. The protective immune response may, in another embodiment, eliminate or reduce the level of infection in an infected subject upon

vaccination.

[0056] An "immunologically effective amount", as used herein, refers to the amount of immunogen that, when administered to a subject, either in a single dose or in a series of doses, is effective for the treatment or prevention of infection by a Plasmodium species. This amount may vary depending upon the health and physical condition of the subject to be treated, as well as on the immunogen. Determination of an effective amount of vaccine for administration to a subject is well within the capabilities of those skilled in the art.

[0057] The term "polymorphism", as used herein, refers to a polymorphic allele and is discussed in the context of the AMA1 protein. It is also contemplated in the context of the RON2 protein. Polymorphism involves one of two or more variants of a particular amino acid/protein sequence. The most common type of polymorphism involves variation at a single amino acid. Polymorphisms can also involve multiple amino acids at different positions and/or long stretches of amino acids, really, any number of changes in the protein sequence in question, for example, AMA1 or RON2.

[0058] An individual referred to as "suffering from" a disease, disorder, and/or condition (e.g. , erythrocytic malaria infection) herein has been diagnosed with and/or displays one or more symptoms of the disease, disorder, and/or condition.

[0059] As used herein, the term "at risk" for disease (such as erythrocytic malaria infection), refers to a subject (e.g. , a human) that is predisposed to contracting the disease and/or expressing one or more symptoms of the disease. Such subjects include those at risk for failing to elicit an immunogenic response to a vaccine against the disease. This predisposition may be genetic (e.g. , a particular genetic tendency to expressing one or more symptoms of the disease, such as heritable disorders, the presence of bacterial species blocking antibodies, the presence of reduced levels of bactericidal antibodies, etc.), or due to other factors (e.g., immune suppressive conditions, environmental conditions, exposures to detrimental compounds, including immunogens, present in the environment, etc.). The term subject "at risk" includes subjects "suffering from disease," i.e., a subject that is experiencing the disease. It is not intended that the present invention be limited to any particular signs or symptoms. Thus, it is intended that the present invention encompasses subjects that are experiencing any range of disease, from sub-clinical infection to full-blown disease, wherein the subject exhibits at least one of the indicia (e.g. , signs and symptoms) associated with the disease.

[0060] Initial manifestations of malaria, common to all malaria species, are similar to flu-like symptoms and can resemble other conditions such as septicemia, gastroenteritis, and viral diseases. The presentation may include headache, fever, shivering, arthralgia (joint pain), vomiting, hemolytic anemia, jaundice, hemoglobinuria, retinal damage, and convulsions. Owing to the non-specific nature of disease presentation, diagnosis of malaria in non-endemic countries requires a high degree of suspicion, which might be elicited by any of the following: recent travel history, splenomegaly (enlarged spleen), fever without localizing signs, thrombocytopenia, and hyperbilirubinemia combined with a normal peripheral blood leukocyte count.

[0061] The classic symptom of malaria is paroxysm— a cyclical occurrence of sudden coldness followed by rigor and then fever and sweating, occurring every two days (tertian fever) in P. vivax and P. ovale infections, and every three days (quartan fever) for P. malariae. P. falciparum infection can cause recurrent fever every 36-48 hours or a less pronounced and almost continuous fever.

[0062] Severe malaria is usually caused by P. falciparum (often referred to as falciparum malaria). Symptoms of falciparum malaria arise 9-30 days after infection

(Bartoloni, et al. 2012 Mediterr J Hematol Infect Dis 4(l):e2012026). Included among the symptoms of malaria (infection) are complications associated with the disease. Among these is the development of respiratory distress, which occurs in up to 25% of adults and 40% of children with severe P. falciparum malaria. Infection with P. falciparum may result in cerebral malaria, a form of severe malaria that involves encephalopathy. It is associated with retinal whitening, which may be a useful clinical sign in distinguishing malaria from other causes of fever. Splenomegaly, severe headache, hepatomegaly (enlarged liver),

hypoglycemia, and hemoglobinuria with renal failure may occur.

[0063] The terms "homologous" and "heterologous", as used herein, refer to polymorphisms within protein, for example, AMA1 alleles among different parasites.

Homologous indicates that the vaccine-type allele and the AMA1 sequence in the parasite used for challenge are similar. Heterologous indicates that the vaccine-type allele and the AMA1 sequence in the parasite used for challenge are different.

[0064] The methods of protecting and/or vaccinating a subject, as disclosed herein, include treating the subject. The terms "treat," "treatment," or "treating", as used herein, refer to any method used to partially or completely alleviate, ameliorate, relieve, inhibit, prevent, delay onset of, reduce severity of and/or reduce incidence of one or more symptoms or features of a disease, disorder, and/or condition {e.g., erythrocytic malaria infection).

Treatment may be administered to a subject who does not exhibit signs of a disease, disorder, and/or condition. In some embodiments, treatment may be administered to a subject who exhibits only early signs of the disease, disorder, and/or condition for the purpose of decreasing the risk of developing pathology associated with the disease, disorder, and/or condition. [0065] As used herein, the terms "immunogenically effective amount,"

"immunologically effective amount", and "antigenically effective amount" refer to that amount of a molecule that elicits and/or increases production of an immune response

(including production of specific antibodies and/or induction of a TCL response) in a host upon vaccination. It is preferred, though not required, that the immunologically-effective (i.e., immunogenically effective) amount is a "protective" amount. The terms "protective" and "therapeutic" amount of a vaccine refer to an amount of the vaccine that prevents, delays, reduces, palliates, ameliorates, stabilizes, and/or reverses disease (for example, erythrocytic malaria infection) and/or one or more symptoms of disease.

[0066] As used herein, the term "vaccination" refers to the administration of a vaccine intended to generate an immune response, for example, to a disease-causing agent. For the purposes of the present invention, vaccination can be administered before, during, and/or after exposure to a disease-causing agent, and, in certain embodiments, before, during, and/or shortly after exposure to the agent. In some embodiments, vaccination includes multiple administrations, appropriately spaced in time, of a vaccinating composition (vaccine).

[0067] The terms "comprises", "comprising", are intended to have the broad meaning ascribed to them in US Patent Law and can mean "includes", "including" and the like.

Additional embodiments of the invention

AMA1/RON2 complex

[0068] The AMA1/RON2 complex may be recombinantly produced, for example, from a genetically engineered system, or it may be a synthetic product, for example, produced by in vitro peptide synthesis or in vitro translation. The AMA1/RON2 complex can, in one embodiment of the invention, be produced by expression of one or more polynucleotides encoding the AMAl and RON2 proteins. For instance, the complex can be produced by expression of a Plasmodium genome encoding an attenuated Plasmodium species, wherein the attenuation does not affect the formation of the complex.

[0069] For the purposes of making a vaccine, recombinant AMAl protein may be made, and the RON2L peptide may be synthesized. The recombinant protein can be expressed in a bacterial system, a yeast system (for example, Pichia), a mammalian system, in vitro, and the like.

[0070] The AMAl protein complex or components thereof may be prepared in a variety of ways in accordance with methods well known in the art. In certain embodiments, the complex is produced by expression of one or more polynucleotides encoding the AMAl and RON2 proteins, fragments of these proteins, or fused molecules. In certain embodiments, in addition to the AMA1 and/or RON2 proteins and/or protein fragments, fused molecules can include other proteins, other fragments, and/or synthetic fragments of amino acid sequence.

[0071] Nucleic acid sequences encoding the AMA1 and RON2 proteins are known in the art and are provided, in whole and/or in part, in public databases such as those at the National Center for Biotechnology Information (NCBI). By way of example, but without limitation, AMA1 and RON2 sequences are provided at GenBank Accession Nos.

XP_001348015.1 (SEQ ID NO:2), XP_729363.1 (SEQ ID NO:4), SEQ ID NO:6, SEQ ID NO:7, and SEQ ID NO: 10. mRNA sequences are provided herein as SEQ ID NO: l and SEQ ID NO:3 and SEQ ID NO:5 and SEQ ID NO:8 and SEQ ID NO:9. These include representative nucleotide sequences of one strain of Plasmodium falciparum parasites.

Sequences of AMA1 and RON2 from other parasites and species and strains are readily available in the literature.

[0072] However, any Plasmodium species comprising an AMA1 and/or RON2 gene locus that is functional or that can be rendered functional via genetic manipulation is suitable for use as a source of the above-mentioned protein complex components. Toxoplasma species (for example, Toxoplasma gondii) are likewise contemplated for use as a source of the AMA1/RON2 complex components, as are other members of the Apicomplexa family of parasites. Furthermore, since AMA1 and RON2 are expressed in Plasmodium sporozoites, the vaccine according to one embodiment of the invention may be used to block this stage of the life cycle, as well.

