US20200261561A9 - Plasmodium falciparum and plasmodium vivax vaccine - Google Patents

Plasmodium falciparum and plasmodium vivax vaccine Download PDF

Info

Publication number
US20200261561A9
US20200261561A9 US16/615,490 US201816615490A US2020261561A9 US 20200261561 A9 US20200261561 A9 US 20200261561A9 US 201816615490 A US201816615490 A US 201816615490A US 2020261561 A9 US2020261561 A9 US 2020261561A9
Authority
US
United States
Prior art keywords
seq
antibody
vaccine
strand
sequence
Prior art date
Legal status (The legal status 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 status listed.)
Pending
Application number
US16/615,490
Other versions
US20200113988A1 (en
Inventor
Ekkehard Werner
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Publication of US20200113988A1 publication Critical patent/US20200113988A1/en
Publication of US20200261561A9 publication Critical patent/US20200261561A9/en
Pending legal-status Critical Current

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/44Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from protozoa
    • C07K14/445Plasmodium
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/20Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans from protozoa
    • C07K16/205Plasmodium
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/70Vectors or expression systems specially adapted for E. coli
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56905Protozoa
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/44Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from protozoa
    • G01N2333/445Plasmodium
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/26Infectious diseases, e.g. generalised sepsis
    • 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

  • the present invention relates to a vaccine V comprising (A) at least one isolated polypeptide strand P comprising or consisting of at least nine consecutive amino acid moieties of the repetitive organellar protein, putative of Plasmodium falciparum or the hypothetical protein PVNK_04523 of Plasmodium vivax or a polynucleotide strand encoding for such polypeptide; and (B) at least one pharmaceutically acceptable carrier or excipient.
  • the present invention refers to an antibody binding to the repetitive organellar protein, putative of Plasmodium falciparum or the hypothetical protein PVNK_04523 of Plasmodium vivax or a polynucleotide strand encoding therefore, and to a method of generating such antibodies and uses thereof.
  • Malaria is a life-threatening disease caused by parasites that are transmitted to people through the bites of infected female Anopheles mosquitoes.
  • parasites There are 5 parasite species that cause malaria in humans, and 2 of these species, P. falciparum and P. vivax , pose the greatest threat.
  • P. falciparum is responsible for most malaria-related deaths globally.
  • P. vivax is the dominant malaria parasite in most countries outside of sub-Saharan Africa.
  • the malaria life cycle begins when the sporozoite stages of the P. falciparum parasite developing inside the salivary glands of an infected Anopheles female mosquito are transmitted to humans during a blood meal. Once inside the bloodstream, sporozoites travel to the liver, where they multiply and transform into infectious stages named merozoites. This represents the liver stage, a pre-erythrocytic stage. Each merozoite released from the liver invades an erythrocyte, inside which it grows and multiplies. Once the intraerythrocytic cycle is completed, the infected erythrocyte membrane lyses, releasing newly formed merozoites that go on to invade more erythrocytes by means of specialized invasion processes. This represents the blood stage. Blood stage parasites are responsible for the clinical manifestations of the disease.
  • the control of the mosquitos (the so-called vector) is the main way to prevent and reduce malaria transmission (vector control).
  • vector control Two forms of vector control—insecticide-treated mosquito nets and indoor residual spraying—are effective in a wide range of circumstances. Much of the success in controlling malaria is due to vector control. However, malaria-endemic areas of sub-Saharan Africa and India are causing significant concern due to high levels of malaria transmission and widespread reports of insecticide resistance. Rotational use of different classes of insecticides for IRS is recommended as one approach to manage insecticide resistance.
  • ACT artemisinin-based combination therapy
  • blood-stage malaria vaccines have been developed. Erythrocyte invasion by merozoites, the blood-stage, is an essential step in Plasmodium falciparum infection and leads to subsequent disease pathology. Proteins both on the merozoite surface and secreted from the apical organelles (micronemes, rhoptries and dense granules) mediate the invasion of erythrocytes; some of the molecules have been regarded as targets in the development of an anti-malaria vaccine.
  • the rhoptry protein Pch ROPE Repeated Organellar Protein; Pch ROPE was first identified, described and characterized in a BALB/c mouse model from the rodent malaria parasite Plasmodium chabaudi .
  • ROPE peptides that could, in principle, have some antigenic properties have been mentioned by Villard et al., 2007, and Kulangara et al., 2009. These documents do, however, not teach a vaccine suitible for efficiently treating or preventing malaria in a patient by means of active immunization. Antigens of Plasmodium vivax are not considered at all.
  • U.S. Pat. No. 8,716,443 teaches that the serum of malaria patients may comprise antibodies against malaria proteins, in particular against a truncated version of 844 amino acid moieties or less of a hypothetical protein of Plasmodium falciparum (MAL6P1.37 (PFF0165c)).
  • MAL6P1.37 PFF0165c
  • PFF0165c Plasmodium falciparum
  • Pch ROPE is encoded by an intronless gene. Its dominant structural features are that of a mainly alpha-helical protein, that will build a coiled-coil structure and has a putative transmembrane anchor sequence close to its C-terminal end. Partial sequences of the Pch ROPE DNA sequence were expressed as a recombinant protein in E. coli (Werner et al., 1998). These Pch ROPE partial protein sequences were use to further characterize Pch ROPE.
  • Pch ROPE is a Natural Antigen
  • BALB/c mice were, after infection with Plasmodium chabaudi 96V, treated with 1 mg of Nivaquine, once 40% of their red blood cells were infected with Plasmodium chabaudi 96V.
  • BALB/c mice survived an otherwise 100% lethal infection with Plasmodium chabaudi .
  • Mice were now immune against any further infection with Plasmodium chabaudi 96V.
  • Sera from these mice were immune-sera. It was shown in Western-blot studies that immune-sera recognize recombinant partial Pch ROPE protein sequences, demonstrating that Pch ROPE is a natural antigen occurring during an infection of BALB/c mice with Plasmodium chabaudi 96V (Werner et al., 1998).
  • Pch ROPE is a Rhoptry Protein
  • Antisera against recombinant fragments of the Pch ROPE protein were raised in Fisher rats. These anti-sera were subsequently used in IF—Indirect Immunofluorescence microscopy on erythrocytes of BALB/c-mice infected with Plasmodium chabaudi 96V.
  • Pch ROPE can be localized in the rhoptry organelles of Plasmodium chabaudi 96V during the more mature blood-stages and Pch ROPE can be localized around the entire erythrocyte membrane during the early stage of the invasion of the BALB/c erythrocyte by Plasmodium chabaudi 96V (Werner et al., 1998).
  • the localization of Pch ROPE in the rhoptry organelles, which are known to be involved in the invasion process, and the localization of Pch ROPE at the membrane of the newly infected erythrocyte suggest a direct role of the Pch ROPE protein during the invasion process of the erythrocyte by Plasmodium chabaudi 96V.
  • Pch ROPE is a Protective Antigen
  • BALB/c mice immunized with recombinant parts of Pch ROPE, were protected from an otherwise 100% lethal infection with Plasmodium chabaudi 96V. It is important to emphasize here, that in the presented BALB/c mice— Plasmodium chabaudi 96V model, which we used over a period of about 4 years, not a single unprotected BALB/c mice did ever survive an infection with Plasmodium chabaudi 96V.
  • Plasmodium falciparum and Plasmodium vivax are Potential Vaccine Candidates
  • Pch ROPE shares some similarities with other vaccine candidates located in the rhoptries, e.g. the Rhoptry-associated membrane antigen (RAMA) (Topolska et al. Infect Immun (2004) June; 72(6):3325-30) of Plasmodium falciparum , for example with respect to its transient location to the erythrocyte membrane during the early invasion stages of the erythrocyte.
  • RFA Rhoptry-associated membrane antigen
  • the homologue (orthologue) of Pch ROPE is the “repetitive organellar protein, putative” of Plasmodium falciparum 3D7 (GenBank: CZT98043.1) [formerly named Plasmodium falciparum 3D7 “Kid domain containing protein” (NCBI Reference Sequence: XP_0013495431)] SEQ ID NO: 2 and SEQ ID NO: 4 and the “hypothetical protein PVNG_04523” of Plasmodium vivax North Korean (GenBank: KNA01322.1) SEQ ID NO: 3 and SEQ ID NO: 5 for the two major human malaria parasite species.
  • the three homologues share a comparable size: Pch ROPE with 1939 amino acids the “repetitive organellar protein, putative” of Plasmodium falciparum 3D7 with 1979 amino acids and the “hypothetical protein PVNG_04523” of Plasmodium vivax North Korean with 1887 amino acids.
  • the three homologues share an identical predicted overall secondary structure: large alpha-helical segments, extended non-helical segments at the N- and C-termini of the protein, a predominantly coiled-coil structure and a sequence at identical sites upstream from the C-termini representing a putative trans-membrane segment, thus suggesting further a homologous biological function of the three homologous (orthologous) proteins.
  • a vaccine comprising an isolated polypeptide strand P of at least nine consecutive amino acid moieties of the repetitive organellar protein, putative of Plasmodium falciparum 3D7 or the hypothetical protein PVNG_04523 of Plasmodium vivax North Korean as well as a polynucleotide strand encoding for such polypeptide serve as effective means for targeting Plasmodium falciparum or Plasmodium vivax . This can be used in a medicinal as well as a non-medicinal context for generating antibodies.
  • a first aspect of the invention relates to a vaccine V comprising:
  • the vaccine V comprises:
  • the vaccine V comprises:
  • the vaccine of the present invention is designated as “vaccine V” in order to ease finding of the vaccine of the present invention in the text. It will be understand that the designation “V” does not have a technical meaning and can also be omitted without altering the scope of the present invention.
  • the polypeptide strand of the present invention is designated as “polypeptide strand P” in order to ease finding of the polypeptide strand of the present invention in the text. It will be understand that the designation “P” does not have a technical meaning and can also be omitted without altering the scope of the present invention.
  • the term “at least nine consecutive amino acid moieties of” in the context of a certain sequence may embrace any peptide strand that overlaps with nine or more consecutive amino acid moieties of the respective protein sequence.
  • polypeptide sequence of the full repetitive organellar protein, putative of Plasmodium falciparum 3D7 (GenBank: CZT98043.1) (SEQ ID NO: 2):
  • the polynucleotide strand encoding for the at least nine consecutive amino acid moieties of SEQ ID NO: 3 can be any sequence suitable for this purpose. Preferably, it is a part of or the whole sequence according to the polypeptide sequence of the full length hypothetical protein PVNG_04523 of Plasmodium vivax North Korean (GenBank: KNA01322.1; GenBank: KQ235267.1_1:98732-104326 Plasmodium vivax North Korean chromosome Unknown supercont.1.94) (SEQ ID NO: 5):
  • the term “isolated” may be understood in the broadest sense as any increase in purity.
  • the isolated polypeptide strand P does not form part of a plasmodium or larger sized fragments of 100 or more nm thereof.
  • the isolated polypeptide strand P represents at least 5% by weight, more preferably at least 10% by weight, even more preferably at least 25% by weight, even more preferably at least 50% by weight, even more preferably at least 75% by weight, in particular at least 90% by weight of the whole polypeptide content of the vaccine V.
  • polypeptide may be understood interchangeably throughout the invention in the broadest sense as any chemical entity mainly composed of amino acid residues and comprising at least nine amino acid residues consecutively linked with another via amide bonds. It will be understood that a protein in the sense of the present invention may or may not be subjected to one or more posttranslational modification(s) and/or be conjugated with one or more non-amino acid moiety/moieties.
  • the termini of the protein may, optionally, be capped by any means known in the art, such as, e.g., amidation, acetylation, methylation, acylation.
  • Posttranslational modifications are well-known in the art and may be but may not be limited to lipidation, phosphorylation, sulfatation, glycosylation, truncation, oxidation, reduction, decarboxylation, acetylation, amidation, deamidation, disulfide bond formation, amino acid addition, cofactor addition (e.g., biotinylation, heme addition, eicosanoid addition, steroid addition) and complexation of metal ions, non-metal ions, peptides or small molecules and addition of iron-sulphide clusters.
