WO2009114202A2 - Vaccine and immunization method using plasmodium antigen 2 - Google Patents

Vaccine and immunization method using plasmodium antigen 2 Download PDF

Info

Publication number
WO2009114202A2
WO2009114202A2 PCT/US2009/001643 US2009001643W WO2009114202A2 WO 2009114202 A2 WO2009114202 A2 WO 2009114202A2 US 2009001643 W US2009001643 W US 2009001643W WO 2009114202 A2 WO2009114202 A2 WO 2009114202A2
Authority
WO
WIPO (PCT)
Prior art keywords
antigen
recombinant
falciparum
dna
vaccine
Prior art date
Application number
PCT/US2009/001643
Other languages
French (fr)
Other versions
WO2009114202A3 (en
Inventor
Denise Doolan
Original Assignee
The United States Of America As Represented By The Secretary Of The Navy
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by The United States Of America As Represented By The Secretary Of The Navy filed Critical The United States Of America As Represented By The Secretary Of The Navy
Publication of WO2009114202A2 publication Critical patent/WO2009114202A2/en
Publication of WO2009114202A3 publication Critical patent/WO2009114202A3/en
Priority to US12/881,604 priority Critical patent/US20110229514A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/002Protozoa antigens
    • A61K39/015Hemosporidia antigens, e.g. Plasmodium antigens
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • Malaria is one of the most devastating parasitic diseases affecting humans. Indeed, 41% of the world's population lives in areas where malaria is transmitted (e.g., parts of Africa, Asia, the Middle East, Central and South America, Hispaniola, and Oceania). The World Health Organization (WHO) and the Centers for Disease Control (CDC) estimate that malaria infects 300-500 million people and kills 700,000-3 million people annually, with the majority of deaths occurring in children in sub-Saharan Africa. Malaria also is a major health concern to U.S. military personnel deployed to tropical regions of the world.
  • WHO World Health Organization
  • CDC Centers for Disease Control
  • Vaccines are the most cost effective and efficient therapeutic interventions for infectious diseases. In this regard, vaccination has the advantage of administration prior to military deployment and likely reduction in non-compliance risks.
  • decades of research and development directed to a malaria vaccine have not proven successful. Recent efforts have focused on developing vaccines against several specific malaria genes and delivery vector systems including adenovirus, poxvirus, and plasmids.
  • Anti-sporozoite vaccines tested include completely synthetic peptides, conjugates of synthetic peptide with proteins such as tetanus toxoid (to provide T cell help), recombinant malaria proteins, particle-forming recombinant chimeric constructs, recombinant viruses, and bacteria and DNA vaccines.
  • Several trials of asexual blood stage vaccines have used either synthetic peptide conjugates or recombinant proteins.
  • transmission blocking vaccines e.g., recombinant P/s25.
  • a recurring problem identified in all of these vaccination strategies is the difficulty in obtaining a sufficiently strong and long lasting immune response in humans, despite the strong immunogenic response in animal models.
  • the malarial parasite infects both mammalian and mosquito hosts during its complex life cycle.
  • the parasite changes hosts, it develops motile stages that can invade the target organ in the new host, heading for the site where it can proliferate to the next motile stage.
  • the parasites encounters host cell barriers that hamper its advance. To circumvent these barriers and penetrate the organ, the parasite invades and traverses host cells. Cell-traversal ability is thus essential for the parasite to establish an infection in a new host.
  • Malarial transmission to the mammalian host is accomplished by sporozoite infection of the hepatocyte, in which it forms a parasitophorous vacuole and develops into thousands of erythrocyte-invasive form.
  • Sporozoites gathered in the mosquito salivary glands are injected into the mammalian host by a mosquito bite.
  • the sporozoites enter the blood circulation and are transported to the liver sinusoid. There they leave the circulation by crossing the liver sinusoidal layer, which is the boundary between the circulation and hepatocytes.
  • motile stages of malarial parasite have no locomotory organelles, such as flagella or cilia, their motility requires substrates and is achieved by highly developed cytoplasmic secretary organelles, called micronemes.
  • a secretory microneme protein named cell -traversal protein of Plasmodium ookinetes and sporozoites (CeITOS or antigen 2, Kariu et al 2006) was found to have crucial role in the parasite invasive motility.
  • CeITOS cell -traversal protein of Plasmodium ookinetes and sporozoites
  • Targeted disruption of the CeITOS gene in Plasmodium berghei reduced parasite infectivity in the mosquito host approximately 200-fold. The disruption also reduced the sporozoite infectivity in the liver and almost abolished its cell-passage ability. Liver infectivity was restored in Kupffer cell-depleted rats, indicating that CeITOS is necessary for sporozoite passage from the circulatory system to hepatocytes through the liver sinusoidal cell layer.
  • antigen 2 (same as "CeITOS", hereafter “Antigen 2” or “Ag2”) is first identified as one of malarial vaccine candidate from the genomic sequences of complex pathogens via an antigen identification strategy, which integrates bioinformaic predictions, HLA-supertype consideration, and in vitro cellular assays. Antigen 2 has also been shown to be highly expressed in the P. falciparum sporozoite/liver proteome as evidenced by MudPIT of P. falciparum sporozoites and P. falciparum sporozoite gene transcript profiles. In immune screening studies, Ag2 is recognized by volunteers either experimentally immunized with radiation-attenuated P.
  • FIG. IA Antigen-specific reactivity in irradiated sporozoite-immunized volunteers - subject response rate.
  • FIG. IB Antigen-specific reactivity in irradiated sporozoite-immunized volunteers - test response rate.
  • FIG. 2 Antigen-specific reactivity in irradiated sporozoite-immunized volunteers protected or non-protected against sporozoite challenge.
  • FIG. 3 Alignment of amino acid sequences of P. falciparum, P. vivax, P. knowlesi and P. voelii Ag2 orthologues.
  • FIG. 4A IFN-g ELIspot of PfAg2 DNA/DNA in immunized inbred and outbred mice.
  • FIG. 4B Intracellular cytokine staining (ICS) of PfAg2 and PyAg2 DNA/DNA or
  • FIG. 5 Ag2 enzyme-linked immunosorbent assay (ELISA) against recombinant
  • Ag2 protein (A) Pre-boost sera. (B) Post-boost/ Pre-challenge sera.
  • the inventive subject matter relates to a method for inducing immune response and protective immunity in humans and animals using antigen 2 or CeITOS or a fragment thereof comprising immunizing with a priming immunization preparation that selected from a group consisting of recombinant virus expression system (such as recombinant poxvirus or recombinant adenovius); a recombinant protein antigen or a recombinant polypeptide; a synthetic peptide; a DNA vector; a whole organism or extract thereof; and a combination thereof, and subsequently immunizing with a boosting immunization preparation that is selected from a group consisting of a recombinant virus expression system such as recombinant poxvirus and recombinant adenovius; a recombinant protein antigen or a recombinant polypeptide; a synthetic peptide; a DNA vector; a whole organism or extract thereof; and a combination thereof.
  • Antigen 2 is selected from the group consisting of PfAg2 ⁇ P. falciparum), PyAg2 (P. yoelii), VvKgI (P. vivax), PkAg2 (P. knowlesi), PoAg2 (P. ovale), PmAg2 (P. malariae) PbAg2 (P. Berghei), PcAg2 (P. Chabaudi), and combination thereof.
  • the term "antigen” means an immunogenic peptide or protein which induces an immune response (see below) to a malarial pathogen capable of infecting a mammal.
  • immune response refers to the development in a subject of a humoral and/or cellular immunological response to an antigen that has been administered to the subject by the methods of this invention.
  • Human immune responses refer to the production of antibodies
  • a “cellular” immune response refers to the activation of T-lymphocytes, particularly cytolytic T-cells ("CTLs") and helper T-cells.
  • CTLs cytolytic T-cells
  • Specific T-cells involved in the cellular immune response include CD4+ and CD8+ T-cells.
  • an "immunogenic fragment” will generally include at least about 5-10 contiguous amino acid residues of the full-length molecule, preferably at least about 15- 25 contiguous amino acid residues of the full-length molecule, and most preferably at least about 20-50 or more contiguous amino acid residues of the full-length molecule, that define an epitope, or any integer between 5 amino acids and the full-length sequence, provided that the fragment in question retains immunogenic activity, as measured by an assay, such as the ones described herein.
  • Regions of a given polypeptide that include an "epitope" can be identified using any number of epitope mapping techniques, well known in the art.
  • T-cell epitopes which are involved in stimulating the cellular arm of the subject's immune system, are short peptides of 8-25 amino acids, and these are not typically predicted by the above-described methods for identifying humoral epitopes.
  • a common way to identify T-cell epitopes is to use overlapping synthetic peptides and analyze pools of these peptides, or the individual ones, that are recognized by T cells from animals that are immune to the antigen of interest, using an enzyme-linked immunospot assay (ELISPOT).
  • ELISPOT enzyme-linked immunospot assay
  • epitopes can also be used in other assays such as the stimulation of cytokine release or secretion, or by the ability to interact with major histocompatibility (MHC) tetramers.
  • MHC major histocompatibility
  • Such immunogenic fragments can also be identified based on their ability to stimulate lymphocyte proliferation in response to stimulation by various fragments from the antigen of interest.
  • epitopope refers to a sequence of at least about 3 to 5, preferably about 5 to 10 or 15, and not more than about 1 ,000 amino acids (or any integer therebetween), which define a sequence that by itself or as part of a larger sequence, binds to an antibody generated in response to such sequence or stimulates a cellular immune response.
  • an epitope for use in the subject invention is not limited to a polypeptide having the exact sequence of the portion of the parent protein from which it is derived. Indeed, there are many known species of Plasmodium and the parasite retains the ability to continue to adapt, and there are several variable domains in the parasite that exhibit relatively high degrees of variability between species. Thus the term "epitope" encompasses sequences identical to the native sequence, as well as modifications to the native sequence, such as deletions, additions and substitutions (generally conservative in nature).