[0073] In one embodiment, a vaccine according to the invention comprises: i) an immunogenic composition comprising a complex of: a) AMA1; and b) RON2 or a fragment thereof; and ii) at least one adjuvant and/or at least one physiologically acceptable carrier. The full-length RON2 protein is 2189 amino acids (aa). Identified herein is one 49 amino acid region sufficient to bind AMA1 and elicit a protective immune response. The exemplified 49aa sequence is within the full length protein of the mouse malaria parasite (Plasmodium yoelli). The corresponding Plasmodium falciparum sequence and others contemplated herein are, for example, and without limitation, provided in the following alignment:

Pb DITQHATDIGMGPSTSCYTSLVPPPKSICIQQTVKAVLTNSTLASMK Py DITQHATDIGMGPSTSCYTSLLPPPKSICIQQTVKTVLTNSTLASMK Pk DITQHASDIGMGPVTSCYTSTIPPPKQVCIQQAVKVTLTNSTQACMK Pv DISQHATDIGMGPATSCYTSTIPPPKQVCIQQAVKATLTSSTQACMK Pf DITQQAKDIGAGPVASCFTTRMSPPQQICLNSVVNTALSTSTQSAMK Tg DIVQHMEDIGGAPPVSCVTNEILG— VTCAPQAIAKATTSAARVATQ wherein Pb is Plasmodium berghei, Py is Plasmodium yoelli, Pk is Plasmodium knowlesi, Pv is Plasmodium vivax, Pf is Plasmodium falciparum, and Tg is Toxoplasma gondii. The SEQ ID NOs for the sequences shown, above, are as follows: for Pb, SEQ ID NO: 11, for Py, SEQ ID NO: 12, for Pk, SEQ ID NO: 13, for Pv, SEQ ID NO: 14, for Pf, SEQ ID NO: 15, and for Tg, SEQ ID NO: 16.

[0074] A shorter fragment within any such contemplated 49 aa region or another fragment of the full-length protein may likewise exhibit AMAl binding capability and elicit a protective immune response.

[0075] In an additional embodiment, at least one protein of the AMA1/RON2 complex is coupled to a carrier protein. Suitable carrier proteins may include, without limitation, albumin, ovalbumin, a toxin, a growth factor, poly-L-lysine, poly-L-glutamine, or manno se- 6-pho sphate .

[0076] In one embodiment, a vaccine according to the invention comprises a vector comprising at least one nucleic acid molecule encoding a Plasmodium protein or a fragment thereof, selected from AMAl, RON2, or a complex of AMAl and RON2. In another embodiment, two or more of such fragments are expressed on a single polypeptide. A vector can be a replicon (plasmid, phagemid, cosmid, baculovirus, bacmid, bacterial artificial chromosome, yeast artificial chromosome, as well as other bacterial, yeast, and viral vectors, such as lentiviruses, retroviruses, adenoviruses, and adeno-associated viruses). The vector can further be an expression vector, which comprises expression control sequences operatively linked to a nucleotide sequence to be expressed.

[0077] The AMA1/RON2 complex is, in one embodiment, prepared by mixing recombinant AMAl protein and RON2L peptide (wherein the ratio of AMAl:RON2L can be adjusted according to specific needs) and incubating at room temperature for about 30 min and formulated with adjuvant. In another embodiment, for human use, the complex is further purified to remove any un-complexed AMAl and/or RON2. In yet another embodiment, the protein binding results in the presentation of an epitope that allows the desired antibody binding to occur.

[0078] A fusion protein can, in one embodiment, be recombinant, created artificially using recombinant DNA technology. A recombinant fusion protein is created through genetic engineering of a fusion gene. This typically involves removing the stop codon from a cDNA sequence coding for the first protein, then appending the cDNA sequence of the second protein in frame through ligation or overlap extension PCR. That DNA sequence will then be expressed by a cell as a single protein. The protein can be engineered to include the full sequence of both original proteins, or only a portion of one or both of the proteins.

[0079] The invention also provides polypeptides and corresponding polynucleotides required for synthesis of an AMA1/RON2 protein complex. The complex includes both naturally occurring and unnaturally occurring polynucleotides and polypeptide products thereof. Naturally occurring biosynthesis products include distinct gene and polypeptide species as well as corresponding species homologs expressed in various Plasmodium strains. Non-naturally occurring biosynthesis products include variants of the naturally occurring products such as analogs and biosynthesis products including covalent modifications. .

[0080] Purified and isolated Plasmodium polynucleotides (e.g., DNA sequences and

RNA transcripts, both sense and complementary antisense strands) encode the bacterial AMAl and/or RON2 biosynthesis gene products. Genomic DNA comprises the protein coding region for a polypeptide of the complex and includes variants that may be found in other Plasmodium strains. "Synthesized," as used herein and is understood in the art, refers to purely chemical, as opposed to enzymatic, methods for producing polynucleotides. "Wholly" synthesized DNA sequences are, therefore, produced entirely by chemical means, and "partially" synthesized DNAs embrace those wherein only portions of the resulting DNA were produced by chemical means. Preferred mRNA sequences encoding Plasmodium AMAl biosynthesis gene products are set out in SEQ ID NO: l and species homologs thereof. Preferred mRNA sequences encoding Plasmodium RON2 biosynthesis gene products are set out in SEQ ID NO:3 and species homologs thereof.

[0081] Autonomously replicating recombinant expression constructions such as plasmid and viral DNA vectors incorporating the biosynthesis gene sequences are also provided. Expression constructs wherein AMAl and/or RON2 biosynthesis polypeptide- encoding polynucleotides are operatively linked to an endogenous or exogenous expression control DNA sequence and a transcription terminator are also provided. The biosynthesis genes may be cloned by PCR, using Plasmodium genomic DNA as the template. For ease of inserting the gene into expression vectors, PCR primers are chosen, so that the PCR- amplified gene(s) has a restriction enzyme site at the 5' end preceding the initiation codon ATG, and a restriction enzyme site at the 3' end after the termination codon TAG, TGA or TAA. If desirable, the codons in the gene(s) are changed, without changing the amino acids, according to E. coli codon preference described by Grosjean et al. (1982) Gene, 18:199-209; and Konigsberg et al. (1983) Proc. Natl. Acad. Sci. USA, 80:687-691. Optimization of codon usage may lead to an increase in the expression of the gene product when produced in E. coli. If a protein gene product is to be produced extracellularly, either in the periplasm of E. coli or other bacteria, or into the cell culture medium, the gene is cloned into an expression vector and linked to a signal sequence.

[0082] According to another aspect of the invention, host cells are provided, including prokaryotic and eukaryotic cells, either stably or transiently transformed, transfected, or electroporated with polynucleotide sequences of the complex proteins in a manner which permits expression of AMA1 and/or RON2 biosynthesis polypeptides.

Potential expression systems of the invention include bacterial, yeast, fungal, viral, parasitic, invertebrate, and mammalian cells systems. Host cells of the invention are a valuable source of immunogen for development of antibodies specifically immunoreactive with the

AMA1/RON2 complex. Host cells of the invention are conspicuously useful in methods for large scale production of AMA1 and/or RON2 biosynthesis polypeptides, wherein the cells are grown in a suitable culture medium and the desired polypeptide products are isolated from the cells or from the medium in which the cells are grown by, for example,

immunoaffinity purification or any of the multitude of purification techniques well known and routinely practiced in the art. Any suitable host cell may be used for expression of the gene product, such as E. coli, other bacteria, including P. multocida, Bacillus and S. aureus, yeast, including Pichia pastoris and Saccharomyces cerevisiae, insect cells, or mammalian cells, including CHO cells, utilizing suitable vectors known in the art. Proteins may be produced directly or fused to a peptide or polypeptide, and either intracellularly or extracellularly by secretion into the periplasmic space of a bacterial cell or into the cell culture medium. Secretion of a protein requires a signal peptide (also known as pre- sequence); a number of signal sequences from prokaryotes and eukaryotes are known to function for the secretion of recombinant proteins. During the protein secretion process, the signal peptide is removed by signal peptidase to yield the mature protein.

[0083] To simplify the protein purification process, a purification tag may be added either at the 5' or 3' end of the gene coding sequence. Commonly used purification tags include a stretch of six histidine residues (US Patent Nos. 5,284,933 and 5,310,663), a streptavidin affinity tag described by Schmidt et al. (1993) Protein Eng., 6: 109-122, a FLAG peptide (Hopp, et al. (1988) Biotechnology, 6: 1205-1210), glutathione 5-transferase (Smith, et al. (1988) Gene, 67:31-40), and thioredoxin (LaVallie, et at. (1993) Bio/Technology, 11: 187-193). To remove these peptide or polypeptides, a proteolytic cleavage recognition site may be inserted at the fusion junction. Commonly used proteases are factor Xa, thrombin, and enterokinase.