  • co-factors in particular cyclic guanidinium monophosphate (cGMP), but optionally also such as, e.g., ATP, ADP, NAD + , NADH+H + , NADP + , NADPH+H + , metal ions, anions, lipids, etc. may be bound to the protein, irrespective on the biological influence of these co-factors.
  • cGMP cyclic guanidinium monophosphate
  • ATP e.g., ATP, ADP, NAD + , NADH+H + , NADP + , NADPH+H +
  • metal ions anions, lipids, etc.
  • the isolated polypeptide strand P does not bear posttranslational modifications, i.e., consists of the plain polypeptide sequence consisting of consecutive amino acid moieties.
  • the terms “pharmaceutically acceptable carrier”, “pharmaceutically acceptable excipient”, “carrier” and “excipient” may be understood interchangeably in the broadest sense as any substance that may support the pharmacological acceptance of the vaccine V.
  • a pharmaceutically acceptable carrier may exemplarily be selected from the list consisting of an aqueous buffer, saline, water, dimethyl sulfoxide (DMSO), ethanol, vegetable oil, paraffin oil or combinations of two or more thereof.
  • the pharmaceutically acceptable carrier may optionally contain one or more detergent(s), one or more foaming agent(s) (e.g., sodium lauryl sulfate (SLS), sodium doceyl sulfate (SDS)), one or more coloring agent(s) (e.g., food coloring), one or more vitamin(s), one or more salt(s) (e.g., sodium, potassium, calcium, zinc salts), one or more humectant(s) (e.g., sorbitol, glycerol, mannitol, propylenglycol, polydextrose), one or more enzyme(s), one or more preserving agent(s) (e.g., benzoic acid, methylparabene, one or more antioxidant(s), one or more herbal and plant extract(s), one or more stabilizing agent(s), one or more chelating agents (e.g., ethylenediaminetetraacetic acid (EDTA), and/or one or more uptake mediator(s) (e.
  • adjuvant may be understood in the broadest sense as any that supports immunologic stimulation. It may be a solution or emulsion of antigen emulsified in mineral oil and used as an immunopotentiator (booster). An adjuvant may also comprise muramyl peptide (MDP). An adjuvant typically significantly enhances the immune response to the isolated polypeptide strand P. Typically, immune response is at least two-fold higher compared to a comparable vaccine lacking the adjuvant.
  • MDP muramyl peptide
  • the vaccine V comprises an adjuvant supporting immunologic stimulation.
  • the excipient of the vaccine V comprises at least one adjuvant, such as, e.g., alum.
  • the vaccine V comprises at least one adjuvant supporting immunologic stimulation selected from the group consisting of alum and an immunostimulatory peptide, and optionally one or more further pharmaceutically acceptable carriers.
  • the combination of an adjuvant such as alum and one or more immunostimulatory peptides may also be designated as composite adjuvant.
  • an adjuvant in the sense of the present invention is a composite adjuvant.
  • such composite adjuvant includes alum and one or more further immunostimulatory agents.
  • such composite adjuvant includes alum and one or more immunostimulatory peptides (e.g., muramyl peptide (MDP) and/or monophosphoryl lipid A (MPL), a modified bacterial coat molecule).
  • an immunostimulatory peptide is selected from the group consisting of muramyl peptide (MDP) and/or monophosphoryl lipid A (MPL).
  • alum bears several technical disadvantages such as rather unsatisfactory enlisting of cytotoxic T cells that may be helpful for fighting malaria (Leslie, 2013). It will be, thus, understood that alum may be replaced by another adjuvant that at least partly overcomes these drawbacks. Such replacement of an adjuvant would not alter the sense of the present invention.
  • peptides derived from SEQ ID NO: 2 or SEQ ID NO: 3 are particularly strong immunogens. Therefore, in some preferred embodiments, an adjuvant such as, e.g., muramyl peptide (MDP), may also be omitted. This may avoid undesired side effects and/or may be more cost-efficient. Accordingly, in another preferred embodiment, the vaccine V does (essentially) not comprise (thus, is (essentially) free of) muramyl peptide (MDP).
  • MDP muramyl peptide
  • the vaccine V does (essentially) not comprise (thus, is (essentially) free of) an adjuvant.
  • the vaccine V comprises at least one isolated polypeptide strand P comprising or consisting of at least nine consecutive amino acid moieties of the repetitive organellar protein, putative of Plasmodium falciparum 3D7 (SEQ ID NO: 2) or the hypothetical protein PVNG_04523 of Plasmodium vivax North Korean (SEQ ID NO: 3) or a polynucleotide strand encoding for said polypeptide strand P.
  • the vaccine V comprises or consists of:
  • the vaccine V comprises or consists of:
  • the vaccine V comprises or consists of:
  • the vaccine V comprises or consists of:
  • the isolated polypeptide strand P is not too short in order to provide various antigens and to improve antigenicity.
  • the isolated polypeptide strand P comprises or consists of at least 10, at least 15, at least 20, at least 50 or at least 100 consecutive amino acid moieties of SEQ ID NO: 2 and/or SEQ ID NO: 3.
  • the polypeptide strand P comprises or consists of from 10 to 250, from 10 to 100, from 15 to 75, from 20 to 50, or from 25 to 45 consecutive amino acid moieties of SEQ ID NO: 2.
  • the polypeptide strand P comprises or consists of from 10 to 250, of from 11 to 100, from 15 to 75, from 20 to 50, or from 25 to 45 consecutive amino acid moieties of SEQ ID NO: 3.
  • the polypeptide strand P comprises or consists of from 10 to 250, of from 11 to 100, from 15 to 75, from 20 to 50, or from 25 to 45 consecutive amino acid moieties of SEQ ID NO: 6.
  • the isolated polypeptide strand P comprises (or consists of) at least 100, at least 200, at least 500, at least 1000, or at least 1800 consecutive amino acids having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the polypeptide sequence of SEQ ID NO: 2 and/or SEQ ID NO: 3.
  • the isolated polypeptide strand P comprises or consists of an amino acid sequence having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the polypeptide sequence of SEQ ID NO: 2.
  • the isolated polypeptide strand P comprises or consists of an amino acid sequence having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the polypeptide sequence of SEQ ID NO: 3.
  • the isolated polypeptide strand P consists of an amino acid sequence having the polypeptide sequence of SEQ ID NO: 2. In a highly preferred embodiment, the isolated polypeptide strand P consists of an amino acid sequence having the polypeptide sequence of SEQ ID NO: 2 obtained from heterologous expression. In a highly preferred embodiment, the isolated polypeptide strand P consists of an amino acid sequence having the polypeptide sequence of SEQ ID NO: 2 obtained from heterologous expression in a bacterium, in particular in Escherichia coli ( E. coli ).
  • the isolated polypeptide strand P consists of an amino acid sequence having the polypeptide sequence of SEQ ID NO: 3. In a highly preferred embodiment, the isolated polypeptide strand P consists of an amino acid sequence having the polypeptide sequence of SEQ ID NO: 3 obtained from heterologous expression. In a highly preferred embodiment, the isolated polypeptide strand P consists of an amino acid sequence having the polypeptide sequence of SEQ ID NO: 3 obtained from heterologous expression in a bacterium, in particular in Escherichia coli ( E. coli ).
  • the vaccine V comprises two isolated polypeptide strands P consisting of amino acid sequence having the polypeptide sequences of SEQ ID NO: 2 and SEQ ID NO: 3.
  • the vaccine V comprises two isolated polypeptide strands P consisting of amino acid sequence having the polypeptide sequences of SEQ ID NO: 2 and SEQ ID NO: 3 each obtained from heterologous expression, preferably obtained from heterologous expression in a bacterium, in particular in Escherichia coli ( E. coli ).
  • the isolated polypeptide strand P comprises or consists of an amino acid sequence having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the polypeptide sequence of SEQ ID NO: 6.
  • the isolated polypeptide strand P comprises or consists of an amino acid sequence having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the polypeptide sequence of SEQ ID NO: 1.
  • the isolated polypeptide strand P comprises or consists of an amino acid sequence having amino acids of positions 1283-1516 (234 amino acids) of SEQ ID NO: 1.
  • the nucleotide sequence usable as antigen encodes for such polypeptide sequence of SEQ ID NO: 2 and/or SEQ ID NO: 3.
  • the nucleotide sequence usable as antigen bears at least 300, at least 600, at least 1500, at least 3000, or at least 5000 consecutive nucleotides having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the sequence of SEQ ID NO: 4 and/or SEQ ID NO: 5.
  • such sequence may optionally be embedded in a larger polynucleotide sequence.
  • the nucleotide sequence encodes for the polypeptide strand P (that may serve as antigen).
  • the nucleotide sequence encoding the polypeptide strand P bears at least 300, at least 600, at least 1500, at least 3000, or at least 5000 consecutive nucleotides having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the sequence of SEQ ID NO: 4.
  • the nucleotide sequence encoding the polypeptide strand P bears at least 300, at least 600, at least 1500, at least 3000, or at least 5000 consecutive nucleotides having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the sequence of SEQ ID NO: 5.
  • the at least one isolated polypeptide strand P consists of or comprises a peptide strand having at least 80% sequence homology to sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression. In a preferred embodiment, the at least one isolated polypeptide strand P consists of or comprises a peptide strand having at least 90% sequence homology to sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression. In a preferred embodiment, the at least one isolated polypeptide strand P consists of or comprises a peptide strand having at least 95% sequence homology to sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression.
  • the at least one isolated polypeptide strand P consists of or comprises a peptide strand having at least 98% sequence homology to sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression. In a preferred embodiment, the at least one isolated polypeptide strand P consists of or comprises a peptide strand having a sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression.
  • isolated polypeptide strand P as used in a vaccine V of the present invention may also form part of a fusion protein, a multi-antigen vaccine, or a viral vector.
  • the isolated polypeptide strand P comprises (essentially) the whole polypeptide sequence of SEQ ID NO: 2 and/or SEQ ID NO: 3.
  • the nucleotide sequence usable as antigen comprises (essentially) the whole polypeptide sequence of SEQ ID NO: 4 and/or SEQ ID NO: 5.
  • the isolated polypeptide strand P may be obtained by any means. In order to obtain a rather pure and polypeptide one effective mean is to employ an effective (over)expression technique. The person skilled in the art is aware of numerous methods suitable for this purpose. In a preferred embodiment, the isolated polypeptide strand P is obtained from heterologous expression.
  • heterologous expression may be understood in the broadest sense as expression of a gene encoding for the polypeptide according to SEQ ID NO: 2 and/or SEQ ID NO: 3 or fragment thereof in a host organism, which does not naturally have this gene or gene fragment. Insertion of the gene in the heterologous host may be performed by recombinant DNA technology which is well-known to the person skilled in the art. For this purpose, the gene may be inserted to the host, wherein it may be integrated into the host DNA causing permanent expression, or may not be integrated causing transient expression of this gene.
  • the isolated polypeptide strand P is obtained from heterologous expression in bacterial or eukaryotic cells.
  • the isolated polypeptide strand P is obtained from heterologous expression in bacterial cells such as E. coli .
  • Such host organism comprising the gene encoding for the polypeptide strand P may also be designated as “expression system”.
  • An advantage of such heterologous expression is that high amounts of the peptide are obtainable in good purity. In contrast to peptide synthesis, toxic agents can be avoided. On the other hand, in particular when expressed in bacteria, the peptides are typically not glycosylated.
  • the vaccine V of the present invention comprises or consists of:
  • the isolated polypeptide strand P may also have a higher homology and/or any of the other properties described herein.
  • the pharmaceutically acceptable carrier and/or the adjuvant may bear more specific properties.
  • the vaccine V comprises or consists of:
  • the vaccine V comprises or consists of:
  • polypeptide strand P may also be isolated from Plasmodium falciparum or Plasmodium vivax . This may optionally be performed by chromatographic means.