  • a "priming” immunization comprising the initial administration of one or more antigens to an animal, especially a human patient, in preparation for subsequent administration(s) of the same antigen.
  • the term "priming”, as used herein defines a first immunization using an antigen which induces an immune response to the desired antigen and recalls a higher level of immune response to the desired antigen upon subsequent re-immunization with the same antigen when administered in the context of the same or a different vaccine delivery system.
  • a “priming immunization” refers to the administration of a composition comprising a preparation containing a malarial antigen.
  • a "priming immunogenic composition or preparation” refers to a preparation containing a malarial antigen or fragment thereof with the preparation being selected from the group consisting of: a recombinant virus expression system; a recombinant protein antigen or a recombinant polypeptide; a synthetic peptide; a polynucleotide vector; a whole organism or extract and combinations thereof.
  • boosting immunization or a “boost” which means the administration of a composition delivering the same malarial antigen as encoded in the priming immunization, but often utilizing a preparation with different platform, such as those selected from the group consisting of: a recombinant virus expression system; a recombinant protein antigen or a recombinant polypeptide; a synthetic peptide; a polynucleotide vector; a whole organism or extract and combinations thereof.
  • a boost is sometimes referred to as an anamnestic response, i.e. an immune response in a previously sensitized animal.
  • a boosting immunization or a boosting immunogenic composition can comprise multiple doses, which may be the same or different amounts.
  • a boosting immunization or a boosting immunogenic composition can comprise one, two, three or multiple doses.
  • the ImmunoSense strategy (Doolan et al. 2003) was designed to identify target antigens and maps T cell epitopes from large and complex genomes, such as P. falciparum, by integrating genomic and proteomic data with bioinformatic predictions, HLA supertype considerations, high-throughput binding assays, and cellular assays.
  • a central component of the strategy is the capacity of the antigen to be recognized by immune responses thought to contribute in protection, allowing for rational antigen selection and prioritization.
  • T cells recognize a complex between a specific MHC type and a particular pathogen-derived linear T cell epitope, and a given epitope will thus elicit a response only in individuals who express an MHC molecule capable of binding that epitope.
  • Human MHC molecules are extremely polymorphic, and different HLA types are expressed at dramatically different frequencies in different ethnicities.
  • HLA supertypes which are characterized by largely overlapping peptide repertoires and are expressed at high frequencies in all major ethnicities (Sette and Sidney 1999).
  • antigens were highly antigenic (recognized by at least 50% of irradiated sporozoite volunteers in at least 25% of assays) three antigens were of intermediate reactivity (recognized by at least 25% of volunteers in at least 15% of assays), and four were of low reactivity (recognized by at least 10% volunteers in at least 5% of assays). Only one protein (Ag2) was recognized by all 8 volunteers. In comparison, CSP was recognized by 3 of 8 volunteers, SSP2 by 5 of 8, LSAl by 2 of 8, and Expl by 1 of 8 volunteers (Table 1).
  • Table 1 Summary of immune reactivities against the panel of 27 putative and four known P. falciparum antigens.
  • the antigens identified as a result of these studies represent, to the best of our knowledge, the first antigens identified from P. falciparum genomic sequence data and shown to be recognized by sporozoite-induced T cell responses. Particularly noteworthy, the immune reactivity against several of the newly identified antigens greatly exceeded the reactivities observed against the well characterized antigens currently considered prime preerythrocytic stage vaccine candidate antigens, namely PfCSP, PfS SP2, PfLSAl and PfExpl. These data suggest that some of the novel antigens identified using the Immunosense screening strategy may represent better candidates for vaccine development.
  • Plasmodium falciparum antigen 2 (PfAg2) appears to be highly expressed specifically in Plasmodium sporozoites based on two large-scale expression analyses. Transcription profiling using an AFFYMETRIX GENE CHIP ® (LeRoch et al. 2003) demonstrated PfAg2 to have the 11 th highest expression level (5,585.2) overall in sporozoites and falling only behind PfCSP (28,684.1) and PfSSP2 (12,475.8) for sporozoite stage- specific genes. Secondly, MudPIT proteomic analysis found Ag2 only matching peptides present in sporozoite stages.
  • Another obstacle to vaccine development is genetic restriction of the host immune response to the CD8+ and CD4+ T cell epitopes that are the targets of T cell mediated protective immunity (Doolan and Hoffman, 1997).
  • an ideal vaccine candidate antigen would contain multiple T cell epitopes recognized in the context of defined panels of HLA supertypes to ensure that the antigen is capable of being recognized by vaccinees of diverse genetic background. Identification of peptide epitopes that bind with high affinity to multiple HLA supertypes is an integral component of the ImmunoSense strategy.
  • PfAg2 contains multiple HLA degenerate HLA- Al, HLA- A2, HLA-A3/A11, HLA-A24 and HLA-B7 degenerate T cell epitopes, and deconvolution of HLA-supertypes used for the initial identification of Ag2 (Doolan et al. 2003) has established the capacity of these epitopes to bind to multiple HLA molecules as assayed by in vitro peptide binding assays, and to be recognized by T cells derived from volunteer immunized with irradiated-sporozoites as assayed by IFN-g ELIspot (Doolan et al. 2003, DL Doolan, unpublished).
  • mice were immunized under various immunization designs.
  • mice were immunized with priming and boosting immunization preparations containing plasmid DNA encoding PfAg2 or PyAg2.
  • mice were primed with DNA encoding PfAg2 or PyAg2, and boosted with DNA containing PfAg2 or PyAg2, or boosted with a recombinant poxvirus expression system containing PfAg2 or PyAg2.
  • mice were primed with either priming preparation containing DNA encoding PfAg2, PyAg2 or PyCSP or a preparation containing recombinant adenovirus expression system containing PyAg 2, PfAg2 or PyCSP.
  • the mice were boosted with preparation containing either a recombinant adenovirus expression system or a recombinant poxvirus expression system, containing PfAg2, PyAg2 or PyCSP.
  • Splenocytes were harvested at 2 weeks post last immunization for assessment of T cell responses by IFN-g ELIspot (FIG. 4A) and multiparameter ICS (FIG. 4B).
  • Table 3 Assessment of antibody responses by IFAT against P. yoelii or P. falciparum sporozoites. (Study 1: DNA/DNA).
  • Table 4 B Assessment of antibody responses by IFAT against P. falciparum sporozoites. (Study 2: DNA/DNA and DNA/POX).
  • Table 5 A Assessment of antibody responses by IFAT against P. yoelii and P. falciparum sporozoites pre-boost. (Study 3: DN A/POX and DNA/ ADENO)
  • Table 5 B Assessment of antibody responses by IFAT against P. yoelii and P. falciparum sporozoites pre-boost. (Study 3: DNA/POX and DNA/ADENO)
  • plasmid DNA vaccine encoding Ag2 are immunogenic in mice, as demonstrated by the induction of antibodies which recognize P. falciparum sporozoites by IFAT after a single IM dose, and by the induction of T cells which recognize pools of PfAg2 synthetic peptides by IFN-g ELIspot after two or three IM immunizations. T cell responses after one immunization were not assessed. In other related studies in our laboratory, with plasmid DNA constructs encoding hypothetical P. falciparum antigens derived from the P.
  • mice were immunized via GeneGun particle mediated gene delivery (typically a better delivery method for induction of antibody responses than IM immunization), and the antisera has been analyzed for immunoreactivity against different stages of the parasite by IFAT (Aguiar et al., 2004).
  • IFAT GeneGun particle mediated gene delivery
  • a total of 22% (21/95) of Gate Way clones induced parasite-specific antibody responses, but only 3% (3/95) induced antibodies that recognized P. falciparum sporozoite stages (antisera tested after two immunizations).
  • mice were primed with either DNA encoding for PfAg2 or PyAg2 or preparation containing recombinant Ag2 adenovirus expression system and boosted with preparation containing recombinant Ag2 adenovirus expression system or recombinant Ag2 poxvirus expression system.
  • Outbred mice immunized with P. yoelii CSP were evaluated in parallel, as a comparator group. Mice were challenged at 2 weeks post last immunization with infectious P. yoelii sporozoites (100 spz for inbred strains; 200 spz for outbred CDl) and monitored for development of blood- stage parasitemia by Giemsa stained blood smears on days 5-14. Mice immunized with Antigen 2 immunization have shown significant protection against P. yoelii sporozoite, which is comparable to PyCSP.
  • Table 6A Protective efficacy of Ag2 against P. yoelii sporozoite challenge. Mice immunized with DNA and boosted with recombinant poxvirus.
  • Table 6 B Protective effecacy of Ag2 against P. yoelii sporozoite challenge. Mice immunized with DNA or recombinant adenovirus and boosted with recombinant poxvirus or recombinant adenovirus.
  • Ag2 is a novel and promising next generation vaccine candidate antigen identified by integrating bioinformatics, genomics, and molecular immunology. It is a single-exon 549 nucleotide gene (182 amino acid protein) located on P. falciparum chromosome 12. It is highly expressed in the P. falciparum sporozoite/liver proteome as evidences by MudPIT of P. falciparum sporozoites and P. falciaprum sporozoite gene transcript profiles. In immune screening studies, Ag2 is recognized by volunteers either experimentally immunized with radiation-attenuated P.
  • Moss B Genetically engineered poxviruses for recombinant gene expression, vaccination, and safety. Proc Natl Acad Sci U S A. 1996 Oct 15;93(21):11341-8.
  • RNA replicons derived from attenuated Venezuelan equine encephalitis virus protect guinea pigs and mice from Ebola hemorrhagic fever virus. Vaccine, 2000. 19(1): p. 142-53.
  • a recombinant vector derived from the host range-restricted and highly attenuated MVA strain of vaccinia virus stimulates protective immunity in mice to influenza virus.