[0084] In one embodiment, the invention employs purified and isolated Plasmodium

AMA1 and/or RON2 biosynthesis polypeptides as described above. The invention also embraces polypeptides that have at least about 99%, at least about 95%, at least about 90%, at least about 85%, at least about 80%, at least about 75%, at least about 70%, at least about 65%, at least about 60%, at least about 55%, and at least about 50% identity and/or homology to the polypeptides of the complex. Percent amino acid sequence "identity" with respect to the preferred polypeptides of the invention is defined herein as the percentage of amino acid residues in the candidate sequence that are identical with the residues in the AMA1 and/or RON2 biosynthesis gene product sequence after aligning both sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and not considering any conservative substitutions as part of the sequence identity. Percent sequence "homology" with respect to the polypeptides of the complex is defined herein as the percentage of amino acid residues in the candidate sequence that are identical with the residues in one of the biosynthesis polypeptide sequences after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent sequence identity, and also considering any conservative substitutions as part of the sequence identity. Conservative substitutions are well known in the art.

[0085] Polypeptides of the complex may be isolated from natural bacterial cell sources or may be chemically synthesized but are preferably produced by recombinant procedures involving host cells. AMA1 and/or RON2 biosynthesis gene products of the invention may be full-length polypeptides, biologically active fragments, or variants thereof which retain specific biological or immunological activity. The biological activity is, in one embodiment, the ability of the two proteins or fragments of either or both to bind to one another. The immunological activity is, in one embodiment, the protective immunological activity of the antigenic gene product, for example, the AMA1/RON2 complex product. Variants may comprise biosynthesis polypeptide analogs wherein one or more of the specified (i.e., naturally encoded) amino acids is deleted or replaced, or wherein one or more non-specified amino acids are added: (1) without loss of one or more of the biological activities or immunological characteristics (activity) specific for the biosynthesis gene product; or (2) with specific disablement of a particular biological activity of the biosynthesis gene product. Deletion variants contemplated also include fragments lacking portions of the polypeptide not essential for biological activity, and insertion variants include fusion polypeptides in which the wild-type polypeptide or fragment thereof have been fused to another polypeptide.

[0086] Variant AMA1 and/or RON2 biosynthesis polypeptides include those wherein conservative substitutions have been introduced by modification of polynucleotides encoding polypeptides of the complex. Conservative substitutions are recognized in the art to classify amino acids according to their related physical properties and are known in the art (see, for example, Lehninger (Biochemistry, Second Edition (1975) W.H. Freeman & Co., pp.71-77).

[0087] Variant AMA1 and/or RON2 biosynthesis products of the complex include mature biosynthesis gene products, i.e., wherein leader or signal sequences are removed, having additional amino terminal residues. Variants contemplated herein also include gene products wherein amino-terminal sequences derived from other proteins have been introduced, as well as variants comprising amino-terminal sequences that are not found in naturally occurring proteins.

[0088] The invention also embraces variant polypeptides having additional amino acid residues resulting from the use of specific expression systems. For example, use of commercially available vectors that express a desired polypeptide as a fusion protein with glutathione-S-transferase (GST) provide the desired polypeptide having an additional glycine residue at position -1 following cleavage of the GST component from the desired

polypeptide. Variants that result from expression using other vector systems are also contemplated.

Antibodies

[0089] Antibodies or epitope-binding fragments thereof which specifically bind to at least a portion of the AMA1 or RON2 or the AMA1/RON2 complex inhibit invasion of merozoites into RBCs (red blood cells). These antibodies can be monoclonal and/or polyclonal antibodies, recombinant antibodies (for example, single-chain antibodies, phage - displayed antibodies, diabodies), and antigen-binding fragments of antibodies, such as Fab or Fv. In one embodiment, the antibodies recognize epitopes of AMA1, RON2, as presented in the AMA1/RON2 complex. Methods for raising and purifying antibodies, including neutralizing antibodies, are well known in the art. In additional embodiments, the antibodies are human or humanized. They can also be human or humanized antibody homologs, chimeric antibodies, chimeric antibody homologs, monomers or dimers of antibody heavy or light chains.

[0090] The antibodies contemplated include other binding proteins specific for

AMA1 and RON2 biosynthesis gene products or fragments thereof. The term "specific for" indicates that the variable regions of the antibodies described herein recognize and bind AMA1 and/or RON2 exclusively (i.e., are oftentimes able to distinguish a single O antigen from related O antigens, but may also interact with other proteins (for example, S. aureus protein A or other antibodies in ELISA techniques) through interactions with sequences outside the variable region of the antibodies, and in particular, in the constant region of the molecule. Screening assays to determine binding specificity of an antibody of the invention are well known and routinely practiced in the art. For a comprehensive discussion of such assays, see Harlow et al. (Eds.); Antibodies A Laboratory Manual (1988) Cold Spring Harbor Laboratory; Cold Spring Harbor, N.Y, chapter 6. Antibodies that recognize and bind fragments of the O antigen of the invention are also contemplated, provided that the antibodies are first and foremost specific for, as defined above, an O antigen of the invention from which the fragment was derived.

[0091] Such antibodies can further comprise glycosylation that has been modulated by expression in yeast cells that have been engineered to add glycan structures to proteins. Other modifications of antibodies are likewise contemplated, including via covalent attachment of a molecule - acylated antibodies, pegylated antibodies, phosphorylated antibodies, and amidated antibodies.

[0092] The antibodies can also be variants having single or multiple amino acid substitutions, deletions, additions, or replacements, as long as they retain their desired biological property(ies), i.e., internalization, binding affinity or avidity, and/or immune effector activity.

[0093] The antibodies can, in further embodiments, be labeled (radioisotope labels, fluorescent labels) or conjugated to various moieties, including detectable moieties and drugs or toxins (bacterial toxins, organic chemicals, inorganic chemicals, and the like).

Treatment/Therapy

[0094] In certain embodiments, the present invention vaccines and methods to treat

{e.g., alleviate, ameliorate, relieve, delay onset of, inhibit progression of, reduce severity of, and/or reduce incidence of one or more symptoms or features of) and/or prevent Plasmodium infection.

[0095] In some embodiments, methods of vaccination and/or treatment (such as those described in the sections below) involve stratification of a patient population based on prior exposure to Plasmodium strains. Such methods involve steps of determining whether a patient has been previously exposed to one or more of the strains. In some embodiments, if it is determined that a patient has been previously been exposed to one or more of the strains, that patient may receive less concentrated, less potent, and/or less frequent doses of the inventive vaccine or composition. If it is determined that a patient has not been previously been exposed to one or more of the Plasmodium strains, that patient may receive more concentrated, more potent, and/or more frequent doses of the inventive vaccine.

[0096] In one embodiment, the vaccine of the invention treats more than one

Plasmodium infection, i.e., infection with more than one Plasmodium strain/species. In another embodiment, the vaccine according to the invention is administered in combination with a distinct therapy.

[0097] A number of factors can be taken into account when determining the distinct therapy: the infecting species of Plasmodium parasite, the clinical situation of the patient (for example, adult, child, or pregnant female, with either mild or severe malaria), and the drug susceptibility of the infecting parasites. Drug susceptibility is determined by the geographic area where the infection was acquired. Different areas of the world have malaria types that are resistant to certain medications.

[0098] Furthermore, while mild malaria can be treated with oral medication; severe malaria (one or more symptoms of either impaired consciousness/coma, severe anemia, renal failure, pulmonary edema, acute respiratory distress syndrome, shock, disseminated intravascular coagulation, spontaneous bleeding, acidosis, hemoglobinuria [hemoglobin in the urine], jaundice, repeated generalized convulsions, and/or parasitemia ([parasites in the blood] > 5%) requires intravenous (IV) drug treatment and fluids in the hospital.

[0099] Known drug treatments of malaria include, but are not limited to, chloroquine, quinine sulfate plus doxycycline, tetracycline, clindamycin, atovaquone-proguanil, artemisinin-derived combination therapy (ACTs)— drug combinations like artesunate- amodiaquine, artesunate-mefloquine, artesunate-pyronaridine, dihydroartemisinin- piperaquine, and chlorproguanil-dapsoneartesunate, and spiroindolones.

Vaccine

[00100] A "vaccine" is a composition that induces an immune response in the recipient or host of the vaccine. The vaccine can induce protection against infection upon subsequent challenge with a bacterial species, herein a Plasmodium species. Protection refers to resistance {e.g., partial resistance) to persistent infection of a host animal with at least one Plasmodium species. Neutralizing antibodies generated in the vaccinated host can provide this protection. In other situations, CTL responses can provide this protection. In some situations, both neutralizing antibodies and cell-mediated immune {e.g., CTL) responses provide this protection. [00101] Vaccines are useful in preventing or reducing infection or disease by inducing immune responses, to an antigen or antigens, in an individual. For example, vaccines can be used prophylactic ally in naive individuals or therapeutically in individuals already infected with at least one Plasmodium species.

[00102] Protective responses can be evaluated by a variety of methods. For example, either the generation of neutralizing antibodies against Plasmodium proteins, specifically, AMA1/RON2, and/or the generation of a cell-mediated immune response against such proteins can indicate a protective response. Protective responses also include those responses that result in lower number of bacteria colonized in a vaccinated host animal exposed to a given inoculum (of the bacteria) as compared to a host animal exposed to the same inoculum, but that has not been administered the vaccine.