  • the at least one isolated polypeptide strand P comprises or consists of at least nine consecutive amino acid moieties of a sequence having at least 80% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • sequences of SEQ ID Nos 7 to 15 are the following:
  • the at least one isolated polypeptide strand P comprises or consists of at least nine consecutive amino acid moieties of a sequence having at least 90% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P comprises or consists of at least nine consecutive amino acid moieties of a sequence having at least 95% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P comprises or consists of at least nine consecutive amino acid moieties of a sequence having at least 98% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P comprises or consists of at least nine consecutive amino acid moieties of a sequence having a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the above peptides may also be at least ten consecutive amino acid moieties, at least 15 consecutive amino acid moieties, at least 20 consecutive amino acid moieties, at least 25 consecutive amino acid moieties, at least 50 consecutive amino acid moieties, or at least 100 consecutive amino acid moieties as far as the respective sequence provided above is long enough.
  • the at least one isolated polypeptide strand P comprises or consists of a sequence having at least 80% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P comprises or consists of a sequence having at least 90% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P comprises or consists of a sequence having at least 95% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P comprises or consists of a sequence having at least 98% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P comprises or consists of a sequence having a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising at least one peptide sequence selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising at least two peptide sequences selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising at least sequences of SEQ ID NO: 7 and 8, SEQ ID NO: 7 and 9, SEQ ID NO: 7 and 10, SEQ ID NO: 7 and 11, SEQ ID NO: 7 and 12, SEQ ID NO: 7 and 13, SEQ ID NO: 7 and 14, SEQ ID NO: 7 and 15, SEQ ID NO: 7 and 16, SEQ ID NO: 8 and 9, SEQ ID NO: 8 and 10, SEQ ID NO: 8 and 11, SEQ ID NO: 8 and 12, SEQ ID NO: 8 and 13, SEQ ID NO: 8 and 14, SEQ ID NO: 8 and 15, SEQ ID NO: 8 and 16, SEQ ID NO: 9 and 10, SEQ ID NO: 9 and 11, SEQ ID NO: 9 and 12, SEQ ID NO: 9 and 13, SEQ ID NO: 9 and 14, SEQ ID NO: 9 and 15, SEQ ID NO: 9 and 16, SEQ ID NO: 10 and 11, SEQ ID NO: 10 and 12, SEQ ID NO: 10 and 12, SEQ ID NO
  • truncated version of SEQ ID NO: 2 means a peptide comprising or consisting of a fraction of SEQ ID NO: 2 truncated by at least one amino acid moiety in length. In other words, in a t truncated version of SEQ ID NO: 2, at least one amino acid moiety is missing. Even if the truncated peptide sequence is extended by one or more amino acid moieties, these are not the same as those of SEQ ID NO: 2.
  • a truncated version of SEQ ID NO: 2 comprises or consists of a fraction of SEQ ID NO: 2 truncated by at least two, at least three, at least five, at least ten, at least 20, at least 50, at least 75, at least 100, at least 200, at least 500, or at least 1000 amino acid moieties in length.
  • the polypeptide strand P which is a truncated version of SEQ ID NO: 2 comprises or consists of from 32 to 1900, from 40 to 1800, from 50 to 1700, from 60 to 1600, from 70 to 1500, from 80 to 1400, from 90 to 1300, from 100 to 1200, from 200 to 1100, from 300 to 1000 amino acid moieties in length.
  • a truncated version of SEQ ID NO: 2 comprises or consists of a fraction of SEQ ID NO: 2 truncated by at least 100 amino acid moieties in length.
  • the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising or consisting of a fraction of SEQ ID NO: 2 truncated by at least 100 amino acid moieties in length and comprising at least two peptide sequences selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising or consisting of a fraction of SEQ ID NO: 2 truncated by at least 100 amino acid moieties in length and comprising all of the peptide sequences SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising at least three peptide sequences selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising at least four peptide sequences selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising at least five peptide sequences selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising at least six, at least seven, at least eight, or least nine, peptide sequences selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising all of the peptide sequences SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • polypeptide strand P may also be obtained by means of heterologous expression. It has been surprisingly found that also polypeptide strands P which were obtained from heterologous expression in bacteria led to a remarkable effectivity. Therefore, also non-glycosylated peptide strands P showed activity as experimentally evidenced.
  • the polypeptide strand P does not comprise (i.e., is (essentially) free of glycosylation.
  • glycosylated peptides are, however, also suitable. Therefore, in an alternative preferred embodiment, the polypeptide strand P comprises one or more glycosylated sides.
  • the polypeptide strand P comprises or consists of an easily assessable epitope. Accordingly, it preferably bears a sequence as laid out above. This is evidenced further by bioinformatic means as laid out in the experimental section below. In a preferred embodiment, the polypeptide strand P is not a hardly assessable sequence.
  • the polypeptide strand P in particular when the polypeptide strand P is not the full-length protein (of any of SEQ ID NOs: 2 or 3), the polypeptide strand P is not and/or comprises no peptide strand that has a length of nine or more consecutive amino acid moieties of at least 80% homology of any of SEQ ID NOs: 17-24. In a preferred embodiment, in particular when the polypeptide strand P is not the full-length protein (of any of SEQ ID NOs: 2 or 3), the polypeptide strand P is not and/or comprises no peptide strand that has a length of nine or more consecutive amino acid moieties of at least 80% homology of any of SEQ ID NOs: 17-21.
  • the polypeptide strand P in particular when the polypeptide strand P is not the full-length protein (of any of SEQ ID NOs: 2 or 3), the polypeptide strand P is not and/or comprises no peptide strand that has a length of nine or more consecutive amino acid moieties of at least 98% homology of any of SEQ ID NOs: 17-21. In a preferred embodiment, in particular when the polypeptide strand P is not the full-length protein (of any of SEQ ID NOs: 2 or 3), the polypeptide strand P is not and/or comprises no peptide strand that has a length of nine or more consecutive amino acid moieties of at least 98% homology of any of SEQ ID NOs: 17-24.
  • the polypeptide strand P in particular when the polypeptide strand P is not the full-length protein (of any of SEQ ID NOs: 2 or 3), the polypeptide strand P is not and/or comprises no peptide strand that has a length of nine or more consecutive amino acid moieties of any of SEQ ID NOs: 17-21. In a preferred embodiment, in particular when the polypeptide strand P is not the full-length protein (of any of SEQ ID NOs: 2 or 3), the polypeptide strand P is not and/or comprises no peptide strand that has a length of nine or more consecutive amino acid moieties of any of SEQ ID NOs: 17-24.
  • the polypeptide strand P is not a sequence having at least 80% sequence homology to a peptide sequence selected from the group consisting of one or more of SEQ ID NOs: 17-24. In a preferred embodiment, the polypeptide strand P is not a peptide strand of a sequence having at least 80% sequence homology to all of SEQ ID NOs: 17-21. In a preferred embodiment, the polypeptide strand P is not a peptide strand of a sequence having at least 80% sequence homology to all of SEQ ID NOs: 17-24.
  • SEQ ID Nos: 17-24 refer to the following sequences:
  • the polypeptide strand P does not have a sequence of SEQ ID NOs: 17-21. In a preferred embodiment, the polypeptide strand P does not have a sequence of SEQ ID NOs: 17-24.
  • a vaccine V based on a polynucleotide strand encoding for the polypeptide strand P may be any kind of polynucleotide.
  • the polynucleotide strand encoding for said polypeptide strand P is double or single stranded DNA or double or single stranded RNA, or an analogue of double or single stranded DNA or double or single stranded RNA.
  • nucleotide analogue may exemplarily comprise or even consist of nucleotide analogues such as, e.g., peptide nucleic acid (PNA), Morpholino and locked nucleic acid (LNA), as well as glycol nucleic acid (GNA) and threose nucleic acid (TNA).
  • PNA peptide nucleic acid
  • LNA Morpholino and locked nucleic acid
  • GNA glycol nucleic acid
  • TAA threose nucleic acid
  • the polynucleotide strand encoding for said polypeptide strand P is a plasmid.
  • the at least one polynucleotide strand encoding for the polypeptide strand P is a polynucleotide strand encoding for a sequence having at least 80% sequence homology, at least 90% sequence homology, at least 95% sequence homology, or at least 98% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the at least one polynucleotide strand encoding for the polypeptide strand P is a polynucleotide strand encoding for a sequence having a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • the vaccine V of the present invention is well suitible for preventing malaria in a patient.
  • a further aspect of the present invention relates to the vaccine V of the present invention for use in a method for preventing malaria in a patient.
  • the present invention relates to a method for preventing malaria in a patient, wherein a sufficient amount of the vaccine V is administered to said patient.
  • This method may be understood as active immunization.
  • malaria is preferably malaria caused by Plasmodium falciparum or Plasmodium vivax .
  • the respective vaccine V (comprising at least one isolated polypeptides P or at least one nucleotide encoding for such) will provide at least partly protection against Plasmodia of the respective type.
  • a vaccine V comprising at least one isolated polypeptides P of a sequence homolog to Plasmodium falciparum or at least one nucleotide encoding for such will particularly prevent the patient from malaria caused by Plasmodium falciparum.
  • a vaccine V comprising at least one isolated polypeptides P of a sequence homolog to Plasmodium vivax or at least one nucleotide encoding for such will particularly prevent the patient from malaria caused by Plasmodium vivax.
  • preventing does not necessarily means that the disease is completely prohibited, but means, in the broadest sense, that the symptoms of malaria are diminish, when the patient is exposed to the respective plasmodia (e.g., Plasmodium falciparum and/or Plasmodium vivax (and/or, regarding rodents, Plasmodium chabaudi )).
  • the effect of diminishing symptoms is the reduction in comparison to a comparable patient exposed to a comparable amount of the respective plasmodia (e.g., Plasmodium falciparum and/or Plasmodium vivax (and/or, regarding rodents, Plasmodium chabaudi )) without administration of the vaccine V.
  • a “sufficient amount” may be understood in the broadest sense as an amount that is suitible to provoke an immune response against plasmodia.
  • a sufficient amount vaccine V is suitible to diminish the quantity of the respective plasmodia (e.g., Plasmodium falciparum and/or Plasmodium vivax (and/or, regarding rodents, Plasmodium chabaudi )) when the patient is exposed to plasmodia.
  • the patient is administered with an amount of an isolated polypeptide strand P in the range of from 0.1 ⁇ g/kg to 1 g/kg, of from 1 ⁇ g/kg to 100 mg/kg, of from 5 ⁇ g/kg to 50 mg/kg, of from 10 ⁇ g/kg to 25 mg/kg, of from 25 ⁇ g/kg to 20 mg/kg, of from 50 ⁇ g/kg to 10 mg/kg, or of from 100 ⁇ g/kg to 5 mg/kg.
  • the patient is administered with a sufficient amount of the vaccine V to diminish the symptoms of malaria or even to prohibit the outbreak of malaria completely. In other words, this may be sufficient to confer protection.
  • the patient may be administered with the vaccine V of the present invention once, twice, three times of more often.
  • the patient may also be administered on a regular basis (e.g., once per month, once per two months, one per three months, once per half a year, once per year or once every five years).
  • the patient is administered only once. This may already be sufficient for developing an immune response to diminish the quantity of the respective plasmodia (e.g., Plasmodium falciparum and/or Plasmodium vivax (and/or, regarding rodents, Plasmodium chabaudi )) when the patient is exposed to plasmodia. This may diminish the symptoms of malaria or even to prohibit the outbreak of malaria completely. In other words, this may be sufficient to confer protection.
  • the respective plasmodia e.g., Plasmodium falciparum and/or Plasmodium vivax (and/or, regarding rodents, Plasmodium chabaudi )
  • the vaccine V is administered at least once to the patient before exposure to the respective plasmodia (e.g., Plasmodium falciparum and/or Plasmodium vivax (and/or, regarding rodents, Plasmodium chabaudi )).
  • the vaccine V is administered to the patient during and/or after exposure to the respective plasmodia.
  • the vaccine V is administered at least once to the patient before exposure to the respective plasmodia and during and/or after exposure to the respective plasmodia
  • the vaccine V of the present invention may be very well be used for generating antibodies binding to Plasmodium falciparum or Plasmodium vivax .