Landscapes

  • Health & Medical Sciences (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

A vaccine comprising an immunogenic preparation containing a Plasmodium antigen or a fragment thereof, wherein said antigen is selected from a group consisting of PyAg2, PfAg2, PvAg2, PkAg2, PoAg2, PmAg2, PbAg2, PcAg2 and a combination thereof. A vaccination method comprising administering a priming immunization preparation containing a Plasmodium antigen or fragment thereof, wherein said antigen is selected from a group consisting of PyAg2, PfAg2, PvAg2, PkAg2, PoAg2, PmAg2, PbAg2, PcAg2 and a combination thereof, and administrating a boosting immunization preparation containing said Plasmodium antigen or fragment, wherein said antigen is selected from a group consisting of PyAg2, PfAg2, PvAg2, PkAg2, PoAg2, PmAg2, PbAg2, PcAg2 and a combination thereof.

Description

VACCINE AND IMMUNIZATION METHOD USING PLASMODIUM ANTIGEN 2
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims priority to provisional application number 61/036,666 filed March 14, 2008.
SEQUENCE LISTING
[0002] Incorporated by reference in its entirety herein is a paper copy of the nucleotide/amino acid sequence listing submitted concurrently herewith.
BACKGROUND OF THE INVENTION
[0003] ' Malaria is one of the most devastating parasitic diseases affecting humans. Indeed, 41% of the world's population lives in areas where malaria is transmitted (e.g., parts of Africa, Asia, the Middle East, Central and South America, Hispaniola, and Oceania). The World Health Organization (WHO) and the Centers for Disease Control (CDC) estimate that malaria infects 300-500 million people and kills 700,000-3 million people annually, with the majority of deaths occurring in children in sub-Saharan Africa. Malaria also is a major health concern to U.S. military personnel deployed to tropical regions of the world. For example, in August 2003, 28% of the 26th Marine Expeditionary Unit and Joint Task Force briefly deployed to Monrovia, Liberia, were infected with the malaria parasite Plasmodium falciparum. In addition, one 157-man Marine Expeditionary Unit sustained a 44% malaria casualty rate over a 12-day period while stationed at Robert International Airport in Monrovia. In all conflicts during the past century conducted in malaria endemic areas, malaria has been the leading cause of casualties, exceeding enemy-inflicted casualties in its impact on "person-days" lost from duty. [0004] To combat malaria during U.S. military operations, preventive drugs, insect repellants, and barriers have been used with some success, but developing drug resistance by the malaria parasite and insecticide resistance by mosquito vectors has limited the efficacy of these agents. Moreover, the logistical burden and side effects associated with the use of these agents often is associated with high non-compliance rates. Vaccines are the most cost effective and efficient therapeutic interventions for infectious diseases. In this regard, vaccination has the advantage of administration prior to military deployment and likely reduction in non-compliance risks. However, decades of research and development directed to a malaria vaccine have not proven successful. Recent efforts have focused on developing vaccines against several specific malaria genes and delivery vector systems including adenovirus, poxvirus, and plasmids. The current status of malaria vaccine development and clinical trials is reviewed in, for example, Graves and Gelband, Cochrane Database Syst. Rev., 1: CDOOO 129 (2003), Moore et al., Lancet Infect. Dis., 2: 737-743 (2002), Carvalho et al., Scand. J. Immunol., 56: 327-343 (2002), Moorthy and Hill, Br. Med. Bull, 62: 59-72 (2002), Greenwood and Alonso, Chem. Immunol, 80: 366-395 (2002), and Richie and Saul, Nature, 415: 694-701 (2002).
[0005] Over the past 15-20 years, a series of Phase 1/2 vaccine trials have been reported using synthetic peptides or recombinant proteins based on malarial antigens or gene-based approaches. Approximately 40 trials were reported as of 1998 (see Engers and Godal, Parisitology Today, 14: 56-64 (1998)). Most of these trials have been directed against the sporozoite stage or liver stage of the Plasmodium life cycle, where the use of experimental mosquito challenges allows rapid progress through Phase 1 to Phase 2a preliminary efficacy studies. Anti-sporozoite vaccines tested include completely synthetic peptides, conjugates of synthetic peptide with proteins such as tetanus toxoid (to provide T cell help), recombinant malaria proteins, particle-forming recombinant chimeric constructs, recombinant viruses, and bacteria and DNA vaccines. Several trials of asexual blood stage vaccines have used either synthetic peptide conjugates or recombinant proteins. There also have been trials of transmission blocking vaccines (e.g., recombinant P/s25). A recurring problem identified in all of these vaccination strategies is the difficulty in obtaining a sufficiently strong and long lasting immune response in humans, despite the strong immunogenic response in animal models.
[0006] The malarial parasite infects both mammalian and mosquito hosts during its complex life cycle. When the parasite changes hosts, it develops motile stages that can invade the target organ in the new host, heading for the site where it can proliferate to the next motile stage. In this migration, the parasites encounters host cell barriers that hamper its advance. To circumvent these barriers and penetrate the organ, the parasite invades and traverses host cells. Cell-traversal ability is thus essential for the parasite to establish an infection in a new host.
[0007] Malarial transmission to the mammalian host is accomplished by sporozoite infection of the hepatocyte, in which it forms a parasitophorous vacuole and develops into thousands of erythrocyte-invasive form. Sporozoites gathered in the mosquito salivary glands are injected into the mammalian host by a mosquito bite. The sporozoites enter the blood circulation and are transported to the liver sinusoid. There they leave the circulation by crossing the liver sinusoidal layer, which is the boundary between the circulation and hepatocytes. Because motile stages of malarial parasite have no locomotory organelles, such as flagella or cilia, their motility requires substrates and is achieved by highly developed cytoplasmic secretary organelles, called micronemes. A secretory microneme protein, named cell -traversal protein of Plasmodium ookinetes and sporozoites (CeITOS or antigen 2, Kariu et al 2006) was found to have crucial role in the parasite invasive motility. [0008] Targeted disruption of the CeITOS gene in Plasmodium berghei reduced parasite infectivity in the mosquito host approximately 200-fold. The disruption also reduced the sporozoite infectivity in the liver and almost abolished its cell-passage ability. Liver infectivity was restored in Kupffer cell-depleted rats, indicating that CeITOS is necessary for sporozoite passage from the circulatory system to hepatocytes through the liver sinusoidal cell layer. Electron microscopic analysis revealed that celtos-disrupted ookinetes invade the midgut epithelial cell by rupturing the cell membrane, but then fail to cross the cell, indicating that CeITOS is necessary for migration through the cytoplasm. These results suggest that conserved cell-passage mechanisms are used by both sporozoites and ookinetes to breach host cellular barriers.
[0009] In this study, antigen 2 (same as "CeITOS", hereafter "Antigen 2" or "Ag2") is first identified as one of malarial vaccine candidate from the genomic sequences of complex pathogens via an antigen identification strategy, which integrates bioinformaic predictions, HLA-supertype consideration, and in vitro cellular assays. Antigen 2 has also been shown to be highly expressed in the P. falciparum sporozoite/liver proteome as evidenced by MudPIT of P. falciparum sporozoites and P. falciparum sporozoite gene transcript profiles. In immune screening studies, Ag2 is recognized by volunteers either experimentally immunized with radiation-attenuated P. falciparum sporozoites or naturally exposed to malaria, and is preferentially recognized by irradiated sporozoite immunized volunteers who were protected against parasite challenge as compared to unprotected volunteers. Furthermore, animal data indicates that Ag2 is protective against P. yoelii challenge.
DETAILED DESCRIPTION OF THE FIGURES
[0010] FIG. IA. Antigen-specific reactivity in irradiated sporozoite-immunized volunteers - subject response rate. [0011] FIG. IB. Antigen-specific reactivity in irradiated sporozoite-immunized volunteers - test response rate.
[0012] FIG. 2. Antigen-specific reactivity in irradiated sporozoite-immunized volunteers protected or non-protected against sporozoite challenge.
[0013] FIG. 3. Alignment of amino acid sequences of P. falciparum, P. vivax, P. knowlesi and P. voelii Ag2 orthologues.
[0014] FIG. 4A. IFN-g ELIspot of PfAg2 DNA/DNA in immunized inbred and outbred mice.
[0015] FIG. 4B. Intracellular cytokine staining (ICS) of PfAg2 and PyAg2 DNA/DNA or
DNA/POX immunized inbred.
[0016] FIG. 5. Ag2 enzyme-linked immunosorbent assay (ELISA) against recombinant
Ag2 protein. (A) Pre-boost sera. (B) Post-boost/ Pre-challenge sera.
SUMMARY OF THE INVENTION
[0017] The inventive subject matter relates to a method for inducing immune response and protective immunity in humans and animals using antigen 2 or CeITOS or a fragment thereof comprising immunizing with a priming immunization preparation that selected from a group consisting of recombinant virus expression system (such as recombinant poxvirus or recombinant adenovius); a recombinant protein antigen or a recombinant polypeptide; a synthetic peptide; a DNA vector; a whole organism or extract thereof; and a combination thereof, and subsequently immunizing with a boosting immunization preparation that is selected from a group consisting of a recombinant virus expression system such as recombinant poxvirus and recombinant adenovius; a recombinant protein antigen or a recombinant polypeptide; a synthetic peptide; a DNA vector; a whole organism or extract thereof; and a combination thereof. Antigen 2 is selected from the group consisting of PfAg2 {P. falciparum), PyAg2 (P. yoelii), VvKgI (P. vivax), PkAg2 (P. knowlesi), PoAg2 (P. ovale), PmAg2 (P. malariae) PbAg2 (P. Berghei), PcAg2 (P. Chabaudi), and combination thereof.
DETAILED DESCRIPTION OF THE INVENTION Definition:
[0018] As used herein, the term "antigen" means an immunogenic peptide or protein which induces an immune response (see below) to a malarial pathogen capable of infecting a mammal.