[00103] The compositions of the present invention are preferably given to an individual in a "prophylactically effective amount" or a "therapeutically effective amount" (as the case may be, although prophylaxis may be considered therapy), this being sufficient to show benefit to the individual. The actual amount administered, and rate and time- course of administration, will depend on the nature and severity of who/what is being treated.

Prescription of treatment, e.g., final decisions on acceptable dosage etc., will be dictated by Vaccine Regulatory Authorities, after review of safety and efficacy data following human immunizations. Thus, a vaccine according to the invention comprises an immunoprotective or immunotherapeutic and non-toxic amount of the vaccine strain. Suitable dosage amounts can be determined by the person skilled in the art.

[00104] In general, a vaccine according to the invention will include a "therapeutic agent" (the immunogenic composition comprising the AMA1/RON2 complex), in addition to one or more inactive, agents such as a sterile, biocompatible pharmaceutical carrier including, but not limited to, sterile water, saline, buffered saline, or dextrose solution. Alternatively or additionally, the vaccine may comprise a pharmaceutically acceptable excipient, which, as used herein, includes any and all solvents, dispersion media, diluents, or other liquid vehicles, dispersion or suspension aids, disintegrating agents, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, buffering agents, solid binders, granulating agents, lubricants, coloring agents, sweetening agents, flavoring agents, perfuming agents, and the like, as suited to the particular dosage form desired. Remington's The Science and Practice of Pharmacy, 21st Ed., A. R. Gennaro, (Lippincott, Williams & Wilkins, Baltimore, Md., 2006; incorporated herein by reference) discloses various excipients used in formulating pharmaceutical compositions and known techniques for the preparation thereof. Except insofar as any conventional excipient medium is incompatible with a substance or its derivatives, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component of the vaccine, its use is contemplated to be within the scope of this invention.

[00105] Thus, vaccines according to the present invention, and for use in accordance with the present invention, may include, in addition to active ingredient, a pharmaceutically or physiologically acceptable excipient, carrier, buffer, stabilizer or other materials well known to those skilled in the art. Such materials should be non-toxic and should not interfere with the efficacy of the active ingredient. The precise nature of the carrier or other material will depend on the route of administration. Examples of techniques and protocols mentioned above can be found in Remington's Pharmaceutical Sciences, 16th edition, Osol, A. (Ed.), 1980.

[00106] Vaccines according to the invention may, in one embodiment, contain an adjuvant. The term "adjuvant", as used herein, refers to any compound which, when injected together with an antigen, non- specifically enhances the immune response to that antigen. Exemplary adjuvants include Complete Freund's Adjuvant, Incomplete Freund's Adjuvant, Gerbu adjuvant (GMDP; C.C. Biotech Corp.), RIBI fowl adjuvant (MPL; RIBI

Immunochemical Research, Inc.), metal salts (aluminum salts, calcium salts), potassium alum, aluminum phosphate, aluminum hydroxide, QS21 (Cambridge Biotech), TITERMAX ® adjuvant (CytRx Corporation), and QUIL A ® adjuvant. Other compounds that may have adjuvant properties include binders such as carboxymethylcellulose, ethyl cellulose, microcrystalline cellulose, or gelatin; excipients such as starch, lactose or dextrins, disintegrating agents such as alginic acid, sodium alginate, PREVIOGEL ® , corn starch and the like; lubricants such as magnesium stearate or STEROTEX ® ; glidants such as colloidal silicon dioxide; sweetening agents such as sucrose or saccharin, a flavoring agent such as peppermint, methyl salicylate or orange flavoring, and a coloring agent.

[00107] Furthermore, a useful compendium of many adjuvants is prepared by the

National Institutes of Health and can be found on the internet

(www.niaid.nih.gov/daids/vaccine/pdf/compendium.pdf). Hundreds of different adjuvants are known in the art and could be employed in the practice of the present invention.

Exemplary adjuvants that can be utilized in accordance with the invention include, but are not limited to, cytokines, aluminum salts (e.g. , aluminum hydroxide, aluminum phosphate, etc.), gel-type adjuvants (e.g. , calcium phosphate, etc.); microbial adjuvants (e.g. ,

immunomodulatory DNA sequences that include CpG motifs; endotoxins such as monophosphoryl lipid A); exotoxins such as cholera toxin, E. coli heat labile toxin, and pertussis toxin; muramyl dipeptide, etc.); oil-emulsion and emulsifier-based adjuvants (e.g. , Freund's Adjuvant, MF59 [Novartis], SAF, etc.); particulate adjuvants (e.g. , liposomes, biodegradable microspheres, etc.); synthetic adjuvants (e.g. , nonionic block copolymers, muramyl peptide analogues, polyphosphazene, synthetic polynucleotides, etc.); and/or combinations thereof. Other exemplary adjuvants include some polymers (e.g. ,

polyphosphazenes), Q57, saponins (e.g. , QS21), squalene, tetrachlorodecaoxide, CPG 7909, poly[di(carboxylatophenoxy)phosphazene] (PCCP), interferon-gamma, block copolymer P1205 (CRL1005), interleukin-2 (IL-2), polymethyl methacrylate (PMMA), etc.

[00108] Vaccines according to the invention may, in another embodiment, be formulated using a diluent. Exemplary "diluents" include water, physiological saline solution, human serum albumin, oils, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents, antibacterial agents such as benzyl alcohol, antioxidants such as ascorbic acid or sodium bisulphite, chelating agents such as ethylene diamine-tetra- acetic acid, buffers such as acetates, citrates or phosphates and agents for adjusting the osmolality, such as sodium chloride or dextrose. Exemplary "carriers" include liquid carriers (such as water, saline, culture medium, saline, aqueous dextrose, and glycols) and solid carriers (such as carbohydrates exemplified by starch, glucose, lactose, sucrose, and dextrans, anti-oxidants exemplified by ascorbic acid and glutathione, and hydrolyzed proteins.

[00109] The AMA1/RON2 protein complex may, for example, be formulated into an emulsion or into biodegradable microspheres or liposomes.

[00110] Vaccines according to the invention may, in still another embodiment, contain an excipient. The term "excipient" refers herein to any inert substance (e.g. , gum arabic, syrup, lanolin, starch, etc.) that forms a vehicle for delivery of an antigen. The term excipient includes substances that, in the presence of sufficient liquid, impart to a composition the adhesive quality needed for the preparation of pills or tablets.

[00111] As mentioned above, in some embodiments, interfering agents and/or binding agents in accordance with the invention may be utilized for prophylactic applications. In some embodiments, prophylactic applications involve systems and methods for preventing, inhibiting progression of, and/or delaying the onset of bacterial infection. In some

embodiments, interfering agents may be utilized for passive immunization (i.e., immunization wherein antibodies are administered to a subject). In some embodiments, vaccines for passive immunization may comprise antibody interfering agents, such as those described herein. In some embodiments, passive immunization occurs when antibodies are transferred from mother to fetus during pregnancy. In some embodiments, antibodies are administered directly to an individual (e.g., by injection, orally, etc.). Of note, it is possible to immunize a person and use their antibodies for passive protection in another individual.

[00112] For example, in some embodiments, interfering agent and/or binding agent polypeptides, nucleic acids encoding such polypeptides, characteristic or biologically active fragments of such polypeptides or nucleic acids, antibodies that bind to and/or compete with such polypeptides or fragments, small molecules that interact with or compete with such polypeptides or with glycans that bind to them, etc. are included in the vaccines described herein. In some embodiments, interfering agents and/or binding agents that are not polypeptides, e.g., that are small molecules, umbrella topology glycans and mimics thereof, carbohydrates, aptamers, polymers, nucleic acids, etc., are included in the vaccines.

Administration

[00113] The mode of administration of a vaccine according to the present invention is any suitable route that delivers an immunoprotective or immunotherapeutic amount of the vaccine to the subject and is described below.

[00114] Vaccines may be administered using any amount and any route of

administration effective for treatment and/or vaccination. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular composition, its mode of administration, its mode of activity, and the like. Vaccines are typically formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the vaccines of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular subject or organism will depend upon a variety of factors including the severity of the malaria infection; the activity of the specific vaccine

composition employed; the half-life of the composition after administration; the age, body weight, general health, sex, and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific components employed; the duration of the treatment; drugs used in combination or coincidental with the specific components employed; and like factors, well known in the medical arts.

[00115] Vaccines according to the invention may be administered by any route. In some embodiments, the vaccines are administered by a variety of routes, including oral (PO), intravenous (IV), intramuscular (IM), intra-arterial, intramedullary, intrathecal, subcutaneous (SQ), intraventricular, transdermal, interdermal, intradermal, rectal (PR), vaginal, intraperitoneal (IP), intragastric (IG), topical or transcutaneous (e.g. , by powders, ointments, creams, gels, lotions, and/or drops), mucosal, intranasal, buccal, enteral, vitreal, sublingual; by intratracheal instillation, bronchial instillation, and/or inhalation; as an oral spray, nasal spray, and/or aerosol, and/or through a portal vein catheter.