  • terms like “binding to”, “directed to”, “targeted” or the like may be understood interchangeably in the broadest sense as interacting selectively and non-covalently with a binding affinity of a dissociation constant (Kd) of 1000 nM or less.
  • Kd dissociation constant
  • the present invention also refers to the generation of antibodies by means of a vaccine V of the present invention.
  • the present invention also refers to the use of a vaccine V of the present invention for preparing an antibody AB binding to Plasmodium falciparum or Plasmodium vivax , in particular, binding to the repetitive organellar protein, putative of Plasmodium falciparum 3D7 or the hypothetical protein PVNG_04523 of Plasmodium vivax North Korean.
  • a further aspect of the present invention refers a method for preparing an antibody AB, binding to Plasmodium falciparum or Plasmodium vivax comprising the following steps:
  • the organism O may be any organism suitible for generating antibodies.
  • the organism is a (typically non-human) mammalian.
  • the organism O may be selected from the group consisting of a mouse, a rat, a rabbit, a goat, a hamster, a donkey, a cow, a pig, or a camel.
  • administration of step (ii) is systemic administration (e.g., intravenously (i.v.), intraarterially (i.a.), intraperitoneally (i.p.), intramusculary (i.m.), subcutaneously (s.c.), transdermally, nasally).
  • administration may also be local administration (e.g., intrathecally or intravitreally).
  • administration is systemic administration, in particular intravenous injection.
  • the waiting time depends on the vaccine V, the species of the organism O and the like. Typically it will take several days up to several weeks until an immune response is obtained. Exemplarily, the waiting time may be between 1 day and twenty weeks, preferably between 2 days and ten weeks, more preferably between 3 days and five weeks, exemplarily between 4 days and three weeks. During this time, the animals are kept under suitable conditions for maintain health for this species.
  • Obtaining antibody-generating cells C of the organism O of step (iii) may be performed by any means known for this purpose in the art.
  • the cells C may be obtained from the blood of the immunized organism O.
  • the cells C may be obtained from the lymph and/or spleen of the immunized organism O.
  • the organism O may be kept alive or may be sacrificed.
  • the antibodies AB of interest may be directly obtained from the cells C or may be obtained from further processed cells.
  • the method comprised the further step (v) of hybridizing the antibody-generating cells of step (iv) with myeloma cells obtaining immortalized antibody-generating cells C1.
  • This may enable obtaining monoclonal antibodies.
  • the person skilled in the art is well aware of methods usable for preparing such hybridoma cells C1.
  • the method comprised the further step (vi) of isolating the nucleotide encoding for the antibody AB of interest and transfer it to another antibody-generating cell type C2 suitible for expressing the antibody AB.
  • This may enable heterologous expression of the antibody AB or fragments thereof.
  • the Fc part of the antibody may be altered.
  • the antibody AB may than be humanized. The person skilled in the art is well aware of methods usable for this purpose. Further, the person skilled in the art will notice that the optional steps (v) and (vi) may optionally also be combined with another.
  • the antibody-generating cells C, C1 or C2 of any of steps (iv) to (vi) are cultivated under conditions enabling the production of the antibody AB followed by isolating the antibody AB.
  • Such isolation step may be performed by any means such as, e.g., by means of chromatographic means such as affinity chromatography using a stationary phase comprising the antigen of the antibody AB.
  • the present invention also embraces each of the antibody-generating cells C, C1 or C2.
  • the present invention also refers to an anti-repetitive organellar protein, putative of Plasmodium falciparum 3D7 or anti-hypothetical protein PVNG_04523 of Plasmodium vivax North Korean antibody.
  • a still further aspect of the present invention refers to an antibody or antibody fragment AB binding a polypeptide strand P of SEQ ID NO: 2 or SEQ ID NO: 3 or a polynucleotide strand encoding for said polypeptide strand P with a dissociation constant of not more than 1000 nM.
  • the antibody or antibody fragment AB binding a polypeptide strand P of SEQ ID NO: 2 or SEQ ID NO: 3 or a polynucleotide strand encoding for said polypeptide strand P with a dissociation constant of not more than 100 nM, more preferably not more than 100 nM, even more preferably not more than 50 nM or not more than 20 nM.
  • antibody may be understood in the broadest sense as any type of immunoglobulin or antigen-binding fraction or mutant thereof known in the art.
  • the antibody of the present invention may be an immunoglobulin A (IgA), immunoglobulin D (IgD), immunoglobulin E (IgE), immunoglobulin G (IgG), immunoglobulin M (IgM), immunoglobulin Y (IgY) or immunoglobulin W (IgW).
  • the antibody is an IgA, IgG or IgD. More preferably, the antibody is an IgG.
  • the type of antibody may be altered by biotechnological means by cloning the gene encoding for the antigen-binding domains of the antibody of the present invention into a common gene construct encoding for any other antibody type.
  • the binding between the antibody and its molecular target structure typically is a non-covalent binding.
  • the binding affinity of the antibody to its antigen has a dissociation constant (Kd) of less than 1000 nM, less than 500 nM, less than 200 nM, less than 100 nM, less than 50 nM, less than 40 nM, less than 30 nM or even less than 20 nM.
  • antibody as used herein may be understood in the broadest sense and also includes what may be designated as an antibody mutant.
  • an antibody mutant may be understood in the broadest sense as any antibody mimetic or antibody with altered sequence known in the art.
  • the antibody mutant may have at least 10%, at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90% or at least 95% of the binding affinity of a corresponding antibody, i.e., bear a dissociation constant (Kd) of less than 10 ⁇ M, less than 1 ⁇ M, less than 500 nM, less than 200 nM, less than 100 nM, less than 50 nM, less than 40 nM, less than 30 nM or even less than 20 nM.
  • Kd dissociation constant
  • the term “antibody fragment” may be understood in the broadest sense as any fragment of an antibody that still bears binding affinity to its molecular target (i.e., its antigen based on the polypeptide strand P of SEQ ID NO: 2 or SEQ ID NO: 3 or a polynucleotide strand encoding for said polypeptide strand P).
  • the antibody fragment may be a fragment antigen binding (Fab fragment), Fc, F(ab′) 2 , Fab′, scFv, a truncated antibody comprising one or both complementarity determining region(s) (CDR(s)) or the variable fragment (Fv) of an antibody.
  • Variable domains are the smallest fragments with an intact antigen-binding domain consisting of one V L and one V H .
  • Such fragments can be generated by enzymatic approaches or expression of the relevant gene fragments, e.g. in bacterial and eukaryotic cells.
  • Different approaches can be used, e.g. either the Fv fragment alone or ‘Fab’-fragments comprising one of the upper arms of the “Y” that includes the Fv plus the first constant domains.
  • These fragments are usually stabilized by introducing a polypeptide link between the two chains which results in the production of a single chain Fv (scFv).
  • dsFv disulfide-linked Fv
  • binding domains of fragments can be combined with any constant domain in order to produce full length antibodies or can be fused with other proteins and polypeptides.
  • a recombinant antibody fragment is the single-chain Fv (scFv) fragment. Dissociation of scFvs results in monomeric scFvs, which can be complexed into dimers (diabodies), trimers (triabodies) or larger aggregates such as TandAbs and Flexibodies.
  • the antibody may be a Fab, a Fab′, a F(ab′)2, a Fv, a disulfide-linked Fv, a scFv, a (scFv) 2 , a bivalent antibody, a bispecific antibody, a multispecific antibody, a diabody, a triabody, a tetrabody or a minibody.
  • antibody may also include an antibody mimetic which may be understood in the broadest sense as organic compounds that, like antibodies, can specifically bind antigens and that typically have a molecular mass in a range of from approximately 3 kDa to approximately 25 kDa.
  • Antibody mimetics may be, e.g., affibody molecules (affibodies), affilins, affitins, anticalins, avimers, DARPins, Fynomers, Kunitz domain peptides, single-domain antibodies (e.g., VHH antibodies or VNAR antibodies, nanobodies), monobodies, diabodies, triabodies, flexibodies and tandabs.
  • the antibody mimetics may be of natural origin, of gene technologic origin and/or of synthetical origin.
  • the antibody mimetics may also include polynucleotide-based binding units.
  • the antibody may also be a CovX-body.
  • the antibody may also be a cameloid species antibody.
  • the antibody or antibody fragment AB binds to its antigen based on the polypeptide strand P of SEQ ID NO: 2 or SEQ ID NO: 3 or a polynucleotide strand encoding for said polypeptide strand P with an at least 10-fold, even more preferably at least 100-fold, even more preferably at least 1000-fold higher binding affinity than to the corresponding (i.e., sequence homologue) antigen of a polypeptide SEQ ID NO: 2 or SEQ ID NO: 3 or the respective polynucleotide strand encoding for said polypeptide.
  • the antibody or antibody fragment AB may be obtained by any means.
  • the antibody or antibody fragment AB is obtained from a method of the present invention.
  • the antibody of the present invention may be a monoclonal antibody, a chimeric antibody and/or a humanized antibody.
  • Monoclonal antibodies are monospecific antibodies that are identical because they are produced by one type of immune cell that are all clones of a single parent cell.
  • a chimeric antibody is an antibody in which at least one region of an immunoglobulin of one species is fused to another region of an immunoglobulin of another species by genetic engineering in order to reduce its immunogenicity. For example murine V L and V H regions may be fused to the remaining part of a human immunoglobulin.
  • a particularly preferred type of chimeric antibodies are humanized antibodies. Humanized antibodies are produced by merging the DNA that encodes the CDRs of a non-human antibody with human antibody-producing DNA. The resulting DNA construct can then be used to express and produce antibodies that are usually not as immunogenic as the non-human parenteral antibody or as a chimeric antibody, since merely the CDRs are non-human.
  • the antibody or antibody fragment AB is a monoclonal humanized antibody.
  • the antibody or antibody fragment AB bears a high affinity to the polypeptide strand P of SEQ ID NO: 2 or SEQ ID NO: 3.
  • the antibody or antibody fragment AB bears a binding affinity to a polypeptide strand P of SEQ ID NO: 2 or SEQ ID NO: 3 of a dissociation constant of not more than 100 nM, not more than 50 nM, not more than 20 nM, not more than 10 nM, or not more than 5 nM.
  • the antibody or antibody fragment AB may be provided and stored in any suitable form.
  • the antibody or antibody fragment AB may optionally be dissolved in any medium suitable for storing said antibody such as, e.g., water, an aqueous buffer (e.g., a Hepes, Tris, or phosphate buffer (e.g. phosphate buffered saline (PBS)), an organic solvent (e.g., dimethyl sulfoxide (DMSO), dimethylformide (DMF)) or a mixture of two or more thereof.
  • PBS phosphate buffered saline
  • organic solvent e.g., dimethyl sulfoxide (DMSO), dimethylformide (DMF)
  • the antibody or mutant thereof according to the present invention may be of any species or origin.
  • any epitope(s) comprised by its molecular target structure e.g., linear epitope(s), structural epitope(s), primary epitope(s), secondary epitope(s), e.g., based on SEQ ID NO: 2 or SEQ ID NO: 3 or a nucleotide encoding therefor.
  • the antibody or mutant thereof may recognize the naturally folded molecular target structure or a domain or fragment thereof (e.g., SEQ ID NO: 2 or SEQ ID NO: 3 or a nucleotide encoding therefor in its natural environment inside the plasmodia).
  • the antibody or mutant thereof may be of any origin an antibody may be obtained from such as, e.g., natural origin, a gene technologic origin and/or a synthetic origin.
  • the antibody may also be commercially available.
  • the antibody may further comprise one or more posttranscriptional modification(s) and/or may be conjugated to one or more further structures such as label moieties or cell-penetrating peptides (CPPs).
  • CPPs cell-penetrating peptides
  • the antibody or antibody fragment may be added to a support, particularly a solid support such as an array, bead (e.g. glass or magnetic), a fiber, a film etc.
  • a support particularly a solid support such as an array, bead (e.g. glass or magnetic), a fiber, a film etc.