[0019] The term "immune response" or "immunization" refer to the development in a subject of a humoral and/or cellular immunological response to an antigen that has been administered to the subject by the methods of this invention. "Humoral" immune responses refer to the production of antibodies, and a "cellular" immune response refers to the activation of T-lymphocytes, particularly cytolytic T-cells ("CTLs") and helper T-cells. Specific T-cells involved in the cellular immune response include CD4+ and CD8+ T-cells. [0020] To stimulate the humoral arm of the immune system, i.e. the production of antigen-specific antibodies, an "immunogenic fragment" will generally include at least about 5-10 contiguous amino acid residues of the full-length molecule, preferably at least about 15- 25 contiguous amino acid residues of the full-length molecule, and most preferably at least about 20-50 or more contiguous amino acid residues of the full-length molecule, that define an epitope, or any integer between 5 amino acids and the full-length sequence, provided that the fragment in question retains immunogenic activity, as measured by an assay, such as the ones described herein. [0021] Regions of a given polypeptide that include an "epitope" can be identified using any number of epitope mapping techniques, well known in the art. (See, e.g., Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66, Glenn E. Morris, Ed., 1996, Humana Press, Totowa, N. J.). Generally, T-cell epitopes which are involved in stimulating the cellular arm of the subject's immune system, are short peptides of 8-25 amino acids, and these are not typically predicted by the above-described methods for identifying humoral epitopes. A common way to identify T-cell epitopes is to use overlapping synthetic peptides and analyze pools of these peptides, or the individual ones, that are recognized by T cells from animals that are immune to the antigen of interest, using an enzyme-linked immunospot assay (ELISPOT). These overlapping peptides can also be used in other assays such as the stimulation of cytokine release or secretion, or by the ability to interact with major histocompatibility (MHC) tetramers. Such immunogenic fragments can also be identified based on their ability to stimulate lymphocyte proliferation in response to stimulation by various fragments from the antigen of interest. The term "epitope" as used herein refers to a sequence of at least about 3 to 5, preferably about 5 to 10 or 15, and not more than about 1 ,000 amino acids (or any integer therebetween), which define a sequence that by itself or as part of a larger sequence, binds to an antibody generated in response to such sequence or stimulates a cellular immune response. There is no critical upper limit to the length of the immunogenic fragment, which may comprise nearly the full-length of the protein sequence, or even a fusion protein comprising two or more epitopes from a single or multiple malarial parasite proteins. An epitope for use in the subject invention is not limited to a polypeptide having the exact sequence of the portion of the parent protein from which it is derived. Indeed, there are many known species of Plasmodium and the parasite retains the ability to continue to adapt, and there are several variable domains in the parasite that exhibit relatively high degrees of variability between species. Thus the term "epitope" encompasses sequences identical to the native sequence, as well as modifications to the native sequence, such as deletions, additions and substitutions (generally conservative in nature). [0022] One component of the methods of the present invention is the use of a "priming" immunization, comprising the initial administration of one or more antigens to an animal, especially a human patient, in preparation for subsequent administration(s) of the same antigen. Specifically, the term "priming", as used herein, defines a first immunization using an antigen which induces an immune response to the desired antigen and recalls a higher level of immune response to the desired antigen upon subsequent re-immunization with the same antigen when administered in the context of the same or a different vaccine delivery system. Specifically as used in this application, a "priming immunization" refers to the administration of a composition comprising a preparation containing a malarial antigen. As used herein, a "priming immunogenic composition or preparation" refers to a preparation containing a malarial antigen or fragment thereof with the preparation being selected from the group consisting of: a recombinant virus expression system; a recombinant protein antigen or a recombinant polypeptide; a synthetic peptide; a polynucleotide vector; a whole organism or extract and combinations thereof.
[0023] Another component of the methods and compositions of the present invention is the use of a "boosting immunization", or a "boost", which means the administration of a composition delivering the same malarial antigen as encoded in the priming immunization, but often utilizing a preparation with different platform, such as those selected from the group consisting of: a recombinant virus expression system; a recombinant protein antigen or a recombinant polypeptide; a synthetic peptide; a polynucleotide vector; a whole organism or extract and combinations thereof. A boost is sometimes referred to as an anamnestic response, i.e. an immune response in a previously sensitized animal. A boosting immunization or a boosting immunogenic composition can comprise multiple doses, which may be the same or different amounts. As used in this application, a boosting immunization or a boosting immunogenic composition can comprise one, two, three or multiple doses.
Genomic and proteomic identifications of malaria vaccine candidate [0024] The ImmunoSense strategy (Doolan et al. 2003) was designed to identify target antigens and maps T cell epitopes from large and complex genomes, such as P. falciparum, by integrating genomic and proteomic data with bioinformatic predictions, HLA supertype considerations, high-throughput binding assays, and cellular assays. A central component of the strategy is the capacity of the antigen to be recognized by immune responses thought to contribute in protection, allowing for rational antigen selection and prioritization. [0025] T cells recognize a complex between a specific MHC type and a particular pathogen-derived linear T cell epitope, and a given epitope will thus elicit a response only in individuals who express an MHC molecule capable of binding that epitope. Human MHC molecules are extremely polymorphic, and different HLA types are expressed at dramatically different frequencies in different ethnicities. One means of circumventing the problem of genetic restriction relies on the selection of epitopes restricted by HLA types that can be grouped in broad families, called HLA supertypes, which are characterized by largely overlapping peptide repertoires and are expressed at high frequencies in all major ethnicities (Sette and Sidney 1999). By targeting HLA-Al, -A2, -A3/A11, -A24, -B7 and -B44 supertypes, coverage of virtually 100% of all populations irrespective of the ethnicity of the target population can be achieved.
[0026] In proof-of-concept studies (Doolan et al. 1993), starting with 27 ORFs defined by multidimensional protein identification technology (Florens et al. 2002), 16 novel proteins were identified and reproducibly recognized by at least two of the eight irradiated sporozoite immunized volunteers tested in two or more assays, but not by any of the four mock immunized controls. Nine antigens were highly antigenic (recognized by at least 50% of irradiated sporozoite volunteers in at least 25% of assays) three antigens were of intermediate reactivity (recognized by at least 25% of volunteers in at least 15% of assays), and four were of low reactivity (recognized by at least 10% volunteers in at least 5% of assays). Only one protein (Ag2) was recognized by all 8 volunteers. In comparison, CSP was recognized by 3 of 8 volunteers, SSP2 by 5 of 8, LSAl by 2 of 8, and Expl by 1 of 8 volunteers (Table 1).
Table 1: Summary of immune reactivities against the panel of 27 putative and four known P. falciparum antigens.
Figure imgf000012_0001
[0027] More specifically, in those studies, to identify for analysis a set of ORFs representing antigens potentially expressed in the hepatic stage of the parasite life cycle, MS/MS spectra of peptide sequences generated by multidimensional protein identification technology (hereafter "MudPIT", Washburn and Yates, 2000) of P. falciparum sporozoite preparations were scanned against the P. falciparum genomic sequence database. A total of 1049 proteins were identified, selecting those ORFs with the highest number of peptide hits. A panel of 27 ORPs representative of putative P. falciparum antigens with a range of expression levels, stage-specificity, and membrane-association were selected. Frequency of recognition in the P, falciparum proteome dataset ranged between 1-16 peptide hits from six different sporozoite runs. CSP, SSP2 and Expl were identified during the proteomic analysis, by between 2 (CSP) to 24 (SSP2) peptide hits associated with between 10% (CSP) to 73% (Expl) sequence coverage. LSAl was not identified since it is expressed only during the hepatic stage and MudPIT studies were not carried out with liver-stage preparations. When searched against the final published P. falciparum database (Gardner et al. 2002) using refined gene model predictions, and taking into consideration genomic sequence information from the Anopheles (vector) (Holt et al. 2002) and human (host) (Venter et al. 2001; Lander et al. 2001) databases, 19 of the 27 antigens were independently identified in either P. falciparum cDNA libraries (P Blair and J Aguiar, unpublished) or as orthologs in P. yoelii laser capture microdissection datasets (J Sacci and J Aguiar).
[0028] Amino acid sequences from the 27 ORFs and the four known P. falciparum antigens: circumsporozoite protein (CSP), sporozoite surface protein 2 (SSP2), liver-stage antigen (LSA) and exported proteins 1 (Exp 1) were scanned with HLA supertype specific algorithms (Al, A2, A3/A11, B7 and DR supertypes) (Sidney and Sette 1999), and peptide epitopes predicted to bind with the highest affinity to each of the five HLA supertypes were identified (top 10 scorers selected from each supertype/protein combination for a maximum of 50 peptides per protein).