[00116] In general, the most appropriate route of administration will depend upon a variety of factors including the nature of the agent being administered (e.g. , its stability upon administration), the condition of the subject (e.g. , whether the subject is able to tolerate a particular mode of administration), etc. In specific embodiments, vaccines may be administered intranasally. In specific embodiments, vaccines may be administered by intratracheal instillation. In specific embodiments, vaccines may be administered by bronchial instillation. In specific embodiments, vaccines may be administered by inhalation. In specific embodiments, vaccines may be administered as a nasal spray. In specific embodiments, vaccines may be administered mucosally. In specific embodiments, vaccines may be administered orally. In specific embodiments, vaccines may be administered by intravenous injection. In specific embodiments, vaccines may be administered by

intramuscular injection. In specific embodiments, vaccines may be administered by subcutaneous injection. The oral or nasal spray or aerosol route (e.g., by inhalation) is most commonly used to deliver therapeutic agents (e.g., an immunogenic composition, comprising the AMA1/RON2 complex) directly to the lungs and respiratory system. However, the invention encompasses the delivery of such a composition by any appropriate route taking into consideration likely advances in the sciences of drug delivery.

[00117] For oral administration, a vaccine according to the invention may be presented as capsules, tablets, dissolvable membranes, powders, granules, or as a suspension. The vaccine may have conventional additives, such as lactose, mannitol, corn starch, or potato starch. The vaccine also may be presented with binders, such as crystalline cellulose, cellulose derivatives, acacia, corn starch, or gelatins. Additionally, the vaccine may be presented with disintegrators, such as corn starch, potato starch, or sodium

carboxymethylcellulose. The vaccine may be further presented with dibasic calcium phosphate anhydrous or sodium starch glycolate. Finally, the vaccine may be presented with lubricants, such as talc or magnesium stearate.

[00118] For parenteral administration, a vaccine according to the invention may be prepared with a sterile aqueous solution, which is preferably isotonic with the blood of the subject. Such a formulation may be prepared by dissolving a solid active ingredient in water containing physiologically compatible substances, such as sodium chloride, glycine, and the like, and having a buffered pH compatible with physiological conditions, so as to produce an aqueous solution, then rendering said solution sterile. The vaccine may be presented in unit or multi-dose containers, such as sealed ampoules or vials. The vaccine also may be delivered by any mode of injection, including any of those described herein.

[00119] Vaccines for rectal or vaginal administration are typically suppositories, which can be prepared by mixing an immunogenic composition with suitable non-irritating excipients such as cocoa butter, polyethylene glycol, or a suppository wax, which are solid at ambient temperature but liquid at body temperature and, therefore, melt in the rectum or vaginal cavity and release the immunogenic composition.

[00120] Solid dosage forms for oral administration include capsules, tablets, pills, powders, dissolvable membranes, and granules. In such solid dosage forms, the immunogenic composition is mixed with at least one inert, pharmaceutically acceptable excipient such as sodium citrate or dicalcium phosphate and/or fillers or extenders (e.g., starches, lactose, sucrose, glucose, mannitol, and silicic acid), binders (e.g., carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia), humectants (e.g., glycerol),

disintegrating agents (e.g. , agar, calcium carbonate, potato starch, tapioca starch, alginic acid, certain silicates, and sodium carbonate), solution retarding agents (e.g. , paraffin), absorption accelerators (e.g. , quaternary ammonium compounds), wetting agents (e.g. , cetyl alcohol and glycerol monostearate), absorbents (e.g., kaolin and bentonite clay), and lubricants (e.g. , talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate), taste/olfactory components, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may comprise buffering agents.

[00121] Dosage forms for topical and/or transdermal administration of a vaccine in accordance with this invention may include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants and/or patches. Generally, the immunogenic composition is admixed under sterile conditions with a pharmaceutically acceptable excipient and/or any needed preservatives and/or buffers as may be required. Additionally, the present invention contemplates the use of transdermal patches, which often have the added advantage of providing controlled delivery of an active therapeutic agent (e.g., the immunogenic composition) to the body. Such dosage forms may be prepared, for example, by dissolving and/or dispensing the immunogenic composition in the proper medium. Alternatively or additionally, the rate may be controlled by either providing a rate controlling membrane and/or by dispersing the compound in a polymer matrix and/or gel. [00122] For transdermal administration, the vaccine according to the invention may be combined with skin penetration enhancers, such as propylene glycol, polyethylene glycol, isopropanol, ethanol, oleic acid, N-methylpyrrolidone, and the like, which increase the permeability of the skin to the immunogenic composition and permit the composition to penetrate through the skin and into the bloodstream. Enhancer and immunogenic

composition also may be further combined with a polymeric substance, such as

ethylcellulose, hydroxypropyl cellulose, ethylene/vinylacetate, polyvinyl pyrrolidone, and the like, to provide the composition in gel form, which may be dissolved in solvent, such as methylene chloride, evaporated to the desired viscosity, and then applied to backing material to provide a patch. The ensuing vaccine may be administered transdermally, at or near the site on the subject where the infection, neoplasm, or other disorder may be localized.

Alternatively, the vaccine may be administered transdermally at a site other than the affected area, in order to achieve systemic administration.

[00123] For intranasal administration (e.g., nasal sprays) and/or pulmonary

administration (administration by inhalation), a vaccine according to the invention, including an aerosol formulation, may be prepared in accordance with procedures well known to persons of skill in the art. Aerosol formulations may comprise either solid particles or solutions (aqueous or non-aqueous). Nebulizers (e.g. , jet nebulizers, ultrasonic nebulizers, etc.) and atomizers may be used to produce aerosols from solutions (e.g. , using a solvent such as ethanol); metered-dose inhalers and dry-powder inhalers may be used to generate small- particle aerosols. The desired aerosol particle size can be obtained by employing any one of a number of methods known in the art, including, without limitation, jet-milling, spray drying, and critical-point condensation.

[00124] Vaccines for intranasal administration may be solid formulations (e.g. , a coarse powder) and may contain excipients (e.g. , lactose). Solid formulations may be administered from a container of powder held up to the nose, using rapid inhalation through the nasal passages. Vaccines for intranasal administration may also comprise aqueous or oily solutions of nasal spray or nasal drops. For use with a sprayer, the vaccine may comprise an aqueous solution and additional agents, including, for example, an excipient, a buffer, an isotonicity agent, a preservative, or a surfactant. A nasal spray may be produced, for example, by forcing a suspension or solution of the immunogenic composition through a nozzle under pressure.

[00125] Formulations of a vaccine according to the invention for pulmonary administration may be presented in a form suitable for delivery by an inhalation device and may have a particle size effective for reaching the lower airways of the lungs or sinuses. For absorption through mucosal surfaces, including the pulmonary mucosa, the formulation may comprise an emulsion that includes, for example, a bioactive peptide, a plurality of submicron particles, a mucoadhesive macromolecule, and/or an aqueous continuous phase. Absorption through mucosal surfaces may be achieved through mucoadhesion of the emulsion particles.

[00126] Vaccines according to the invention for use with a metered-dose inhaler device may include a finely-divided powder containing the immunogenic composition as a suspension in a non-aqueous medium. For example, the composition may be suspended in a propellant with the aid of a surfactant (e.g., sorbitan trioleate, soya lecithin, or oleic acid). Metered-dose inhalers typically use a propellant gas (e.g., a chlorofluorocarbon, a

hydrochlorofluorocarbon, a hydrofluorocarbon, or a hydrocarbon) stored in a container (e.g., a canister) as a mixture (e.g., as a liquefied, compressed gas). Inhalers require actuation during inspiration. For example, actuation of a metering valve may release the mixture as an aerosol. Dry-powder inhalers use breath- actuation of a mixed powder.

[00127] A vaccine according to the invention also may be released or delivered from an osmotic mini-pump or other timed-release device. The release rate from an elementary osmotic mini-pump may be modulated with a microporous, fast-response gel disposed in the release orifice. An osmotic mini-pump would be useful for controlling release, or targeting delivery, of the vaccine.

[00128] A vaccine according to the invention may be administered or introduced to a subject by known techniques used for the introduction of drugs, including, for example, injection and transfusion.

[00129] A vaccine according to the invention may be administered to a subject, either alone or in combination with one or more drugs used to treat the infection or a symptom of the same. A vaccine according to the invention may be administered alone or in combination with other treatments, either simultaneously or sequentially dependent upon the condition to be treated.