  • bead e.g. glass or magnetic
  • the antibody or antibody fragment AB may be conjugated with any kind of detectable label moiety. Then, the antibody or antibody fragment AB and the label moiety may be covalently or non-covalently conjugated with another, either directly of via a spacer.
  • conjugated with may be understood in the broadest sense as any kind of covalent or non-covalent attachment or linkage of one component with another component. Such conjugate can be obtained by chemical means and/or by genetic engineering and biotechnological means.
  • the label moiety may be any moiety that is detectable, preferably detectable by an imaging method.
  • the label moiety may be a fluorescent moiety.
  • a fluorescent moiety may be a fluorescent polypeptide moiety (e.g., cyan fluorescent protein (CFP), green fluorescent protein (GFP) or yellow fluorescent protein (YFP), red fluorescent protein (RFP), mCherry, etc.), a small-molecule dye moiety (e.g., an Atto dye moiety (e.g., ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 520, ATTO 532, ATTO 550, ATTO 565, ATTO 590, ATTO 594, ATTO 610, ATTO 611X, ATTO 620, ATTO 633, ATTO 635, ATTO 637, ATTO 647, ATTO 647N, ATTO 655, ATTO 665, ATTO 680, ATTO 700, ATTO 725, ATTO 740), a Cy dye moiety (e.g., Cy3, Cy5, Cy5.5, Cy 7), an Alexa
  • VivoTag680, VivoTag750 an S dye (e.g., S0387), a DyLight fluorophore moiety (e.g., DyLight 750, DyLight 800), an IRDye moiety (e.g., IRDye 680, IRDye 800), a fluorescein dye moiety (e.g., fluorescein, carboxyfluorescein, fluorescein isothiocyanate (FITC)), a rhodamine dye moiety (e.g., rhodamine, tetramethylrhodamine (TAMRA)), a HOECHST dye moiety, a quantum dot moiety or a combination of two or more thereof.
  • fluorescent label moiety may be used in fluorescence microscopy.
  • the label moiety may be metal atom, metal ion or metal bead (e.g., a (colloidal) gold such as a gold bead).
  • metal bead e.g., a (colloidal) gold such as a gold bead.
  • Such metal bead may be used in electron microscopy.
  • the label moiety may be radioactive label such as, e.g., 3 H, 14 C, 123 I, 124 I, 131 I, 32 P, 99m Tc or lanthanides (e.g., 64 Gd).
  • a radioactive label may or may not be suitable for scintillation assays, computer tomography (CT), single-photon emission computed tomography (SPECT) or as a label suitable for Positron Emission Tomography (PET) (e.g., 11 C, 13 N, 15 O, 18 F, 82 Rb).
  • the present invention also refers to the medicinal and non-medicinal uses of the antibody or antibody fragment AB of the present invention.
  • a still further aspect of the present invention refers to an antibody or antibody fragment AB of the present invention for use in a method for treating or preventing malaria in a patient.
  • the present invention refers to a method for treating or preventing malaria in a patient, said method comprising administration of antibody or antibody fragment AB of the present invention to said patient in an amount suitible for treating or preventing malaria in said patient.
  • administration is systemic administration (e.g., intravenously (i.v.), intraarterially (i.a.), intraperitoneally (i.p.), intramusculary (i.m.), subcutaneously (s.c.), transdermally, nasally).
  • administration may also be local administration (e.g., intrathecally or intravitreally).
  • administration is systemic administration, in particular intravenous injection.
  • the administration frequency may be adapted to the individual patient. Administration may be performed once, twice, or more often or continuously (e.g., via drip). Exemplarily, administration may be performed three times daily, twice daily, or every two days or less often.
  • a still further aspect of the present invention refers to an antibody or antibody fragment AB for use in a method for diagnosing malaria in a patient.
  • the present invention refers to a method for diagnosing malaria in a patient, said method comprising administration of antibody or antibody fragment AB of the present invention to said patient in an amount suitible for detecting Plasmodium falciparum or Plasmodium vivax in said patient.
  • the antibody or antibody fragment AB is preferably labelled, i.e., conjugated to a label moiety, as described above.
  • the antibody or antibody fragment AB according to the present invention may also be used for staining in vitro.
  • detection may be performed by enzyme-linked immunosorbent assay (ELISA), flow cytometry, fluorescence activated cell sorting (FACS), magnetic activated cell sorting (MACS), antibody-based dipsticks, etc.
  • ELISA enzyme-linked immunosorbent assay
  • FACS fluorescence activated cell sorting
  • MCS magnetic activated cell sorting
  • antibody-based dipsticks etc.
  • a further aspect of the present invention relates to a method for staining Plasmodium falciparum or Plasmodium vivax in vitro, said method comprising the steps of:
  • fluorescence microscopy may comprise one or more of the following methods: laser scanning microscopy (LSM), two-photon fluorescence microscopy, fluorescence molecular imaging (FMI), fluorescence energy transfer (FRET), fluorescence correlation spectroscopy (FCS), and/or fluorescence cross-correlation spectroscopy (FCCS). All these techniques as such are well-known to those skilled in the art.
  • LSM laser scanning microscopy
  • FMI fluorescence molecular imaging
  • FRET fluorescence energy transfer
  • FCS fluorescence correlation spectroscopy
  • FCCS fluorescence cross-correlation spectroscopy
  • the polypeptide strand P of SEQ ID NO: 1, SEQ ID NO: 2, SEQ IND NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16 or nucleotide encoding for at least one thereof may be fluorescently stained by a fluorescently labeled antibody AB1 or unlabeled AB1 in combination with labelled AB2. The excess fluorescently labeled antibody may be washed away and the localization and intensity is determined spatially resolved in the sample.
  • a still further aspect of the present invention refers to the (preferably in vitro) use of any of the polypeptides of the present invention as described herein (e.g. a polypeptide selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and/or SEQ ID NO: 16) for detecting Plasmodium falciparum or Plasmodium vivax in a patient's body fluid.
  • a polypeptide selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and/or SEQ ID NO: 16 for detecting Plasmodium fal
  • patient's body fluid is blood, blood plasm, blood serum or a fraction thereof.
  • a still further aspect of the present invention refers to a method of detecting Plasmodium falciparum or Plasmodium vivax (preferably in vitro), said method comprising the step of detecting the presence of any of the polypeptides of the present invention as described herein (e.g. a polypeptide selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and/or SEQ ID NO: 16) in a patient's body fluid.
  • a further step may optionally be treating malaria in the patient accordingly.
  • a still further aspect of the present invention refers to a polypeptide of the present invention as described herein (e.g. a polypeptide selected from the group consisting of (SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and/or SEQ ID NO: 16) for use in a method for diagnosing a Plasmodium falciparum or Plasmodium vivax infection in a patient.
  • the patient is further subjected to a malaria treatment.
  • a still further aspect of the present invention refers to a method of detecting Plasmodium falciparum or Plasmodium vivax (preferably in vitro), said method comprising the step of detecting the presence of an antibody specific for any of the polypeptides of the present invention as described herein (e.g. a polypeptide selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and/or SEQ ID NO: 16) in a patient's body fluid.
  • a further step may optionally be treating malaria in the patient accordingly.
  • a still further aspect of the present invention refers to an antibody specific for any of the polypeptides of the present invention as described herein (e.g. a polypeptide selected from the group consisting of (SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and/or SEQ ID NO: 16) for use in a method for diagnosing a Plasmodium falciparum or Plasmodium vivax infection in a patient.
  • the patient is further subjected to a malaria treatment.
  • a still further aspect of the present invention refers to a primer that is complementary to a fraction of any of SEQ ID NO: 4, SEQ ID NO: 5, or SEQ ID NO: 6 for detecting Plasmodium falciparum or Plasmodium vivax in a patient's body fluid.
  • the primer is a single chain nucleotide strand of a length of between five to 50 nucleotide moieties in length, between seven to 40 nucleotide moieties in length, between eight to 35 nucleotide moieties in length, or between nine to 30 nucleotide moieties in length.
  • a still further aspect of the present invention refers to a pair of primers that are both complementary to a fraction of any of SEQ ID NO: 4, SEQ ID NO: 5, or SEQ ID NO: 6 for detecting Plasmodium falciparum or Plasmodium vivax in a patient's body fluid.
  • a still further aspect of the present invention refers to a method of detecting Plasmodium falciparum or Plasmodium vivax (preferably in vitro) in a patient, said method comprising the steps of
  • a still further aspect of the present invention refers to a method of detecting Plasmodium falciparum or Plasmodium vivax (preferably in vitro) in a patient, said method comprising the steps of
  • a cDNA library of a Plasmodium chabaudi 96V was constructed in pBluescript SK (Werner et al. 1998). From the cDNA library the recombinant plasmid pBluescript SK (-) 70 was isolated by screening. It contains part of the Pch ROPE sequence coding for amino acids 1283-1516 (234 amino acids) as an insert.
  • the insert was sub cloned in the pGEX-1T vector, transformed in E. coli JM 109 bacteria and expressed as a glutathione-S-transferase fusion protein in E. coli JM 109.
  • the fusion protein was isolated using glutathione-agarose beads, followed by thrombin cleavage to obtain the 234 amino acid Pch ROPE protein fragment in a pure form, called rec 700.
  • mice were immunized at day 0 and day 21 with:
  • mice were infected with 5 ⁇ 10 6 Plasmodium chabaudi 96V parasitized erythrocytes.
  • Balb/c mice were immunized only two times and without any supplementation of an adjuvant such as muramyl peptide (MDP).
  • MDP muramyl peptide
  • ROPE is a particularly effective immunogen. It is indicating a very high protection capacity of the ROPE protein when used as a malaria vaccine. Moreover, ROPE is very effective for generating specific antibodies.
  • Steps towards the development of a Pf ROPE or Pv ROPE malaria vaccine include generation and testing of antibodies against ROPE fragments, testing of antibodies against ROPE peptide-microarrays covering the entire ROPE sequence, and preclinical and clinical trials.
  • Plasmodium falciparum (Pf) ROPE as peptides, raising soluble scFvs (single chain fragment variable) antibodies against these peptides and testing the capacity to block invasion of human red blood cells by Plasmodium falciparum in vitro in culture.
  • Antibodies showing the strongest inhibition will be used to produce antibodies that can be used for passive immunization in humans, similar to an anti-malaria drug.
  • Human antibodies are generated against above Pf ROPE peptides by phage display using human antibody gene libraries (Kugler et al. 2015).
  • biotinylated Pf ROPE peptides are immobilized on streptavidin coated microtiter plates.
  • the libraries are incubated with the peptides, non-binding antibody phage particles removed by rigid washing steps.
  • the bound antibody phages are eluted by trypsin and re-amplified using E. coli XL1-Blue and the M13-K07 helper phage.
  • two further panning rounds are performed.
  • Monoclonal antibodies are produced as soluble scFvs (single chain fragment variable) antibodies, using the phage display vector pHAL30 and identified by screening ELISA on the immobilized Pf ROPE peptides. This step is needed to discard non-specific binders of the corresponding antibody phage particles.
  • the monoclonal scFvs—single chain fragment variable-antibodies are re-cloned into the bivalent scFv-Fc format and produced in mammalian cells.
  • the mammalian vector pCSE2.6-hIgG-Fc-XP and HEK293 6E cells are used. This is an IgG like bivalent molecule and also effector functions are established. (Jager et al., 2013).
  • IgG-like antibodies are used to test for their capacity to block invasion of human red blood cells by Plasmodium falciparum in vitro in culture.
  • An in vitro growth inhibition activity assay (GIA) is used to measure the efficacy of the soluble scFvs antibodies directed against our peptides in blocking merozoite invasion (Kennedy et al., 2002).
  • Peptides identified as protective in the context of this in vitro inhibition can be used for the production of chemically synthesized vaccines, either as single peptides or as a fusion of several peptides or can be used as diagnostics.
  • Peptide microarrays covering the entire Pf ROPE sequence are prepared. Sera from malaria patients are used to identify the immunodominant parts of the Pf ROPE protein during the course of an infection with Plasmodium falciparum.