[0029] Control peptides from PfCSP, SSP2, LSAl and Expl predicted according to the same criteria applied to the 27 ORFs were also synthesized. Analysis revealed that the predictive algorithms were highly effective in identifying the majority (approximately 81%) of previously validated epitopes from those antigens. Peptides representing predicted epitopes were synthesized and then tested by IFN-γ ELIspot for their capacity to induce recall IFN-γ immune responses using PBMC from volunteers immunized with irradiated P. falciparum sporozoites (n=4), or mock immunized control volunteers (n=4). Peptides were tested as pools, at 1 ug/ml each peptide, with each antigen represented by a separate pool, Positive and negative control epitopes from well characterized antigens (CMV, Influenza, EBV, HIV) were also included. A total of 16 of the 27 previously untested antigens were reproducibly recognized as antigens by at least two of eight irradiated sporozoite immunized volunteers tested in two or more assays, but not by any of the four mock immunized controls (Doolan et al. 2003) (Table 1, FIG. IA & IB).
[0030] Nine of the 27 antigens were recognized by at least 50% of irradiated sporozoite volunteers in at least 25% of assays and classified as highly antigenic. These antigens could be prioritized for vaccine development accordingly to their immune reactivity (frequency and magnitude of response). Three antigens were recognized by at least 25% of volunteers in at least 15% of assays and classified as intermediately antigenic. Four antigens were recognized by two volunteers and classified as weakly antigenic. Finally, 11 of the 27 untested antigens failed to induce reproducible IFN-γ responses of sufficient magnitude to meet our criteria of positivity. Positive control peptides derived from CMV, FLU and EBV were recognized by 87.5% of the volunteers tested. Pools of predicted epitopes from the known antigens, PfCSP, PfSSP2, PfLSAl and PfExpl, were recognized by about one third of the irradiated sporozoite volunteers.
[0031] The antigens identified as a result of these studies represent, to the best of our knowledge, the first antigens identified from P. falciparum genomic sequence data and shown to be recognized by sporozoite-induced T cell responses. Particularly noteworthy, the immune reactivity against several of the newly identified antigens greatly exceeded the reactivities observed against the well characterized antigens currently considered prime preerythrocytic stage vaccine candidate antigens, namely PfCSP, PfS SP2, PfLSAl and PfExpl. These data suggest that some of the novel antigens identified using the Immunosense screening strategy may represent better candidates for vaccine development. [0032] The most antigenic protein amongst those identified by the ImmunoSense approach was a single-exon 549-nucleotide gene (182 amino acid protein) located on P. falciparum chromosome 12 (PFL0800c), and was designated "Ag2". In immune T cell screening studies (Doolan et al. 2003), Ag2 was reproducibly recognized in the context of multiple genetic restrictions by 100% (8/8) volunteers experimentally immunized with radiation-attenuated P. falciparum sporozoites, but not by any mock-immunized control volunteers, in more than 50% of assays (Table 1).
[0033] Moreover, it appeared to be preferentially recognized by irradiated sporozoite- immunized voluteers who were protected against parasite challenge as compared to unprotected volunteers (FIG. 2). Interestingly, responses to the previously characterized antigens (CSP, etc) have been detected predominantly in nonprotected volunteers. In all in vitro studies to date, immune responses to Ag2 have been markedly better than responses to PfExpl, PfLSAl, PfCSP or PfSSP2, as evidenced by the frequency of responders (eg., 8/8 vs. 1/8, 2/8, 3/8 and 5l%; p = 0.001, 0.007, and 0.026), and magnitude of response (122.7 vs 41.3, 34.2, 41.6, 97.2) (Doolan et al. 2003) (Table 1). The relative immunodominance of Ag2 as compared to the historic antigens has been consistently demonstrated in subsequent studies. [0034] In independent studies, seven of the 27 ORFs evaluated in the ImmunoSense approach were assayed in a humoral immunoscreening assay designed to evaluate whether putative antigens could be recognized by malaria-immune sera. Of those, 4/4 antigens classified with high immune reactivity and 1/1 antigens classified with intermediate immune reactivity were shown to be recognized by malaria-immune sera but not by malaria-naϊve sera, in as many as 5/8 independent experiments (Regis et al. mns submitted). For Ag2, responses that met criteria of positivity were detected in 2 of 3 independent experiments. Expression analysis ofΛg2
[0035] Plasmodium falciparum antigen 2 (PfAg2) appears to be highly expressed specifically in Plasmodium sporozoites based on two large-scale expression analyses. Transcription profiling using an AFFYMETRIX GENE CHIP® (LeRoch et al. 2003) demonstrated PfAg2 to have the 11th highest expression level (5,585.2) overall in sporozoites and falling only behind PfCSP (28,684.1) and PfSSP2 (12,475.8) for sporozoite stage- specific genes. Secondly, MudPIT proteomic analysis found Ag2 only matching peptides present in sporozoite stages. Moreover, peptides identified in sporozoites were numerous (SeqCount = 17; SpecCount = 56) and achieved high sequence coverage (49.5%). These data relatively translate to PfAg2 being in the top four most abundant sporozoite specific antigens via MudPIT analysis. Only PfCSP, PfSSP2, and PFL0945w (hypothetical protein) possessed higher SpecCounts for sporozoite specific antigens.
Identification oforthologs ofPfAg2 in other Plasmodium spp
[0036] No ortholog of Pf Ag2 could be identified in the annotated P. yoelii genome (Carlton et al. 2002). However, upon scanning the corresponding region along chromosome 12 common stretch of sequence and gene homology was achieved between P. yoelii and P. falciparum. It was noticed that genes flanking Ag2 in the P. falciparum chromosome 12 genome all had orthologs present in P. yoelii. After obtaining the corresponding P. yoelii contig (chrPyl 00049) and scanning for ORFs a previously uncharacterized ORF was identified that shares size (single exon; predicted 185 amino acids for P. yoelii, 182 amino acids for P. falciparum) and sequence identify (44% identity at the amino acid level; 57% identify at the nucleotide level). Subsequently, PfAg2 orthologs were determined in corresponding contigs in P. vivax (185 amino acids; 42% identity at the amino acid level; 54% identify at the nucleotide level) and P. knowlesi (185 amino acids; 43% identity at the amino acid level; 56% identify at the nucleotide level), using BLAST searches against the incomplete sequence databases (Table 2). Table 2; Amino acid and nucleotide sequence identity
Figure imgf000017_0002
Figure imgf000017_0001
Sequence conservation ofAg2 relative to well characterized P. falciparum antigens [0037] The level of sequence identity between the identified Plasmodium spp orthologs of Ag2 is greater than that observed between PyHEP 17 and PfExp-1 (37% identity at the amino acid level), P. falciparum and P. yoelii circumsporozoite protein (26%) (Dame et al. 1984; LaI et al. 1987); P. falciparum and P. knowlesi circumsporozoite protein (25%); and P. falciparum and P. yoelii sporozoite surface protein (28%) (Robson et al. 1999, 1990; Rogers et al. 1992a, b); and similar to that between the P. falciparum and P. knowlesi sporozoite surface protein 2 (36%). There are no known P. knowlesi or P. vivax orthologs of PyHEP 17/PfExp-l, and no known P. yoelii, P. knowlesi or P. vivax orthologs of PfLSAl, so sequence identity between species cannot be assessed in those cases. The level of identity between Plasmodium orthologs of Ag2 is also greater than for leading blood-stage candidate antigens, merozoite surface protein- 1 (MAD20, 29%; Wellcome, 30%) and rhoptry antigen (overall identity 4%; best identity 11%). (FIG. 3) [0038] Analysis of the sequence of Ag2 from a number of different P. falciparum strains and isolates revealed a high degree of conservation between P. falciparum strains, and showed that the polymorphism was restricted to a limited number of amino acid variants.
Recognition ofΛg2 in the context of multiple genetic restrictions
[0039] Parasite polymorphism of identified targets of CD 8+ and CD4+ T cell mediated protective immunity poses a major obstacle to the development of an effective vaccine, since such polymorphism provides a means for the parasite to evade the host immune response (Doolan and Hoffman, 1997). Thus, an ideal vaccine candidate antigen would be highly conserved between Plasmodium strains and species. The high level of sequence identity noted between P. falciparum strains and between the P. falciparum, P. vivax, P. knowlesi, and P. yoelii orthologs of Ag2 (FIG. 3) support the potential of Ag2 as a good candidate antigen for vaccine development.
[0040] Another obstacle to vaccine development is genetic restriction of the host immune response to the CD8+ and CD4+ T cell epitopes that are the targets of T cell mediated protective immunity (Doolan and Hoffman, 1997). Thus, in addition to being conserved between species, an ideal vaccine candidate antigen would contain multiple T cell epitopes recognized in the context of defined panels of HLA supertypes to ensure that the antigen is capable of being recognized by vaccinees of diverse genetic background. Identification of peptide epitopes that bind with high affinity to multiple HLA supertypes is an integral component of the ImmunoSense strategy. PfAg2 contains multiple HLA degenerate HLA- Al, HLA- A2, HLA-A3/A11, HLA-A24 and HLA-B7 degenerate T cell epitopes, and deconvolution of HLA-supertypes used for the initial identification of Ag2 (Doolan et al. 2003) has established the capacity of these epitopes to bind to multiple HLA molecules as assayed by in vitro peptide binding assays, and to be recognized by T cells derived from volunteer immunized with irradiated-sporozoites as assayed by IFN-g ELIspot (Doolan et al. 2003, DL Doolan, unpublished).