Dosages

[00130] The dosage of a vaccine (or other composition) according to the invention can be determined by, for example, first identifying doses effective to elicit a prophylactic and/or therapeutic immune response. This may be accomplished by measuring the serum titer of Plasmodium-specific immunoglobulins (anti-AMAl or -RON2 or -AMA1/RON2 antibodies) and/or by measuring the inhibitory ratio of antibodies in serum samples. The dosages can be determined from animal studies, including animals that are not natural hosts to the parasite species in question. For example, the animals can be dosed with a vaccine candidate, e.g., a vaccine according to the invention, to partially characterize the immune response induced and/or to determine if any neutralizing antibodies have been produced. In addition, routine human clinical studies can be performed to determine the effective dose for humans.

[00131] In one embodiment, a vaccine dose consists of a range of about 1 μg to about

1 mg total protein. In another embodiment, the range is about 0.1 mg to about 1.0 mg total protein. Such a dosage could be adjusted based on the amount of polypeptide delivered. More precise dosages can further be determined by assessing the immunogenicity of the AMA1/RON2 complex.

[00132] Effective doses may be extrapolated from dose-response curves derived from in vitro and/or in vivo animal models. For example, various immunization schedules could be evaluated for optimum ensuing protection (and therapy).

[00133] An immunologically effective amount, based upon human studies, would, in one embodiment, be sufficient to stimulate an acceptable level of protective immunity in a population. For some vaccines (in certain embodiments), this immunologically effective level would provide an 80% efficacy against a malaria. For other vaccines (in other embodiments), an immunologically effective amount would be one that protects against severe malaria but may not protect against all symptoms of the infection.

[00134] In one embodiment, a vaccine according to the invention may be administered to a subject at risk of developing malaria, in an amount effective to prevent the disorder in the subject. As used herein, the phrase "effective to prevent the disorder" includes effective to hinder or prevent the development or manifestation of clinical impairment or symptoms resulting from the disorder, or to reduce in intensity, severity, and/or frequency, and/or delay of onset of one or more symptoms of the disorder.

Combinations

[00135] Compositions and vaccines according to the invention can be administered to a subject either alone or in combination with one or more other therapeutic agents including, but not limited to, vaccines and/or antibodies. By "in combination with," it is not intended to imply that the agents must be administered at the same time or formulated for delivery together, although these methods of delivery are within the scope of the invention.

Compositions and vaccines according to the invention can be administered concurrently with, prior to, or subsequent to, one or more other desired therapeutics or medical procedures. It will be appreciated that therapeutically active agents utilized in combination may be administered together in a single composition or administered separately in different compositions. In general, each agent will be administered at a dose and/or on a time schedule determined for that agent.

[00136] In general, each agent (in this context, one of the "agents" is a composition or vaccine according to the invention) will be administered at a dose and on a time schedule determined for that agent. Additionally, the invention encompasses the delivery of the compositions in combination with agents that may improve their bioavailability, reduce or modify their metabolism, inhibit their excretion, or modify their distribution within the body. Although the compositions (including vaccines) according to the invention can be used for treatment and/or vaccination of any subject, they are preferably used in the treatment and/or vaccination of humans.

[00137] The particular combination of therapies (e.g., therapeutics or procedures) to employ in a combination regimen will take into account compatibility of the desired therapeutics and/or procedures and the desired therapeutic effect to be achieved. It will be appreciated that the therapies employed may achieve a desired effect for the same purpose (for example, an agent useful for treating, preventing, and/or delaying the onset of a bacterial (or other microorganism) infection may be administered concurrently with another agent useful for treating, preventing, and/or delaying the onset of the bacterial infection), or they may achieve different effects (e.g., prevention of severe illness or control of adverse effects).

[00138] A vaccine according to certain embodiments of the invention may further comprise one or more antigens. Such antigens may, for example, be derived from a

Plasmodium species and may be capable of eliciting an immune response directed to the Plasmodium species.

[00139] In general, it is expected that agents utilized in combination will be utilized at levels that do not exceed the levels at which they are utilized individually. In some embodiments, the levels utilized in combination will be lower than those utilized

individually.

Kits

[00140] Kits comprising an immunogenic composition comprising a recombinant

AMA1/RON2 complex or a vaccine according to the invention are provided in an additional embodiment. Kits can include one or more other elements including, but not limited to, instructions for use; other reagents, e.g., a diluent, devices or other materials for preparing the vaccine or composition for administration; pharmaceutically acceptable carriers; and devices or other materials for administration to a subject. Instructions for use can include instructions for therapeutic application (e.g., DNA vaccination and protein boosting) including suggested dosages and/or modes of administration, e.g., in a human subject, as described herein.

[00141] In another embodiment, a kit according to the invention can further contain at least one additional reagent, such as a diagnostic or therapeutic agent, e.g., a diagnostic agent to monitor an immune response to the vaccines according to the invention in the subject, or an additional therapeutic agent as described herein (see, e.g., the section herein describing combination therapies).

EXAMPLES

[00142] The following examples are provided for illustrative purposes only and are not intended to limit the scope of the invention.

Example 1

[00143] Recombinant P. yoelli AMAl with a c-terminal His tag was expressed in E. coli, purified on a nickel column and subsequently refolded. The refolded protein was confirmed to bind a conformation-dependent mAb to verify proper folding.

[00144] PyRON2L peptide comprising the 49-amino acid region that binds AMAl was synthesized (Lifetein LLC). Mice (5/group) were immunized with PBS (control), PyAMAl,

PyRON2L, PyAMAl + PyRON2L (the two antigens injected at separate sites), or

PyAM A 1 /PyRON2L (AMA1-RON2L preformed complex). AMA1/RON2L complex was prepared by mixing AMAl and RON2L (1:2 ratio) and allowing complex formation to occur at room temperature for 30 min.

[00145] Mice were immunized with the respective antigens formulated with Freund' s complete adjuvant, followed by boosting with Freund' s incomplete adjuvant, each two weeks apart. The AMA1+RON2L group received l( g of each antigen in two separate sites (2C^g total/mouse), while the AMAl, RON2L, and AMA1/RON2L groups received lC^g antigen per animal. Two weeks after the last boost, the mice were challenged intravenously with 10 5 parasites of a lethal strain of P. yoelli (XL). The effect of the vaccination was assessed by counting daily parasitemia from Geimsa-stained blood smears (Fig. 1) and monitoring survival of infected mice (Fig. 2). Control animals injected with PBS developed parasitemia that killed all the mice by Day 5. Similarly, animals that were either immunized with the single antigens (PyAMAl or PyRON2L) or with both antigens but at different sites

(PyAMAl +PyRON2L) developed parasitemia similar to control mice and succumbed to infection by Day 5. In marked contrast, all of the animals immunized with the

PyAMAl /PyRON2L complex were able to control parasitemia and were protected. Example 2

[00146] AMA1-RON2L complex, but not AMA1, protects against lethal P. yoelli challenge.

[00147] Recombinant protein production and peptide synthesis

[00148] E. coli expression: synthetic codon optimized P. yoelii YM AMA1 (residues

59-479; PlasmoDB accession # PYYM_0916000 (SEQ ID NO: 17)) with a C-terminal histidine tag was cloned into pET24a vector. Solubilized inclusion bodies were refolded and affinity purified essentially as described (Miura, et al. 2013 Infect Immun 81(5): 1491-1501). In brief, solubilized protein was purified on a Ni Sepharose 6 FF column (GE Healthcare, NJ), followed by separation on Q Sepharose FF column (GE Healthcare, NJ) using 20 mM Tris and a NaCl gradient at pH 8.0. The EcPyAMAl eluates were pooled and polished on a S75 size-exclusion column (GE Healthcare, NJ) with a mobile phase consisting of phosphate buffered saline, pH 7.4. Purified recombinant EcPyAMAl was characterized by Coomassie blue-stained SDS-PAGE gel electrophoresis, Western blot using a protective, conformational mAb 45B1 (Narum, et al. 2000 Infect Immun 68(5):2899-2906), reversed-phase-HPLC and analytical size-exclusion column chromatography with online multi-angle light scattering- HPLC (Fig. 9), essentially as previously described (Plassmeyer, et al. 2009 J Biol Chem 284(39):26951-26963).

[00149] P. pastoris expression: recombinant his-tagged P 3D7 and P FVO AMA1 full- length ectodomain (residues 25-546) as described previously (Ellis, et al. 2012 PLoS One 7(10):e46094) were used in the studies.

[00150] Peptide synthesis: all peptides were synthesized by Lifetein LLC (New Jersey,

USA) and are listed in Figure 10. KLH conjugation to RON2L was also performed by Lifetein LLC.

[00151] Parasites and mouse infections

[00152] P. yoelii YM parasites were maintained by serial blood passage in BALB/c mice (Charles River Laboratory). For challenge studies following vaccination, the indicated numbers of infected RBCs (iRBCs) were injected intravenously and parasitemia was monitored by counting the number of iRBCs on Giemsa stained blood smears ( parasitemia = (number of iRBCs* 100)/ number of total RBCs). All experiments were performed in accordance with NIH-approved animal study protocol LMVR-1 IE.