  • the whole amino acid sequence of the target protein is retrieved from a public database and translated into 15-mer peptides with a peptide-peptide overlap of e.g. 12 amino acids.
  • the peptide arrays with the corresponding peptides are produced by the company PEPperPRINT GmbH (Heidelberg, Germany) in a laser printing process on glass slides, coated with a PEGMA/PMMA graft copolymer, which are functionalized with a ßAla-ßAla-linker.
  • a layer of amino acid particles, containing Fmoc-amino acid pentafluorophenyl esters, is printed layer after layer onto the functionalized glass slides, with intermittent melting (i.e. coupling) steps at 90° C. and chemical washing and capping steps (Stadler et al., 2008), based on the same principle as Merrifield's solid-phase peptide synthesis. Peptides are generated in duplicates on the arrays, which are screened for IgG and IgM responses in human sera.
  • peptide microarrays are placed in incubation trays (PEPperPRINT GmbH, Heidelberg, Germany) and blocked for 30 min at room temperature with western blot blocking buffer MB-070 (Rockland, USA). Then, sera are diluted 1:1000 in PBS buffer with 0.05% Tween 20 pH 7.4 (PBS-T) and 10% blocking buffer, incubating the sera for 16 h at 4° C. and 50 RPM orbital shaking. Peptide microarrays are washed three times shortly with PBS-T, followed by an incubation with a 1:2500 dilution of the secondary fluorescently labeled antibody, together with a control antibody for 30 min at room temperature.
  • the peptide microarrays are washed with PBST and rinsed with deionized water. After drying in a stream of air, fluorescent images are acquired using an Odyssey Imaging System (LI-COR, USA) at 700 nm. Image analysis and quantification is performed with the PepSlide Analyzer software (Sicasys Software GmbH, Heidelberg, Germany).
  • LI-COR Odyssey Imaging System
  • the DNA sequence coding for the entire Pf ROPE protein or parts of it is amplified by PCR of genomic Pf DNA and cloned into the pET-21a (+)-plasmid at the multiple cloning site (MCS).
  • MCS multiple cloning site
  • the MCS is under the control of a T7 promoter and flanked by a T7- and a HIS-tag.
  • the recombinant pET-21a (+)-plasmid containing a sequence coding for Pf ROPE is transformed into E. coli BL21(DE3), a strain that allows high-efficiency protein expression of any gene that is under the control of a T7 promoter.
  • the expressed recombinant Pf ROPE protein carries a histidine-tag at its C-terminus and is purified on a Nickel-column.
  • the recombinant Pf ROPE protein is mixed with a pharmaceutically acceptable carrier or excipient.
  • a vaccine is obtained. This is applied to Aotus monkeys and once proven to be efficient in human trials.
  • a Plasmodium chabaudi protein contains a repetitive region with a predicted spectrin-like structure. Mol Biochem Parasitol. 94:185-196.

Abstract

The present invention relates to a vaccine V comprising (A) at least one isolated polypeptide strand P comprising or consisting of at least nine consecutive amino acid moieties of the repetitive organellar protein, putative of Plasmodium falciparum or the hypothetical protein PVNG_04523 of Plasmodium vivax or a polynucleotide strand encoding for such polypeptide; and (B) at least one pharmaceutically acceptable carrier or excipient. Furthermore, the present invention refers to an antibody binding to the repetitive organellar protein,putative of Plasmodium falciparumor the hypothetical protein PVNG_04523 of Plasmodium vivax or a polynucleotide strand encoding therefor, to a method of generating such antibody and uses thereof.

Description

  • The present invention relates to a vaccine V comprising (A) at least one isolated polypeptide strand P comprising or consisting of at least nine consecutive amino acid moieties of the repetitive organellar protein, putative of Plasmodium falciparum or the hypothetical protein PVNK_04523 of Plasmodium vivax or a polynucleotide strand encoding for such polypeptide; and (B) at least one pharmaceutically acceptable carrier or excipient. Furthermore, the present invention refers to an antibody binding to the repetitive organellar protein, putative of Plasmodium falciparum or the hypothetical protein PVNK_04523 of Plasmodium vivax or a polynucleotide strand encoding therefore, and to a method of generating such antibodies and uses thereof.
  • Malaria is a life-threatening disease caused by parasites that are transmitted to people through the bites of infected female Anopheles mosquitoes. There are 5 parasite species that cause malaria in humans, and 2 of these species, P. falciparum and P. vivax, pose the greatest threat. P. falciparum is responsible for most malaria-related deaths globally. P. vivax is the dominant malaria parasite in most countries outside of sub-Saharan Africa.
  • The malaria life cycle begins when the sporozoite stages of the P. falciparum parasite developing inside the salivary glands of an infected Anopheles female mosquito are transmitted to humans during a blood meal. Once inside the bloodstream, sporozoites travel to the liver, where they multiply and transform into infectious stages named merozoites. This represents the liver stage, a pre-erythrocytic stage. Each merozoite released from the liver invades an erythrocyte, inside which it grows and multiplies. Once the intraerythrocytic cycle is completed, the infected erythrocyte membrane lyses, releasing newly formed merozoites that go on to invade more erythrocytes by means of specialized invasion processes. This represents the blood stage. Blood stage parasites are responsible for the clinical manifestations of the disease.
  • According to the latest WHO estimates, released in December 2016, there were 212 million cases of malaria in 2015 and about 429 000 deaths. Some population groups are at considerably higher risk of contracting malaria, and developing severe disease, than others. Children under 5 are particularly susceptible to infection, illness and death; more than two thirds (70%) of all malaria deaths occur in this age group. Partial immunity against malaria is developed over years of exposure, and while it never provides complete protection, it does reduce the risk that malaria infection will cause severe disease.
  • The control of the mosquitos (the so-called vector) is the main way to prevent and reduce malaria transmission (vector control). Two forms of vector control—insecticide-treated mosquito nets and indoor residual spraying—are effective in a wide range of circumstances. Much of the success in controlling malaria is due to vector control. However, malaria-endemic areas of sub-Saharan Africa and India are causing significant concern due to high levels of malaria transmission and widespread reports of insecticide resistance. Rotational use of different classes of insecticides for IRS is recommended as one approach to manage insecticide resistance.
  • Malaria can be prevented through chemoprophylaxis (with antimalarial drugs), which suppresses the blood stage of malaria infections, thereby preventing malaria disease. Resistance to antimalarial medicines is a recurring problem. Resistance of P. falciparum to previous generations of medicines, such as chloroquine and sulfadoxine-pyrimethamine (SP), became widespread in the 1950s and 1960s. A good available treatment, particularly for P. falciparum malaria, is artemisinin-based combination therapy (ACT). An ACT contains both, the drug artemisinin and a partner drug. In recent years, parasite resistance to artemisinin has been detected in 5 countries of the Greater Mekong subregion: Cambodia, Lao People's Democratic Republic, Myanmar, Thailand and Viet Nam. In parallel, there were reports of increased resistance to ACT partner drugs in some settings.
  • All these means for treating bear considerable drawbacks. It has also been tried to prepare a variety of vaccines against malaria. Malaria vaccines are considered amongst the most important modalities for potential prevention of malaria disease and reduction of malaria transmission. Research and development in this field has been an area of intense effort by many groups over the last few decades.
  • Despite this, there is currently no licensed malaria vaccine. Malaria vaccines can address either the pre-erythrocytic-stage or the blood-stage (more rarely transmission-blocking vaccines are investigated). Also pre-erythrocytic malaria vaccines have been developed. RTS, S/AS01 (RTS, S) is an injectable pre-erythrocytic vaccine, directed against sporozoites, that provides partial protection against malaria in young children. The vaccine is being evaluated in sub-Saharan Africa as a complementary malaria control tool that potentially could be added to (and not replace) the core package of WHO-recommended preventive, diagnostic and treatment measures.
  • Furthermore, blood-stage malaria vaccines have been developed. Erythrocyte invasion by merozoites, the blood-stage, is an essential step in Plasmodium falciparum infection and leads to subsequent disease pathology. Proteins both on the merozoite surface and secreted from the apical organelles (micronemes, rhoptries and dense granules) mediate the invasion of erythrocytes; some of the molecules have been regarded as targets in the development of an anti-malaria vaccine.
  • Unfortunately, the anti-malaria vaccines known in the art so far bear rather limited effectiveness. Therefore, there is still the need for providing further effective anti-malaria vaccines. It has been shown that several rhoptry polypeptides generally qualify as potential vaccine candidates. This is of particular interest, because there are only about thirty proteins present in the rhoptries. One example could be the Rhoptry-associated membrane antigen (RAMA) (Topolska et al., 2004).
  • The rhoptry protein Pch ROPE—Repeated Organellar Protein; Pch ROPE was first identified, described and characterized in a BALB/c mouse model from the rodent malaria parasite Plasmodium chabaudi. Examples for ROPE peptides that could, in principle, have some antigenic properties have been mentioned by Villard et al., 2007, and Kulangara et al., 2009. These documents do, however, not teach a vaccine suitible for efficiently treating or preventing malaria in a patient by means of active immunization. Antigens of Plasmodium vivax are not considered at all.
  • U.S. Pat. No. 8,716,443 teaches that the serum of malaria patients may comprise antibodies against malaria proteins, in particular against a truncated version of 844 amino acid moieties or less of a hypothetical protein of Plasmodium falciparum(MAL6P1.37 (PFF0165c)). However, no vaccine based on rhoptry protein Pf ROPE or Pv ROPE is taught or suggested.
  • The sequence of ROPE was originally obtained from cDNA—and genomic DNA—libraries of Plasmodium chabaudi 96V (Werner et al., 1998, Mol Biochem Parasitol. 94(2):185-96). (SEQ ID NO: 1): polypeptide sequence of the Repeat Organellar Protein of Plasmodium chabaudi (cf., also GenBank: AAC63403):
  • MIFNLKKSKKNEDGSNKDSKKTNETSGIEKKEKSNKWYNKIVNNSTKKDK
    DKNNDSIVYDDESKVGENDHHMKEYELEDQLKETLKSITALSIKVKEYEV
    KIEELEKELKLEKEKQINKEYEKELNEKSEFIKRQMELLKEKELNINLKE
    NKINNKEIITLKREEKLNDIESEYIEKNKEKEKLNYEVTNIKMSLDKLTC
    EVQEKKDNLEKINKKVIEKENNLRELKEFMKEKNEIIESLDGTINDKKNA
    YEKLEISFEEKRKMIEMLDSKLIEKEENFANKQAKLEKENEIIIEKLKDI
    ESREKDFKSKEEKFASMENELNTLKSDLSKNACQMEVYKLEIKDLSQSLV
    EKEREIFEIKNEYDDKINNMKEKLSSINDKGIDNTVLHSEEEKINKLLKE
    KETELNEIHKKYNLEIETIKNELNEKEEELEKNKKAHTVEVTNLTKEIKL
    LEKKTEDAKEGHKNELNELNNQLSKLNKEKDNIKNENTELNDKISSLNSE
    VNILNKDKQTLGNDIKTLNDLINNLKNEINTSDNKMNKMKEDLAMLNEEM
    EGKCVVIDEIEKKYKNEIFMLEEKLKEKENYADLNDEISILRNSIYVKEK
    EFIEMKEFYENKINLFNKNFEEKKNIYENELNSLRLKYDNEQGLIKQIDE
    LNIQKLKTEEKYLQLYNDNMHMFRSICTKIDMPYSENIKGSDLVDFVTAY
    IKRRDESSSDANPDTTHKEMVAELEKRHAAIVAELEEKHKEEIAKLGEGH
    KEVVLRLGEQHKEETIILEEKHKDVVTKLGEQHKENIIKLEEEHKDVVTK
    LGDQYKEEIAKLKEEHAVVVAELEEKHKLGEGHKEMVDELEKRHADFVEG
    LEEKHKAETAKLEEGHKSEMNEVEKRHADFVEGLEEKHKAETAKLGEGHR
    EVVAGLEEKHKEVVAELEEKHKEEIAKLEEGHKEVMAELGEKHKEVVAGL
    EAKHNLEEGHKEMVAELEKRHADLVAVLEEQHKAEIIKLGEEHKEVVAGI
    EEKYKVEAIKLAEEHKDVVTKLGEQHKEEIAKLEDGHKEVVNEVEKKNAS
    LLNMLEENHKNEMIKLKEEHKESASDLVEKLYQKDEEVKNSNNKIEELTN
    VIKDLNDSIMCYKKQILEEVEKRNEYNEEINKLKIVQNEMKDMNDKKILE
    KENEIKKLNKKLSNYKVFETKENTYKNSEMVVNENKERIIVDSVCKENIS
    ESDVEGKGGNLKMTLSLKKKERNIFSINDNKNESSELVDTIKSAYINKIE
    MYKKEIEDNGKNIEDLKNKILDLSNELINLENMKNVLTDENNNLKKEIEI
    KDNKLNEKEKNENTEILNLNDDIIKLKKEISEWKDEEEKLTKENIKLKND
    IEQINKEYKIKEENLMIKFNENINEVTSLKNQIEIEKMKLEELNKNYELL
    LAEKRETNMSISNDDNKIVENNILEDTDSKQNNLNKNVEDKTGDDINCEK
    NNDQAKEISYLKDEIKKISMLYGEELNRKNSYDEKVKNLTNELKELKIRN
    KKGEEAIAELNKLKNIKEKNKSVKQNDESSSNNIITKDGDKTPEYVSNDD
    KIQKDWKANLVLKLKEKPDLWDNINSLEKENFRVMSIVKENKNVQNDKIV
    GIYSYFKKCEKELKNDMLVICLVLKDILSILFLNDNFVNLFEKIDKILWK
    QMYIPTEIRILFLRYFSFLDKLRNYVKCVNEEYVNNERYEYSWALFQTYL
    ETASNLKKEMIYYVLEKAEKDSCENNSSNFDKPKITDILNFSKDSIRLKT
    IAQLRKELNFEREAKNILNYDYQIILNKYHECLRKLKIVKNMARELDFNY
    NVSSKFSIKKELEMCSDENDEFKYNNIKNNEEKNDTIKDPKHNNLIQKII
    NLQRNKKTEKKKNNLVNEINTMYPGDTTPKGKIFTTNDNSKQNEILKKKD
    NINNNITHKNVYTGQVKNIFNEPVERKVRISFIHKSPFN
  • (Pch ROPE is encoded by an intronless gene. Its dominant structural features are that of a mainly alpha-helical protein, that will build a coiled-coil structure and has a putative transmembrane anchor sequence close to its C-terminal end. Partial sequences of the Pch ROPE DNA sequence were expressed as a recombinant protein in E. coli (Werner et al., 1998). These Pch ROPE partial protein sequences were use to further characterize Pch ROPE.