[0041] Additionally, sera from 20/20 genetically diverse individuals resident in a hyperendemic region of Kenya recognized PfAg2 by western blot analysis of cells transfected with plasmid DNA encoding PfAg2 (data not presented), demonstrating the unrestricted nature of the antibody response to Ag2. Chicken embryo cells infected with recombinant PfAg2 poxvirus or 293 cells infected with recombinant PfAg2 adenovirus were also recognized by sera from malaria-exposed individuals (data not presented). [0042] Studies assessing the immunogenicity of PfAg2 and PyAg2 in vivo support the contention that Ag2 is recognized in the context of multiple genetic elements. In the studies, BALB/C (H-2d), C57BL/6 (H-2b), Bl 0.BR (H-2k), A/J (H-2a) and CDl (outbred) mice were immunized under various immunization designs. In study 1, mice were immunized with priming and boosting immunization preparations containing plasmid DNA encoding PfAg2 or PyAg2. In study 2, mice were primed with DNA encoding PfAg2 or PyAg2, and boosted with DNA containing PfAg2 or PyAg2, or boosted with a recombinant poxvirus expression system containing PfAg2 or PyAg2. In study 3, mice were primed with either priming preparation containing DNA encoding PfAg2, PyAg2 or PyCSP or a preparation containing recombinant adenovirus expression system containing PyAg 2, PfAg2 or PyCSP. The mice were boosted with preparation containing either a recombinant adenovirus expression system or a recombinant poxvirus expression system, containing PfAg2, PyAg2 or PyCSP. Splenocytes were harvested at 2 weeks post last immunization for assessment of T cell responses by IFN-g ELIspot (FIG. 4A) and multiparameter ICS (FIG. 4B). [0043] Sera was harvested at 2-4 weeks post each immunization for assessment of antibody responses by IFAT against P. yoelii or P. falciparum sporozoites. Table 3 shows antibody responses by IFAT against P. yoelii or P. falciparum sporozoites for study 1. Tables 4A & 4B shows antibody responses by IFAT against P. yoelii or P. falciparum sporozoites for study 2, in which sera post last immunization were pooled and assayed in triplicate. IFAT data for Study 3 are shown in Tables 5A & 5B. ELISA against recombinant PfAg2 protein are shown in FIG. 5 A & 5B, in which pooled sera post last immunization were assayed in quadruplicate. Other studies have established that PfAg2 antisera does not recognize blood-stage parasites (liver stage IFAT not done).
Table 3: Assessment of antibody responses by IFAT against P. yoelii or P. falciparum sporozoites. (Study 1: DNA/DNA).
CD-1 A/J C57BL/6 BALEVc B10.BR
1 40 40 80 40 0
2 160 80 40 80 0
3 320 320 80 40 0
4 160 80 20 40 0
5 40 40 80 160 0
6 320 80 320 80 0
7 640 80 1260 80 0
8 20 80 40 40 0
9 40 40 40 0 0
10 0 40 160 0 0
GeoMeaπ 109 70 92 62 0
Range 0-640 40-320 40-1280 0-160 0
Table 4 A: Assessment of antibody responses by IFAT against P. yoelii sporozoites. (Study 2: DNA/DNA and DN A/POX)
Figure imgf000021_0001
Table 4 B: Assessment of antibody responses by IFAT against P. falciparum sporozoites. (Study 2: DNA/DNA and DNA/POX).
Figure imgf000021_0002
Table 5 A: Assessment of antibody responses by IFAT against P. yoelii and P. falciparum sporozoites pre-boost. (Study 3: DN A/POX and DNA/ ADENO)
P. yoelii Ag2 & P. falciparum Ag2 Pre-Boost
GROUP IMMUNOGENS GEOMEAN RANGE
1 PyAg2 DNATPOX 120 20-320
2 PyAg2 DNA/ADENO 23 20-40
3 PyAg2ADENO/POX 35 20-160
4 PfAg2 DNA/POX) 67 20-320
5 Pf Ag2 DNA/ADENO 48 20-160
6 Pf Ag2 ADENO/POX 31 20-40
7 PyCSP DNA/POX 80 20-320
8 PyCSP DNA/ADENO 36 20-160
. 9 PyCSP ADENO/POX) 40 20-160
Figure imgf000022_0001
PyAg2 DNA prcboost 54 20-320
PfAg2 DNA preboost 57 20-320
PyCSP DNA preboost 53 20-320
Table 5 B: Assessment of antibody responses by IFAT against P. yoelii and P. falciparum sporozoites pre-boost. (Study 3: DNA/POX and DNA/ADENO)
P. yoelii Ag2 & P. falciparum Ag2 Pre-Challenge (Post-Boost)
GROUP IMMUNOGENS GEOMEAN RANGE
1 Py Ag2DNA/POX 214 80-320
2 Py Ag2DNA/ADENO 135 40-320
3 Py Ag2 ADENO/POX 254 160-320
4 PfAg2 DNA/POX 135 80-320
5 Pf Ag2 DNA/ADENO 127 80-640
6 PfAg2 ADENO/POX 285 80-640
7 PyCSP DNA/POX 269 20-20480
8 PyCSP DNA/ADENO 320 20-20480
9 PyCSP ADENO/POX 285 20-2560
[0044] Thus, we have established that plasmid DNA vaccine encoding Ag2 are immunogenic in mice, as demonstrated by the induction of antibodies which recognize P. falciparum sporozoites by IFAT after a single IM dose, and by the induction of T cells which recognize pools of PfAg2 synthetic peptides by IFN-g ELIspot after two or three IM immunizations. T cell responses after one immunization were not assessed. In other related studies in our laboratory, with plasmid DNA constructs encoding hypothetical P. falciparum antigens derived from the P. falciparum genome and generated using the Gate Way recombinatorial cloning system, mice were immunized via GeneGun particle mediated gene delivery (typically a better delivery method for induction of antibody responses than IM immunization), and the antisera has been analyzed for immunoreactivity against different stages of the parasite by IFAT (Aguiar et al., 2004). In those studies, a total of 22% (21/95) of Gate Way clones induced parasite-specific antibody responses, but only 3% (3/95) induced antibodies that recognized P. falciparum sporozoite stages (antisera tested after two immunizations). Those genes were not selected based on stage-specific expression and it is possible that only a small percentage may be expressed on the sporozoite. Nonetheless, the capacity of plasmid DNA encoding Ag2 to induce anti-sporozoite antibodies after a single IM immunization attests to the proposed immunogenicity of Ag2 in vivo, consistent with its demonstrated antigenicity in vitro.
Protective efficacy ofAg2
[0045] The capacity of Ag2 to protect against P. yoelii sporozoite challenge was determined in inbred and outbred mice. BALB/C (H-2d), C57BL/6 (H-2b), Bl O.BR (H-2k), A/J (H-2a) and CDl (outbred) mice were primed with plasmid DNA encoding PfAg2 or PyAg2, and boosted with recombinant Ag2 poxvirus (table 6A). Outbred mice were primed with either DNA encoding for PfAg2 or PyAg2 or preparation containing recombinant Ag2 adenovirus expression system and boosted with preparation containing recombinant Ag2 adenovirus expression system or recombinant Ag2 poxvirus expression system. Outbred mice immunized with P. yoelii CSP were evaluated in parallel, as a comparator group. Mice were challenged at 2 weeks post last immunization with infectious P. yoelii sporozoites (100 spz for inbred strains; 200 spz for outbred CDl) and monitored for development of blood- stage parasitemia by Giemsa stained blood smears on days 5-14. Mice immunized with Antigen 2 immunization have shown significant protection against P. yoelii sporozoite, which is comparable to PyCSP.
Table 6A: Protective efficacy of Ag2 against P. yoelii sporozoite challenge. Mice immunized with DNA and boosted with recombinant poxvirus.
P. yoelii Ag 2 DNA/POX P. falciparum Ag 2 DNA/POX
Strain H-2
#ιnfect/#challenge % protection #ιnfect/#challenge % protection
BALB/c H-2d 0/9 0 1/9 11.1
C57BL/6 H-2b 1/9 11.1 0/9 0
A/J H-2a 1/9 11.1 0/9 0
B10 BR H-2k 1/9 11.1 1/9 11.1
CD-1 outbred 4/9 44.4 5/9 55.6
Table 6 B: Protective effecacy of Ag2 against P. yoelii sporozoite challenge. Mice immunized with DNA or recombinant adenovirus and boosted with recombinant poxvirus or recombinant adenovirus.
0 wks 8 wks Sterile Pr otection
Group Strain Prime Boost # protect % protect
1 CD-1 P yoelii Ag2 DNA P yoelii Ag2 POX 6/14 43%
2 CD-1 P yoelii Ag2 DNA P yoelii Ag2 Adeno 7/14 50%
3 CD-1 P yoelii Ag2 adeno P yoelii Ag2 POX 9/14 64%
4 CD-1 P falciparum Ag2 DNA P falciparum Ag2 POX 5/14 36%
5 CD-1 P falciparum Ag2 DNA P falciparum Ag2 Adeno 8/14 57%
6 CD-1 P falciparum Ag2 adeno P falciparum Ag2 POX 10/14 71%
7 CD-1 P yoelii CSP DNA P yoelii CSP POX 7/14 50%
8 CD-1 P yoelii CSP DNA P yoelii CSP Adeno 8/14 57%
9 CD-1 P yoelii CSP adeno P yoelii CSP POX 8/14 57%
10 CD-1 Infectivity control - 200 spz - 0/14 0%
11 CD-1 Infectivity control - 50 spz) - 0/7 0%
12 CD-1 Infectivity control - 10 spz) - 1/7 14%
13 CD-1 Infectivity control - 2 spz) - 2/7 29%
14 CD-1 Infectivity control - 0 4 spz) - 3/7 43%
DNA dose = 100 ug route = IM
POX dose = 1E7, route = IM
Adeno dose = 1E8, route = IM
Spz chl = 200 spz Conclusion - The potential ofΛg2for vaccine development
[0046] In summary, Ag2 is a novel and promising next generation vaccine candidate antigen identified by integrating bioinformatics, genomics, and molecular immunology. It is a single-exon 549 nucleotide gene (182 amino acid protein) located on P. falciparum chromosome 12. It is highly expressed in the P. falciparum sporozoite/liver proteome as evidences by MudPIT of P. falciparum sporozoites and P. falciaprum sporozoite gene transcript profiles. In immune screening studies, Ag2 is recognized by volunteers either experimentally immunized with radiation-attenuated P. falciparum sporozoites or naturally exposed to malaria, and is preferentially recognized by irradiated sporozoite immunized volunteers who were protected against parasite challenge as compared to unprotected volunteers. Significantly, Ag2 has proved to be more immunogenic than all well- characterized P. falciparum antigens currently in clinical evaluation. Overall, an abundance of data implicates Ag2 as an extremely promising candidate antigen for malaria vaccine development. Table 7: Nucleotide and Amino Acid Sequences for Ag 2 Orthologs
Figure imgf000025_0001
REFERENCES
Aguiar, J. C. et al., (2004)." High throughput generation of P. falciparum functional molecules by recombinational cloning" Genome Research. In Press.
Allen, T. M., et al. (2000). "Induction of AIDS virus-specific CTL activity in fresh, unstimulated peripheral blood lymphocytes from rhesus macaques vaccinated with a DNA prime/modified vaccinia virus Ankara boost regimen." J Immunol 164(9): 4968-78.
Amara, R. R., et al. (2001) Control of a mucosal challenge and prevention of AIDS by a multiprotein DNA/MVA vaccine. Science. 292(5514):69-74.
Barouch, D. H., et al. (2001a). Elicitation of high-frequency cytotoxic T-lymphocyte responses against both dominant and subdominant simian-human immunodeficiency virus epitopes by DNA vaccination of rhesus monkeys. J Virol. 2001 Mar;75(5):2462-7.
Barouch, D. H., et al. (2001b). Reduction of simian-human immunodeficiency virus 89.6P viremia in rhesus monkeys by recombinant modified vaccinia virus Ankara vaccination. J Virol. 75(11):5151-8.
Beadle, C. and S. L. Hoffman (1993). "History of malaria in the United States Naval Forces at war: World War I through the Vietnam conflict." Clin.Infect.Dis. 16: 320-329.
Bender, B. S., Rowe, C. A., Taylor, S. F., Wyatt, L.S ., Moss B., and Small P. A. Jr. (1996). Oral immunization with a replication-deficient recombinant vaccinia virus protects mice against influenza. J Virol. 70(9):6418-24.
Berglund, P., et al. (1998). "Enhancing immune responses using suicidal DNA vaccines." Nat Biotechnol 16(6): 562-5.
Berglund, P., et al. (1999). "Immunization with recombinant Semliki Forest virus induces protection against influenza challenge in mice." Vaccine 17(5): 497-507.
Carlton, J. M., et al. (2002). "Genome sequence and comparative analysis of the model rodent malaria parasite Plasmodium yoelii yoelii." Nature 419: 520-526.