[00153] AMA1 -RON2L complex preparation, immunization

[00154] AMA1-RON2L complex was prepared by mixing 10 μg of AMA1 with 30 μg of RON2L in 50 μΐ ^ PBS and incubating at room temperature for 30 min. The complex was emulsified in 50 μΐ ^ of Freund's adjuvant. For AMA1 alone or RON2L-KLH alone, 10 μg and 30 μg, respectively, were added to 50 μΐ ^ of PBS and emulsified in an equal volume of adjuvant. B ALB/c mice were immunized subcutaneously three times (Freund' s complete followed by two injections in Freund's incomplete adjuvant) in three week intervals. The control mice received 50 μΐ ^ of PBS emulsified in an equal volume of adjuvant. Challenge with P_ YM iRBCs were done 3 weeks after the last immunization. For injecting antigens in separate sites, 10 μg PyAMAl and 30 μg PyRON2L, respectively, in 50 μΐ ^ of PBS were emulsified separately with adjuvant and injected on opposite sides.

[00155] To test whether AMA1 or RON2L can protect against a lethal PyYM parasite challenge, animals were immunized with recombinant PyAMAl or RON2L peptide conjugated to KLH and challenged intravenously with P_ YM-infected RBCs (iRBCs). All animals succumbed to the infection similar to control animals (Fig. 3A). This lack of protection against a homologous parasite challenge in animals vaccinated with AMA1 resembles the results from controlled human trials using P. falciparum (Spring, et al. 2009 PLoS One 4(4):e5254). Strikingly however, all animals immunized with the complex were protected against the lethal parasite challenge (Fig. 3A and 3B). The amounts of anti-AMAl antibodies were similar between the groups immunized with the complex or AMA1 alone (Fig. 3C), suggesting that the differences observed between the groups were due to qualitative differences in the antibody specificity.

[00156] ELISA

[00157] A detailed description of the assay is described elsewhere (Miura, et al. 2008 J

Immunol 181(12):8776-8783). ELISA plates were coated overnight with 1 μg/mL

recombinant AMA1 or 4 μg/mL RON2L peptide. For measuring relative antibodies in immunized mice, a serial dilution of the sera was performed and the dilution that produced an OD >0.5 (in the linear detection range of the assay) was used to compare the anti-AMAl and anti-RON2L antibodies between the groups.

[00158] Antigen- specific ELISA units for P AMAl and P RON2L were measured by first generating a standard curve using serially diluted IgG mixture containing either anti- AMAl (IgG from four rats immunized with AMA1) or RON2L (IgG from four rats immunized with RON2L-KLH). Antibody units of the standards were assigned based on the reciprocal of the dilution giving an OD 405 = 1, and all samples were tested against the same standard as described (Miura, et al. 2008 J Immunol 181(12):8776-8783).

[00159] Competition ELISA was performed as described above with the addition of

0.5 μg/mL biotinylated P RON2L peptide along with the IgG dilutions (containing the indicated AMA1-EU) to measure the ability of the antibodies to inhibit RON2L binding to P 3D7AMA1. Streptavidin conjugated to alkaline phosphatase (Life technologies # S-921) was used (1:2000) to measure the amount of biotinylated RON2L bound to AMAl.

[00160] If immunity to virulent infection were due to vaccination with a functional

AMA1-RON2L complex, it would be expected that mutating the cysteine residues in

RON2L, which prevent complex formation with AMAl (Srinivasan, et al. 2011 PNAS USA 108(32): 13275-13280) (Fig. 3D and 3E), would fail to protect mice. Consistent with this, immunization with a mixture of AMAl-RON2Lc/a failed to protect mice against P_yYM (Fig. 3F and Fig. 6A).

[00161] To determine if protection is simply due to an additive effect of immunizing with two antigens or if a complex is required, animals were immunized with the AMA1- RON2L complex or the two antigens, AMAl and RON2L (AM A 1 +RON2L) , injected in two separate sites (Fig. 3G). While mice immunized with the AMA1-RON2L complex were protected, immunizing with the two antigens separately did not protect mice against Py YM (Fig. 3G and Fig. 6B). The data indicate that protection against lethal P_yYM parasites requires vaccination with a pre-formed AMA1-RON2L complex.

Example 3

[00162] AMA1-RON2 complex-induced protection is largely IgG-mediated.

[00163] Passive transfer studies

[00164] IgG from animals immunized with the P_yAMAl-RON2L complex or control

PBS as described above were purified on protein G agarose beads (GE health sciences) and dialyzed against RPMI 1640. On days -1, 0, and +1, 400 μg total IgG was injected

intravenously into recipient mice and were challenged with 105 P_ YM iRBCs on day 0. T cells from immunized animals were purified using the mouse pan T cell isolation kit

(Miltenyi # 130-095-10). All preparations used contained greater than 80% live cells as measure by counting trypan blue-stained cells. 2xl0 6 purified T cells were injected on days - 1, 0, and +1 and challenged with 105 P_ YM iRBCs on day 0. Figures 3A and 3G are two independent experiments with mice immunized with AMAl and/or RON2L separately. Data from two out of three independent experiments were performed with AMA1-RON2L complex are shown in Fig. 3A and 3G. A third experiment with AMA1-RON2L complex in Montanide ISA720 adjuvant also showed complete protection. Data from one out of three independent immunizations performed with AMAl-RON2Lc/a (that does not form a complex) is shown in Fig. 3F. [00165] For P. falciparum studies, four Sprague Dawley rats per group (Charles River

Laboratory) were immunized subcutaneously with P 3D7 full-length AMA1 (10 μg), RON2L-KLH (10 μg), or AMA1-RON2L complex (10 μg AMA1 mixed with 30 μg

RON2L), emulsified in Freund's complete, followed by two injections in Freund's incomplete adjuvant in three week intervals, as described above. IgG from sera of individual rats were purified on protein G column (GE health sciences) and dialyzed against RPMI 1640. Rat immunizations were carried out in accordance with NIH-approved animal study protocol LMVR-1.

[00166] The contribution of antibody or T cells in conferring protection was evaluated through passive transfer studies. Total IgG (400 μg) or T cells (2 x 10 6 ) from animals immunized with AMA1-RON2L complex were injected intravenously in naive mice on days -1, 0, and +1 and challenged on day 0 with P_yYM. The data suggest that the complex- dependent protection was largely antibody- mediated, as IgG, but not T cells, were able to transfer partial protection (Fig. 3H and 31 and Fig. 6C). However, the data does not rule out the possibility of a role for T cells together with antibody in mediating complete protection. Example 4

[00167] P AMA1-RON2L complex induces qualitatively better growth inhibitory antibodies.

[00168] The surprising ability of the complex to confer complete protection in mice against virulent P_yYM challenge prompted the evaluation of the potential of AMA1-RON2L complex as a blood- stage vaccine candidate for human malaria parasite P. falciparum {Pj).

[00169] P. jalciparum parasite culture

[00170] Parasites were maintained in standard in vitro cultures as described (27) with modifications as follows. Briefly, parasites were grown in RPMI 1640 supplemented with 25 mM HEPES and 50 μg ml "1 hypoxanthine (KD Medical), 0.5% Albumax (Invitrogen), 0.23% sodium bicarbonate (Gibco) using 0+ RBCs ( Interstate Blood Bank, Jackson, TN) and monitored daily by Giemsa-stained blood smears.

[00171] In the absence of an easily accessible in vivo model for human malaria, an in vitro growth inhibition activity (GIA) assay is routinely used to measure efficacy of antibodies to Pj blood- stage antigens (Kennedy, et al. 2002 Inject Immun 70(12):6948-6960).

[00172] Growth inhibition assay ( GIA )

[00173] Purified IgG at the desired concentration was dialyzed against RPMI 1640

(KD Medical) and incubated with iRBCs for 40 h. Parasitemia was quantified by biochemical measurement using a P lactate dehydrogenase assay as described previously (28). GIA reversal was performed by mixing the desired concentration of recombinant proteins with 2mg/mL pooled IgG from four rats in each group before adding to the GIA wells. All assays were performed in duplicate.

[00174] Statistical analysis

[00175] Differences in GIA responses between IgG from P/AMA1 and P/AMA1-

RON2L groups were measured using non-parametric Mann-Whitney test. Inhibition of RON2L binding to AMAl was measured by plotting a non-linear regression curve fit of the individual data points and comparing the EC 50 of the two curve fits.

[00176] GIA was used to compare the efficacies of anti-P/AMAl and anti-P/AMAl-

RON2L antibodies in blocking merozoite invasion.