  • Pch ROPE is a Natural Antigen
  • BALB/c mice were, after infection with Plasmodium chabaudi 96V, treated with 1 mg of Nivaquine, once 40% of their red blood cells were infected with Plasmodium chabaudi 96V. Here BALB/c mice survived an otherwise 100% lethal infection with Plasmodium chabaudi. Mice were now immune against any further infection with Plasmodium chabaudi 96V. Sera from these mice were immune-sera. It was shown in Western-blot studies that immune-sera recognize recombinant partial Pch ROPE protein sequences, demonstrating that Pch ROPE is a natural antigen occurring during an infection of BALB/c mice with Plasmodium chabaudi 96V (Werner et al., 1998).
  • Pch ROPE is a Rhoptry Protein
  • Antisera against recombinant fragments of the Pch ROPE protein were raised in Fisher rats. These anti-sera were subsequently used in IF—Indirect Immunofluorescence microscopy on erythrocytes of BALB/c-mice infected with Plasmodium chabaudi 96V. Pch ROPE can be localized in the rhoptry organelles of Plasmodium chabaudi 96V during the more mature blood-stages and Pch ROPE can be localized around the entire erythrocyte membrane during the early stage of the invasion of the BALB/c erythrocyte by Plasmodium chabaudi 96V (Werner et al., 1998).The localization of Pch ROPE in the rhoptry organelles, which are known to be involved in the invasion process, and the localization of Pch ROPE at the membrane of the newly infected erythrocyte suggest a direct role of the Pch ROPE protein during the invasion process of the erythrocyte by Plasmodium chabaudi 96V.
  • Pch ROPE is a Protective Antigen
  • BALB/c mice, immunized with recombinant parts of Pch ROPE, were protected from an otherwise 100% lethal infection with Plasmodium chabaudi 96V. It is important to emphasize here, that in the presented BALB/c mice—Plasmodium chabaudi 96V model, which we used over a period of about 4 years, not a single unprotected BALB/c mice did ever survive an infection with Plasmodium chabaudi 96V.
  • The Pch ROPE Homologues (orthologues) of Plasmodium falciparum and Plasmodium vivax are Potential Vaccine Candidates
  • Survival of mice immunized with recombinant parts of Pch ROPE is a direct proof of Pch ROPE being a protective antigen in this model and thus suggesting that the Pch ROPE protein homologue (orthologue) of the human malaria parasites (Plasmodium falciparum and Plasmodium vivax) being potential vaccine candidates for human malaria disease.
  • In this context it seems to be noteworthy that Pch ROPE shares some similarities with other vaccine candidates located in the rhoptries, e.g. the Rhoptry-associated membrane antigen (RAMA) (Topolska et al. Infect Immun (2004) June; 72(6):3325-30) of Plasmodium falciparum, for example with respect to its transient location to the erythrocyte membrane during the early invasion stages of the erythrocyte. The homologue (orthologue) of Pch ROPE is the “repetitive organellar protein, putative” of Plasmodium falciparum 3D7 (GenBank: CZT98043.1) [formerly named Plasmodium falciparum 3D7 “Kid domain containing protein” (NCBI Reference Sequence: XP_0013495431)] SEQ ID NO: 2 and SEQ ID NO: 4 and the “hypothetical protein PVNG_04523” of Plasmodium vivax North Korean (GenBank: KNA01322.1) SEQ ID NO: 3 and SEQ ID NO: 5 for the two major human malaria parasite species.
  • The three homologues (orthologues) share a comparable size: Pch ROPE with 1939 amino acids the “repetitive organellar protein, putative” of Plasmodium falciparum 3D7 with 1979 amino acids and the “hypothetical protein PVNG_04523” of Plasmodium vivax North Korean with 1887 amino acids. The three homologues (orthologues) share an identical predicted overall secondary structure: large alpha-helical segments, extended non-helical segments at the N- and C-termini of the protein, a predominantly coiled-coil structure and a sequence at identical sites upstream from the C-termini representing a putative trans-membrane segment, thus suggesting further a homologous biological function of the three homologous (orthologous) proteins.
  • A vaccine comprising an isolated polypeptide strand P of at least nine consecutive amino acid moieties of the repetitive organellar protein, putative of Plasmodium falciparum 3D7 or the hypothetical protein PVNG_04523 of Plasmodium vivax North Korean as well as a polynucleotide strand encoding for such polypeptide serve as effective means for targeting Plasmodium falciparum or Plasmodium vivax. This can be used in a medicinal as well as a non-medicinal context for generating antibodies.
  • Accordingly, a first aspect of the invention relates to a vaccine V comprising:
      • (A) at least one isolated polypeptide strand P comprising or consisting of at least nine consecutive amino acid moieties of the repetitive organellar protein, putative of Plasmodium falciparum 3D7 (SEQ ID NO: 2) and/or the hypothetical protein PVNG_04523 of Plasmodium vivax North Korean (SEQ ID NO: 3) or a polynucleotide strand encoding for said polypeptide strand P; and
      • (B) at least one pharmaceutically acceptable carrier or excipient.
  • In a preferred embodiment, the vaccine V comprises:
      • (A) at least one isolated polypeptide strand P comprising or consisting of at least nine consecutive amino acid moieties of SEQ ID NO: 2 or SEQ ID NO: 3 or a polynucleotide strand encoding for said polypeptide strand P; and
      • (B) at least one pharmaceutically acceptable carrier or excipient.
  • In another preferred embodiment, the vaccine V comprises:
      • (A) at least one isolated polypeptide strand P comprising or consisting of at least nine consecutive amino acid moieties of SEQ ID NO: 2 and (concomitantly) SEQ ID NO: 3 or a polynucleotide strand encoding for said polypeptide strand P; and
      • (B) at least one pharmaceutically acceptable carrier or excipient.
  • As used herein, the vaccine of the present invention is designated as “vaccine V” in order to ease finding of the vaccine of the present invention in the text. It will be understand that the designation “V” does not have a technical meaning and can also be omitted without altering the scope of the present invention. As used herein, the polypeptide strand of the present invention is designated as “polypeptide strand P” in order to ease finding of the polypeptide strand of the present invention in the text. It will be understand that the designation “P” does not have a technical meaning and can also be omitted without altering the scope of the present invention.
  • It will be understood that the term “at least nine consecutive amino acid moieties of” in the context of a certain sequence may embrace any peptide strand that overlaps with nine or more consecutive amino acid moieties of the respective protein sequence.