Dame, J. B., et al. (1984). "Structure of the gene encoding the immunodominant surface antigen on the sporozoite of the human malaria parasite Plasmodium falciparum." Science 225: 593-599.
Doolan, D. L., et al. (1996a). "Characterization of the protective Hepatocyte Erythrocyte Protein 17 kDa gene of Plasmodium yoelii: homo log of P. falciparum Exported Protein- 1." J. Biol. Chem. 271 : 17861-17868.
Doolan, D. L., et al. (1996b). "Circumventing genetic restriction of protection against malaria with multi-gene DNA immunization: CD8+ T cell, interferon- D, nitric oxide dependent immunity." J. Exp. Med. 183: 1739-1746. Doolan, D. L. and S. L. Hoffman (1997). "Multi-gene vaccination against malaria: a multistage, multi-immune response approach." Parasitol. Today 13: 171-178.
Doolan, D. L. and S. L. Hoffman (1999). "IL-12 and NK cells are required for antigen- specific adaptive immunity against malaria initiated by CD8+ T cells in the Plasmodium yoelii model." J Immunol 163: 884-892.
Good, M. F. and D. L. Doolan (1999). "Immune effector mechanisms in malaria." Curr Opin Immunol 1 1 : 412-419.
Doolan, D. L. and S. L. Hoffman (2000). "The complexity of protective immunity against liver- stage malaria." J Immunol 165: 1453-1462.
Doolan, D. L. and S. L. Hoffman (2001). "DNA-based vaccines against malaria: status and promise of the Multi-Stage Malaria DNA Vaccine Operation." Int J Parasitol 31(8): 753-62.
Doolan, D. L., et al. (2003). "Identification of new P. falciparum antigens by antigenic analysis of genomic and proteomic data." Proc Natl Acad Sci U S A 100(17): 9952-9957.
Eloit, M., et al., Construction of a defective adenovirus vector expressing the pseudorabies virus glycoprotein gp50 and its use as a live vaccine. J Gen Virol, 1990. 71(Pt 10): p. 2425- 31.
Epstein, J. E., et al. (2002). "Safety, Tolerability, and Lack of Antibody Responses After Administration of a PfCSP DNA Malaria Vaccine via Needle or Needle-Free Jet Injection, and Comparison of Intramuscular and Combination Intramuscular/Intradermal Routes." Hum Gene Ther 13(13): 1551-60.
Florens, L., M. P. Washburn, et al. (2002). "A proteomic view of the Plasmodium falciparum life cycle." Nature 419: 520-526.
Gardner, M. J., N. Hall, et al. (2002). "Genome sequence of the human malaria parasite Plasmodium falciparum." Nature 419(6906): 498-511.
Gardner, M. J., et al. (2002). "Genome sequence of the human malaria parasite Plasmodium falciparum." Nature 419(6906): 498-511.
Hevey, M., et al., Marburg virus vaccines based upon alphavirus replicons protect guinea pigs and nonhuman primates. Virology, 1998. 251(1): p. 28-37.
Hirsch, V.M., Fuerst, T.R., Sutter, G., Carroll, M.W., Yang, L., Goldstein, S., Piatak, M., Elkins, W.R., Alvord, W.G., Montefiori, D.C., Moss, B., and Lifson, J.D. (1996). Patterns of viral replication correlate with outcome in simian immunodeficiency virus (SΙV)-infected macaques: effect of prior immunization with a trivalent SIV vaccine in modified vaccinia virus Ankara. J. Virol. 70: 3741-3752.
Hoffman, S. L, et al. (2002). "Protection of humans against malaria by immunization with radiation-attenuated Plasmodium falciparum sporozoites." J Infect Dis 185(8): 1155-64.
Hoffman, S. L. and Doolan, D. L. (2000). "Malaria Vaccines - Targeting the infected hepatocyte." Nature Med. 6: 1218-1219.
Holt, R. A., G. M. Subramanian, et al. (2002). "The genome sequence of the malaria mosquito Anopheles gambiae." Science 298(5591): 129-49.
Kariu, T., Ishino, T., Yano, K., Chinzei, Y, and Yuda, M. (2006). "CeITOS, a novel malarial protein that mediates transmission to mosquito and vertebrate hosts." Molecular Micro. 59(5): 1369-1379.
Klonjkowski B, et al. (1999). Adenoviral vectors for vaccines., In Adenoviruses: Basic biology to gene therapy, S. P, Editor. RG Landers Company: Austin, p. 163-173.
Kumar, S., et al. (2002). "A multilateral effort to develop DNA vaccines against falciparum malaria." Trends Parasitol 18(3): 129-35.
LaI, A. A., et al. (1987). "Structure of the gene encoding the circumsporozoite protein of Plasmodium yoeli." J.Biol.Chem. 262: 2937-2940.
Lander, E. S., et al. (2001). "Initial sequencing and analysis of the human genome." Nature 409(6822): 860-921.
Lasonder, E., et al. (2002). "Analysis of the Plasmodium falciparum proteome by high- accuracy mass spectrometry." Nature 419(6906): 537-42.
Le Roch, K. G., et al. (2003). "Discovery of gene function by expression profiling of the malaria parasite life cycle." Science 301(5639): 1503-8.
Le, T. P., et al. (2000). "Safety, tolerability and humoral immune responses after intramuscular administration of a malaria DNA vaccine to healthy adult volunteers." Vaccine 18(18): 1893-1901.
Leitner, W., et al. (2000). Enhancement of tumor-specific immune response with plasmid DNA replicon vectors. Cancer Res. 60:51.
Martin, T., et al. (1999). "Plasmid DNA malaria vaccine: the potential for genomic integration following intramuscular injection." Human Gene Ther 10: 759-768.
Mayr, G.A., J. Chinsangaram, and M.J. Grubman. (1999). Development of replication- defective adenovirus serotype 5 containing the capsid and 3 C protease coding regions of foot- and-mouth disease virus as a vaccine candidate. Virology. 263(2): p. 496-506.
Moss B. Genetically engineered poxviruses for recombinant gene expression, vaccination, and safety. Proc Natl Acad Sci U S A. 1996 Oct 15;93(21):11341-8.
Moss, B., Carroll, M.W., Wyatt, L., Bennink, J.R., Hirsch, V.M., Goldstein, S., Elkins, W., Fuerst, T., Lifson, J., Piatak, M., Restifo, N., Overwijk, W., Chamberlain, R., Rosenberg, S. A., and Sutter, G. (1996). Host range restricted, non-replicating vaccinia virus vectors as vaccine candidates. Adv. Exp. Med. Biol. 397: 7-13.
Nardin, E., F. Zavala, et al. (1999). "Pre-erythrocytic malaria vaccine: mechanisms of protective immunity and human vaccine trials." Parassitologia 41(1-3): 397-402.
Ourmanov, I., Brown, C. R., Moss, B., Carroll, M., Wyatt, L., Pletneva, L., Goldstein, S., Venzon, D., and Vm., H. (2000). Comparative efficacy of recombinant modified vaccinia virus Ankara expressing simian immunodeficiency virus (SIV) Gag-Pol and/or Env in macaques challenged with pathogenic SIV. J. Virol. Methods 74: 2740-2751.
Parker, S. E., F. Borellini, et al. (1999). "Plasmid DNA malaria vaccine: tissue distribution and safety studies in mice and rabbits." Hum. Gene Ther. 10: 741-758.
Parker, S. E., D. Monteith, et al. (2001). "Safety of a GM-CSF adjuvant-plasmid DNA malaria vaccine." Gene Ther 8(13): 101 1-23.
Plebanski, M. and A. V. Hill (2000). "The immunology of malaria infection." Curr Opin Immunol 12(4): 437-41.
Polo et al. (1999). Stable alphavirus packaging cell lines for Sindbis virus and Semliki Forest virus-derived vectors. Proc.Natl.Acad.Sci.USA 96:4598.
Pushko, P., et al. (2000). Recombinant RNA replicons derived from attenuated Venezuelan equine encephalitis virus protect guinea pigs and mice from Ebola hemorrhagic fever virus. Vaccine, 2000. 19(1): p. 142-53.
Pushko, P., et al., Replicon-helper systems from attenuated Venezuelan equine encephalitis virus: expression of heterologous genes in vitro and immunization against heterologous pathogens in vivo. Virology, 1997. 239(2): p. 389-401.
Richie, T. L. and A. Saul (2002). "Progress and challenges for malaria vaccines." Nature 415(6872): 694-701.
Robson, K. J., et al. (1988). "A highly conserved amino-acid sequence in thrombospondin, properdin and in proteins from sporozoites and blood stages of a human malaria parasite." Nature 335: 79-82.
Robson, K. J., et al. (1990). "Polymorphism of the TRAP gene of Plasmodium falciparum." Proc. R. Soc. Lond. B. Biol. Sci. 242: 205-216.
Rogers, W. O., et al. (1992). "Characterization of Plasmodium falciparum sporozoite surface protein 2." Proc. Natl. Acad. Sci. USA 89: 9176-9180.
Rogers, W. O., et al. (1992). "Characterization of the gene encoding sporozoite surface protein 2, a protective Plasmodium yoelii sporozoite antigen." MoI. Biochem. Parasitol. 53:
45-52.
Sacci, J.B., Aguiar, J. C, Lau, A.O., Hoffman, S.L., (2002) "
Schneider, J., Gilbert, S.C., Hannan, CM., Degano, P., Prieur, E., Sheu, E.G., Plebanski, M., and Hill, A. V. (1999). Induction of CD8+ T cells using heterologous prime-boost immunisation strategies. Immunol. Rev. 170: 29-38.
Sedegah, M., et al. (2002). "Persistence of protective immunity to malaria induced by DNA priming and poxvirus boosting: characterization of effector and memory CD8(+)-T-cell populations." Infect Immun 70(7): 3493-9.
Sedegah, M., et al. (1994). "Protection against malaria by immunization with plasmid DNA encoding circumsporozoite protein." Proc. Natl. Acad. Sci. USA 91 : 9866-9870.
Sedegah, M., et al. (1998). "Boosting with recombinant vaccinia increases immunogenicity and protective efficacy of malaria DNA vaccine." Proc Natl Acad Sci U S A 95(13): 7648-53.
Sedegah, M., et al. (2000). "Improving protective immunity induced by DNA-based immunization: priming with antigen and GM-CSF-encoding plasmid DNA and boosting with antigen-expressing recombinant poxvirus." J Immunol 164(11): 5905-12.
Sette, A. and J. Sidney (1999). "Nine major HLA class I supertypes account for the vast preponderance of HLA-A and — B polymorphism." Immunogenetics 50: 201-212.
Shiver, J. W., et al. (2002). Replication-incompetent adenoviral vaccine vector elicits effective anti-immunodeficiency-virus immunity. Nature. 415(6869):331-335.
Srivastava, I. K. and M. A. Liu (2003). "Gene vaccines." Ann Intern Med 138(7): 550-9.
Sullivan, N. J., Sanchez, A., Rollin, P. E., Yang, Z. Y., Nabel, G. J. (2000). Development of a preventive vaccine for Ebola virus infection in primates. Nature. 408(6812):605-609.
Sutter, G., Wyatt, L. S., Foley, P. L., Bennink, J. R., and Moss, B. (1994). A recombinant vector derived from the host range-restricted and highly attenuated MVA strain of vaccinia virus stimulates protective immunity in mice to influenza virus. Vaccine 12: 1032-1040.
Venter, J. C, et al. (2001). "The sequence of the human genome." Science 291(5507): 1304- 51.
Wang, R., D. L. Doolan, et al. (1998). "Malaria DNA vaccine induces antigen specific genetically restricted cytotoxic T lymphocytes in humans." Science 282: 476-480.
Wang, R., et al. (2001). "Induction of CD4(+) T cell-dependent CD8(+) type 1 responses in humans by a malaria DNA vaccine." Proc Natl Acad Sci U S A 98(19): 10817-22.
Washburn, M. P. and J. R. Yates, 3rd (2000). "Analysis of the microbial proteome." Curr Opin Microbiol 3(3): 292-7.
Yarosh, O.K., et al., (1996). Human adenovirus type 5 vectors expressing rabies glycoprotein. Vaccine, 14(13): p. 1257-64.
Ying, H., et al. (1999). "Cancer therapy using a self-replicating RNA vaccine." Nat Med 5(7): 823-827.