[00177] IgG from rats immunized with P 3D7-allele AMA 1 -RON2L complex showed significantly higher inhibition of merozoite invasion against homologous P 3D7 parasites (Fig. 4A). This occurred despite comparable levels of antibodies to AMAl in the animals immunized with AMAl or AMA1-RON2L complex (Fig. 4B). Furthermore, antibodies to P/RON2L did not block merozoite invasion at the concentrations tested (Fig. 4C), even though the amount of RON2L- specific antibodies were higher in the RON2L-KLH group than in animals immunized with the complex (Fig. 7). Mixing anti-AMAl and RON2L IgG (AMA 1 +RON2L- KLH) did not recapitulate the increase in GIA observed with P/AMA1- RON2L complex (Fig. 4C), suggesting that RON2L- specific antibodies did not contribute significantly to GIA.

Example 5

[00178] Inhibitory antibodies induced by the complex mainly target AMAl.

[00179] The molecular basis of the qualitative difference in the antibodies induced by

AMAl and AMA1-RON2L complex was examined. Competition experiments were performed by adding recombinant P 3D7AMA1 (rAMAl) to the GIA assays. If the increase in GIA observed with IgG from the P AMA1-RON2L- immunized rats was due to antibodies targeting new epitopes formed by the complex, one would expect rAMAl not to completely reverse the GIA of IgG from AMAl-RON2L-immunized rats. Interestingly, a concentration- dependent reversal of GIA is observed when rAMAl was added to IgG from both P/AMA1- and P AMAl-RON2L-immunized rats (Fig. 4D). This suggests that inhibitory antibodies targeting AMAl still comprise a major part of the GIA of IgG induced by Ρ ΆΜΑ 1 -RON2L complex. The qualitative increase in GIA may, therefore, be due to a difference in the proportion of inhibitory antibodies in the IgG from AMAl- and AMA1-RON2L complex- immunized rats. This is supported by the observation herein that IgG from P complex- immunized animals inhibited RON2L binding to AMA1 significantly higher than IgG induced by Pf AM Al (Fig. 4E). However, the contribution of antibodies targeting new epitopes formed by the P AMA1-RON2L complex cannot be ruled out, which, in the absence of antibodies to AMA1, may not be sufficient to show significant GIA in this in vitro assay. Example 6

[00180] P AMA1-RON2L complex induces a switch in the proportion of antibodies to loops surrounding the RON2L binding site.

[00181] Homology modeling of the PyAMAl-PyRON2_D3 complex

[00182] The structural model for PyAMAl (Asn53 - Glu383; XP_729363.1 (SEQ ID

NO:4)) was generated using Modeller 9v8 through the Chimera interface (Pettersen, et al. 2004 J Comput Chem 25(13): 1605-1612; Eswar, et al. 2006 Curr Protoc Bioinformatics Chapter 5:Unit 56), based off of a hybrid model of Pf AM Al (PDB ID 3ZWZ) and PvAMAl (PDB ID 1Z40), with which it shares 52 and 56 % identity, respectively. The region of the DII loop (Lys296 - Ser332) disordered in the Pf AM Al co-structure with P/RON2_D3 and in the apo structure of PvAMAl was removed due to uncertainty in its position while in complex with P_ RON2_D3. The final model of P_ AMAl was chosen based on its low value of the normalized Discrete Optimized Protein Energy value (zDOPE).

[00183] The core 30 residues of P RON2_D3 (His2068 to Val2097; XP_727536.1

(SEQ ID NO: 10)) were modelled based on P RON2_D3 from the published co-structure with P AMAl (Vulliez-Le Normand, et al. 2012 PLoS Pathog 8(6):el002755), and initially docked into the P_ AMAl groove using ProtlnfoPPC (Kittichotirat, et al. 2009 Nucleic Acids Res 37(Web Server issue):W519-525). The P AMAl-PyRON2_D3 model was refined using Rosetta FlexPepDock (London, et al. 2011 Nucleic Acids Res 39(Web Server issue):W249- 253) with the complex showing the lowest Rosetta energy score chosen and validated by visual inspection, PISA (Krissinel, et al. 2007 J Mol Biol 372(3):774-797), ProQ (Wallner, et al. 2003 Protein Sci 12(5): 1073- 1086), ERRAT (Colovos, et al. 1993 Protein Sci 2(9): 1511- 1519), and MolProbity (Chen, et al. 2010 Acta Cry stallo gr D Biol Crystallogr 66(Ptl): 12- 21).

[00184] The hydrophobic groove in AMA1 is formed by two cysteine-rich domains

(Pizarro, et al. 2005 Science 308(5720):408-411), and binding of RON2L displaces the conserved loop (DII) in domain 2 (Vulliez-Le Normand, et al. 2012 PLoS Pathog

8(6):el002755). In addition, domain 1 loops DIb and DIf that were disordered in the apo structure (Fig. 5A) become visible in the complex structure (Fig. 5B) and a conformational change in loop Die is also observed (Fig. 5C). Previous studies have demonstrated a positive correlation between the amounts of antibodies to loop Did and in vitro GIA (Ouattara, et al. 2013 J Infect Dis 207(3):511-519), suggesting that loop regions could be targets of protective immune responses. In order to determine whether some of the conformational changes and/or stabilization of the loops upon RON2 binding may have contributed to the qualitative increase in inhibitory antibodies induced by the complex, the proportion of antibodies against synthetic biotinylated loop peptides was examined by ELISA (Fig. 5D).

[00185] Antibodies to different loop region peptides was measured by first coating individual biotinylated peptides (4 μg/mL) to streptavidin coated plates for 2 hrs at RT followed by the standard ELISA method as described previously. In order to compare the proportion of antibodies between the AMA1 and AMA1-RON2L group, each IgG sample was adjusted to have the same amount of anti-AMAl ELISA units.

[00186] Surprisingly, while the proportion of antibodies against loop Did appears to be higher in the group immunized with AMA1 alone, antibodies to loops Die and DIf appear to be higher in the group immunized with the AMA1-RON2L complex (Fig. 5D). Interestingly, the immune response against the DII loop, which undergoes the most conformational change, appears to be similar between the two groups (Fig. 5D). Did contains some of the highly polymorphic residues including E197, which has been shown to be a target of strain- specific anti-AMAl antibodies (Coley, et al. 2006 Infect Immun 74(5):2628-2636), while loops Die and DIf are less polymorphic (Fig. 8). The data suggest that a switch in the proportion of antibodies targeting these loops may contribute to the enhanced AMA1-RON2L complex vaccine efficacy. This suggestion is supported by a recent study showing that monoclonal antibodies targeting loop Die potently inhibited parasite invasion (Dutta, et al. 2013 PLoS Pathog 9(12):el003840).

Example 7

[00187] Non-human primate challenge using virulent human malaria parasite P.

falciparum.

[00188] Aotus nancymaae was used as an in vivo human malaria parasite P. falciparum model to evaluate the efficacy of AMA1-RON2L complex vaccine. Three groups of monkeys were vaccinated with wither buffer (control), AMA1 alone (40 μg per injection), or AMA1-RON2L complex (1:3 to 1:5 ratio) three times in three week intervals. Four weeks after the last vaccination, all of the monkeys were challenged intravenously with 10 5 highly virulent FVO strain of human malaria parasite P. falciparum. This parasite strain was chosen to ensure the most stringent model to test protective efficacy of the vaccine. [00189] Vaccine efficacy was analyzed by monitoring the infection status and parasite load of the animals by counting parasites in their blood every day. Animals were treated with mefloquine, an antimalarial drug, when the parasitemia reached 200,000 infected cells per microliter blood or when the hematocrit fell below 25%. Antibodies purified from vaccinated animals were also used to directly measure the ability of IgG to block parasite invasion of RBCs using an in vitro growth inhibition assay as previously described.

[00190] While an approximate range of 1:3 to 1:5 ratio is described above, a range of about 1:3 to about 1: 10 ratio (including any range within that range of ratios) is contemplated. Furthermore, any ratio resulting in vaccine efficacy is contemplated herein.

[00191] Thus, a novel approach towards developing a vaccine against the disease causing forms of the malaria parasite is disclosed herein. The lack of protection in humans immunized with AMA1 has been attributed to insufficient amounts of antibody generated using currently available adjuvants and the polymorphisms between the vaccine-type and parasite strains in the field. However, the lack of protection against a controlled, homologous parasite challenge despite the vaccine inducing high anti-AMAl titers suggests that these may not be the only reason for the failure in human trials.

[00192] Evidence is provided herein using P. yoelii and P. falciparum, two

independent host-parasite systems, that, despite similar AMA1 antibody titers, the AMA1- RON2L complex is more effective in inducing invasion-inhibitory, protective antibodies. The results indicate that the increased inhibitory activity of IgG induced by P AMA1-RON2L complex is, at least in part, due to antibodies that target new AMA1 epitopes surrounding the RON2 binding site. The fact that some of these target sites are less polymorphic bodes well for the development of an effective AMA1 -based vaccine.

[00193] Certainly, the results described herein have important implications for developing an effective blood-stage malaria vaccine. For instance, a multi-allele AMA1 (to cover polymorphisms) in complex with RON2L can be effective in protecting against both homologous and heterologous parasites.