Claims

What is claimed is:
1. A method to induce protective immunity comprising: a. administrating a priming immunization preparation containing a Plasmodium antigen or a fragment thereof; and b. administrating a boosting immunization preparation containing said Plasmodium antigen or fragment thereof.
2. The method of claim 1, wherein said Plasmodium antigen is selected from a group consisting of PfAg2, PyAg2, PvAg2, PkAg2, PoAg2, PmAg2, PbAg2, PcAg2 and a combination thereof.
3. The method of claim 1, wherein said priming immunization preparation containing said antigen or fragment thereof, is selected from a group consisting of: a. a recombinant virus expression system; b. a recombinant protein antigen or a recombinant polypeptide; c. a synthetic peptide; d. a DNA vector; e. a whole organism or extract thereof; and f. a combination thereof.
4. The method of claim 1 , wherein said boosting immunization preparation containing said antigen or fragment thereof, is selected from a group consisting of a. a recombinant virus expression system; b. a recombinant protein antigen or a recombinant polypeptide; c. synthetic peptide; d. a DNA vector; e. a whole organism or extract thereof; and f. a combination thereof.
5. The method of Claim 3 or claim 4, wherein said recombinant virus expression system comprises Plasmodium antigen 2, fragment thereof, or epitope derived from said antigen, or combination thereof.
6. The method of claim 3 or claim 4, wherein said recombinant virus expression system is selected from the group consisting of poxvirus, adenovirus, adeno-associated virus, and retrovirus.
7. The method of Claim 5, wherein said poxvirus is selected from the group consisting of cowpox, canarypox, monkeypox, and fowlpox.
8. The method of claim 1, wherein said antigen is native or recombinant.
9. The method of Claim 1, wherein the number of doses of priming agent is 1-4 and the number of doses of boosting agent is 1-4.
10. The method of claim 1, wherein said immunization preparations are administered by routes selected from the group consisting of subcutaneous, intramuscular, intradermal, mucosal, oral, transcutaneous, and by injection devices, or combinations thereof.
1. A vaccine comprising an immunogenic preparation containing a Plasmodium antigen or fragment thereof, wherein said antigen is selected from a group consisting of PyAg2, PfAg2, PvAg2, PkAg2, PoAg2, PmAg2 and a combination thereof.
PCT/US2009/001643 2008-03-14 2009-03-16 Vaccine and immunization method using plasmodium antigen 2 WO2009114202A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/881,604 US20110229514A1 (en) 2009-03-16 2010-09-14 Vaccine and immunization method using plasmodium antigen 2

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US3666608P 2008-03-14 2008-03-14
US61/036,666 2008-03-14

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/881,604 Continuation-In-Part US20110229514A1 (en) 2009-03-16 2010-09-14 Vaccine and immunization method using plasmodium antigen 2

Publications (2)

Publication Number Publication Date
WO2009114202A2 true WO2009114202A2 (en) 2009-09-17
WO2009114202A3 WO2009114202A3 (en) 2009-12-30

Family

ID=41065740

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/001643 WO2009114202A2 (en) 2008-03-14 2009-03-16 Vaccine and immunization method using plasmodium antigen 2

Country Status (1)

Country Link
WO (1) WO2009114202A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012134591A3 (en) * 2011-03-25 2013-12-12 The United States Of America As Represented By The Secretary Of The Navy Plasmodium falciparum antigens
US9169304B2 (en) 2012-05-01 2015-10-27 Pfenex Inc. Process for purifying recombinant Plasmodium falciparum circumsporozoite protein

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040202667A1 (en) * 2002-03-04 2004-10-14 Daniel Carucci Antigens and their use as diagnostics and vaccines against species of plasmodium
US20060147477A1 (en) * 2002-06-11 2006-07-06 Glaxo Group Limited Immunogenic compositions

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040202667A1 (en) * 2002-03-04 2004-10-14 Daniel Carucci Antigens and their use as diagnostics and vaccines against species of plasmodium
US20060147477A1 (en) * 2002-06-11 2006-07-06 Glaxo Group Limited Immunogenic compositions

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012134591A3 (en) * 2011-03-25 2013-12-12 The United States Of America As Represented By The Secretary Of The Navy Plasmodium falciparum antigens
US9169304B2 (en) 2012-05-01 2015-10-27 Pfenex Inc. Process for purifying recombinant Plasmodium falciparum circumsporozoite protein
US9849177B2 (en) 2012-05-01 2017-12-26 Pfenex Inc. Process for purifying recombinant plasmodium falciparum circumsporozoite protein

Also Published As

Publication number Publication date
WO2009114202A3 (en) 2009-12-30

Similar Documents

Publication Publication Date Title
Schneider et al. Enhanced immunogenicity for CD8+ T cell induction and complete protective efficacy of malaria DNA vaccination by boosting with modified vaccinia virus Ankara
Doolan et al. DNA-based vaccines against malaria: status and promise of the Multi-Stage Malaria DNA Vaccine Operation
Hill Pre-erythrocytic malaria vaccines: towards greater efficacy
Moore et al. Progress in DNA‐based heterologous prime‐boost immunization strategies for malaria
Schneider et al. A prime-boost immunisation regimen using DNA followed by recombinant modified vaccinia virus Ankara induces strong cellular immune responses against the Plasmodium falciparum TRAP antigen in chimpanzees
Wang et al. Boosting of DNA vaccine-elicited gamma interferon responses in humans by exposure to malaria parasites
Walther et al. Safety, immunogenicity, and efficacy of prime-boost immunization with recombinant poxvirus FP9 and modified vaccinia virus Ankara encoding the full-length Plasmodium falciparum circumsporozoite protein
AU2017219613B2 (en) Novel antigen for use in malaria vaccine
US20050208020A1 (en) Enhancement of vaccine-induced immune responses and protection by heterologous boosting with alphavirus replicon vaccines
US20050031592A1 (en) Methods and compositions for inducing immune responses and protective immunity by priming with alpha virus replicon vaccines
US10919946B2 (en) Chimeric molecule useful in immunotherapy for leishmaniasis, which includes a fragment of the PFR1 protein of leishmania infantum with specific immunodominant epitopes
Richie High road, low road? Choices and challenges on the pathway to a malaria vaccine
WO2009114202A2 (en) Vaccine and immunization method using plasmodium antigen 2
Todryk et al. Building better T‐cell‐inducing malaria vaccines
US20110229514A1 (en) Vaccine and immunization method using plasmodium antigen 2
US9694062B2 (en) Plasmodium falciparum sporozoite and liver stage antigens
Oliveira-Ferreira et al. Protective CD8+ T cell responses against the pre-erythrocytic stages of malaria parasites: an overview
WO2020229805A1 (en) Vaccine immunogens
EP2451477A1 (en) Plasmodium falciparum hla class i restricted t-cell epitopes
Richie et al. Plasmodium falciparum pre-erythrocytic vaccines: previous experience and the challenge of selecting new candidates
Doolan et al. DNA-based vaccines against malaria and other diseases-from the laboratory to the clinic
Grubb A genomic approach to the identification of novel malaria vaccine antigens
HEPPNER JR et al. PROSPECTS FOR A TRAVELERS'
Freilich et al. Boosting of DNA Vaccine-Elicited Gamma

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09720827

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 09720827

Country of ref document: EP

Kind code of ref document: A2