  • The polypeptide sequence of the full repetitive organellar protein, putative of Plasmodium falciparum 3D7 (GenBank: CZT98043.1) (SEQ ID NO: 2):
  • MVFTFKNKKKKKEASSDKVSKESFNEEDNENNEKREKSDSWYKKIIETKG
    KSKTKYKNDNSLDDNINEDIINNNNNNNNDNNNDNNNDNNNDNNNDNNND
    NNNENNNDNNNFNNYSDEISKNIIHKDNELENQLKDTLKSISSLSNKIVN
    YESKIEELEKELKEVKDKNIDNNDYENKLKEKEDFVKQKIDMLNEKENLL
    QEKELDINKREKKINEKEKNIIKKEETFHNIEKEYLEKNKERETISIEII
    DIKKHLEKLKIEIKEKKEDLENLNKKLLSKENVLKELKGCVKEKNETINS
    LNDNIIEKEKKYKLLEYELEEKNKQIDLLNKQEKEKEKEKEREKEKEREK
    EKEKEYDTLIKELKDEKISILEKVHSIKVREMDIEKREHNFLHMEDQLKD
    LKNSFVKNNNQLKVYKCEIKNLKTELEKKEKELKDIENVSKEEINKLINQ
    LNEKEKQILAFNKNHKEEIHGLKEELKESVKITKIETQELQEMVDIKQKE
    LDQLQEKYNAQIESISIELSKKEKEYNQYKNTYIEEINNLNEKLEETNKE
    YTNLQNNYTNEINMLNNDIHMLNGNIKTMNTQISTLKNDVHLLNEQIDKL
    NNEKGTLNSKISELNVQIMDLKEEKDFLNNQIVDLSNQIDLLTRKMEEKE
    NKMLEQENKYKQEMELLRGNIKSSENILNNDEEVCDLKRKLSLKESEMKM
    MKEEHDKKLAELKDDCDVRIREMNEKNEDKINMLKEEYEDKINTLKEQNE
    DKINTLKEQNEDKINTLKEEYEHKINTMKEEYEHKINTLNEQNEHKINTL
    NEQNEHKINTMKEEYEDKMNTLNEQNEDKMNSLKEEYENKINQINSNNEI
    KIKDVVNEYIEEVDKLKVTLDEKKKQFDKEINYAHIKAHEKEQILLTEME
    ELKCQRDNKYSDLYEKYIKLIKSICMIINIECCDDIENEDIIRRIEEYIN
    NNKGLKKEVEEKEHKRHSSFNILKSKEKFFKNSIEDKSHELKKKHEKDLL
    SKDKEIEEKNKKIKELNNDIKKLQDEILVYKKQSNAQQVDHKKKSWILLK
    DKSKEKIKDKENQINVEKNEEKDLKKKDDEIRILNEELVKYKTILYNLKK
    DPLLQNQDLLSKIDINSLTINEGMCVDKIEEHILDYDEEINKSRSNLFQL
    KNEICSLTTEVMELNNKKNELIEENNKLNLVDQGKKKLKKDVEKQKKEIE
    KLNKQLTKCNKQIDELNEEVEKLNNENIELITYSNDLNNKFDMKENNLMM
    KLDENEDNIKKMKSKIDDMEKEIKYREDEKKRNLNEINNLKKKNEDMCIK
    YNEMNIKYGDICVKYEEMSLTYKETSLKYEQIKVKYDEKCSQYDEIRFQY
    DEKCFQYDEINKKYGALLNINITNKMVDSKVDRNNNEIISVDNKVEGIAN
    YLKQIFELNEEIIRLKGEINKISLLYSNELNEKNSYDINMKHIQEQLLFL
    EKTNKENEEKIINLTSQYSDAYKKKSDESKLCGAQFVDDVNIYGNISNNN
    IRTNEYKYEEMFDTNIEEKNGMHLSKYIHLLEENKFRCMKIIYENENIKS
    SNKIIGLYNYSRYYGLREDLCKEEIVPSKIGNISNKNENNNKKNNTCDGY
    DEKVTIVLCIILNEIIKFLFLNDEYVLLFEKIHKNVWKRMYIPEEIKFFI
    LKYITLLNNLRDYIISVHNNMKNEKYDECWFLFQHYFERSSDVRKEMVHF
    LLERKSQENLISFKSKLKSKKEKILTMDILNFSKEHMQLKTIAHLRKEIN
    YEKLSKDTLNRDYNLLLYKYQECVSKLKRVKNLMKEINQNVFIEKYDDIS
    KELDNFSDGYNEQNEQHVMDPILLNNNKNKNNKLITEHNNPIINRLTNFT
    QNRDSKYKNKIMDDVKQRKINSTMNNTNKNGINIIYNHYENLNKPNYNDN
    INRLNSYHQNIHIANSIHPNRNQNKSFLTNQANSTYSVMKNYINSDKPNL
    NGKKSVRNIFNEIVDENVNKTFVHKSVFF
  • The polypeptide sequence of the full length hypothetical protein PVNG_04523 of Plasmodium vivax North Korean (GenBank: KNA01322.1) (SEQ ID NO: 3):
  • MVFKFKKKKKEESSDKLSKQSQNDEGNANEEAEKKDHKSNSWYKKIIDNA
    IITKSKHDDKEEQEEEKNGEGNDSRAMERNKDYQLEEQLKETLRSITSLS
    TKIVNYETKIEDLEKELKMEKDKQVDKAYEKELKEKENFIKQKIGMLNEK
    ENLLNEKELDINMREEKINDREMFISKKEDKLNDMQEQYLEKNKEKEKLH
    FEIADIKISLEKLKYEVKDKKDCLENVSNKVILKENTLRELKEFIREKNE
    MIESLNEKITEKEKIYEQLGKDVEEKRKIIELLDMKANEKEKYFEEKIKE
    LEKEQNALLQKLNNVKMREKEVETRENDFLHMEDELNDLRSSFSKNDCQL
    KIYKLEIKDLSSALVEKEREILDLKNTYDGEICSLKDQIKEKEKEIAKGS
    SSGGDVGAQDEPASEVESEEKADPKEEGVENSLTDLLKMKERELHEMKEK
    YAKEIDTLNSELNEKKKEFVEAKNSHINQINNLNDEIEESESKMAELKSG
    YEMEIKKLRSEINAVHEEKYLLSNEKQTLSGEIDKLNEEKKSLASEKEEL
    HNKITTLNSEIGTLHVEKQALTGEINTLNDLIHTLKNEISSSDNLISKLK
    EQMNAINEEKEGKEKLITEIENNYKNEINALKEKLKDTDNQVSISIREEM
    DHLKCVLGETEKENKQMKEDYHKKIKQYDEELLSKQQYFEEELNNIRIKS
    HEKEQILILKNDELKESKLKTEEKYLKLYDDKMSLLRNMCSKVGLPYSDE
    VSVEELLERVGNYVSGMGEPGGAAHRGEQSEEPHEGQSIVEETNEPLLSA
    QTADNANSLEDKTTLQALQKELESVQEEYREEVAKMKSYLAMKEKTIEES
    NHTIAELTGKINSLNDTISFFKVNHSEEKINSYMDEINSLSLTLSELKAN
    NEQEQLENRNEIARLSEELSGYKRRADEQCRKRSSEKERSESKRGDTRGD
    SEKEQISESDVEGGGNLKSFLHFPLRKIKGKKRKASKTEKEIQTELRRNE
    PENEQSEKNEKAPRGDSLEVDQYKKELEEKAKIIEDLKDKICTLTNEVMD
    LKNLKNELAERDSSLAKAGEEAERQREQLDTLSAQLGGANGEVERLSEEV
    ERLNEEVEKLKEGEAQSWGEAEKWKGEAEKWKEDAAKWEADTVKLKEDAA
    KWESDAVKWESDAETWRKEAEELRSSANQLNEELCSKENNYVLKLNENVG
    VIQKMKDSIDAREKEKENYVREINDLRNELEGLKLKHDALSETYKQLEGK
    SSPPSGDDPPGGDNYTSEGENKLSIPNENCEMDQAEEANANPGVPKSEIA
    TEGGVSSLAVNDYISEIAHLKEEINRLTLLYSNELNEKNSSDIRTKELLS
    QLKELEVRDKENEEKIAALSKMNEKMKAKNEKLKSGKWLSRKDHAPNEEV
    DIAGEERKKKEKEKVPHPDVKEESLSSEHVNTLEGNTYRVMRIVDESSPA
    GGGQIIGSYLYTKKVEDLHAVNGANVADAQLAEKNAITVVCLILSEILSL
    LFLNDQFVNAFERINKSLWKLMYMPEEIKALLLRYFSFMSKLRDYAKEVH
    GRVENERYEDSQRQDNQRYDDSWLLFQNYLETSSSIKRDLVCFILEEKEN
    ELAELGEHYGGGMRKGEEVIGGVRGVRGGKIADIINLSKDEMRLKTIAQL
    RRDLDFEKKSKTLLSRDYQLLLYKYQECVRKLKRVKNMIRQLNLNDHSNR
    GSFALNRELDRCSEVSNERGFNEEGGDEDSHGNYKNCILQDNNNNSSVNN
    YNSSNTKLESRENVLIKDLINLRRAQKVKGNNLIHWGRPSMMMGGRCHQD
    ASHVVRAMVNGPKISSQNIFAHMNRLSNAPKISDHLDDMKKMKNIFNEFV
    ETRGDVTFVHRSPFCET
  • The polynucleotide strand encoding for the at least nine consecutive amino acid moieties of SEQ ID NO: 2 can be any sequence suitable for this purpose. Preferably, it is a part of or the whole sequence according to the repetitive organellar protein, putative of Plasmodium falciparum 3D7 (GenBank: CZT98043.1): 1..1979/locus_tag=“PF3D7_0203000”/coded_by=“complement (LN999943.1:141625..147564)” (SEQ ID NO: 4):
  • ATGGTATTCACGTTCAAAAATAAGAAAAAGAAAAAAGAAGCTAGTTCAGA
    TAAAGTAAGCAAAGAATCATTTAATGAGGAAGATAATGAAAATAATGAAA
    AGAGGGAAAAAAGCGATTCATGGTATAAAAAAATAATAGAAACTAAAGGA
    AAAAGTAAAACTAAATATAAAAATGATAATTCCTTAGATGATAATATTAA
    TGAGGACATAATAAATAATAATAATAATAATAATAATGATAATAATAATG
    ATAACAATAATGATAATAATAATGATAATAATAATGATAATAATAATGAT
    AATAATAATGAGAATAATAATGATAATAATAATTTTAATAATTATAGCGA
    TGAAATATCAAAAAATATTATACATAAAGACAATGAGCTAGAAAACCAGC
    TTAAGGATACATTAAAGTCCATTAGTTCGTTGTCGAATAAAATTGTGAAT
    TACGAAAGTAAAATTGAAGAATTAGAAAAAGAATTAAAAGAAGTAAAGGA
    TAAGAATATTGATAATAATGATTATGAAAATAAATTAAAAGAAAAAGAAG
    ATTTTGTTAAACAAAAAATTGATATGCTAAATGAAAAAGAAAATCTTTTA
    CAAGAAAAAGAATTAGATATTAATAAAAGAGAAAAGAAAATTAATGAAAA
    AGAAAAGAATATAATAAAAAAGGAAGAAACATTTCATAATATAGAAAAAG
    AGTATTTAGAAAAAAATAAAGAAAGAGAAACGATTTCTATAGAAATTATA
    GATATTAAAAAACATCTAGAAAAACTAAAAATAGAAATAAAAGAAAAAAA
    AGAAGATTTAGAAAATTTAAATAAAAAATTGTTATCAAAAGAAAATGTAC
    TAAAAGAATTAAAAGGATGTGTTAAGGAAAAAAATGAAACCATTAATTCA
    TTGAATGATAATATTATTGAAAAAGAAAAAAAATATAAATTATTAGAATA
    TGAGTTGGAAGAAAAAAATAAACAAATTGATTTATTAAACAAACAAGAAA
    AAGAAAAGGAAAAGGAGAAGGAAAGGGAAAAGGAGAAGGAAAGGGAAAAG
    GAAAAAGAAAAGGAATATGATACATTAATCAAAGAATTAAAAGATGAAAA
    GATTTCCATTTTAGAAAAAGTTCATTCCATTAAAGTAAGAGAAATGGATA
    TTGAAAAAAGAGAACATAATTTCCTTCATATGGAAGATCAATTAAAAGAT
    TTAAAAAATAGTTTTGTAAAGAATAATAATCAATTAAAAGTATATAAATG
    TGAAATAAAGAATCTTAAAACCGAATTAGAAAAAAAAGAAAAAGAATTAA
    AAGATATAGAAAATGTATCTAAAGAAGAAATAAATAAATTAATAAACCAA
    TTAAATGAAAAGGAGAAACAAATTCTTGCGTTTAATAAAAATCATAAAGA
    AGAAATTCATGGATTGAAAGAAGAATTAAAAGAATCTGTGAAAATAACCA
    AAATAGAAACACAAGAGTTACAAGAAATGGTAGACATCAAACAAAAAGAG
    TTAGACCAATTGCAGGAAAAATATAACGCACAAATAGAAAGTATAAGCAT
    CGAATTAAGTAAAAAAGAGAAGGAATATAATCAATATAAAAATACTTATA
    TAGAAGAAATAAATAATTTAAATGAAAAATTAGAAGAAACTAATAAAGAA
    TATACGAATTTACAAAATAATTATACAAATGAAATAAATATGTTAAATAA
    TGATATACATATGTTAAATGGCAATATAAAAACCATGAATACACAAATAA
    GTACTTTAAAAAATGATGTACATTTGTTAAATGAACAAATAGATAAATTA
    AATAATGAAAAGGGTACATTAAATAGTAAAATTAGTGAATTGAATGTTCA
    AATTATGGATTTAAAAGAGGAAAAAGATTTCTTAAATAATCAAATTGTAG
    ATTTAAGTAATCAAATTGATTTGTTAACAAGAAAAATGGAAGAGAAGGAA
    AATAAAATGTTGGAACAGGAGAATAAGTATAAACAAGAGATGGAACTCTT
    AAGGGGGAATATAAAAAGTTCTGAGAATATTTTAAACAATGACGAAGAGG
    TGTGTGATTTAAAAAGGAAATTAAGTTTGAAGGAAAGTGAAATGAAAATG
    ATGAAGGAGGAACATGATAAGAAGTTGGCTGAGTTGAAAGATGATTGTGA
    TGTGAGGATACGGGAGATGAATGAAAAGAATGAAGATAAAATTAATATGT
    TAAAGGAAGAATATGAAGATAAAATTAATACGTTGAAGGAACAAAATGAA
    GATAAAATTAATACGTTAAAGGAACAAAATGAAGATAAAATTAATACATT
    GAAAGAAGAGTATGAACATAAAATTAATACGATGAAGGAAGAATATGAAC
    ATAAAATTAATACGTTGAATGAACAAAATGAACATAAAATTAATACGTTG
    AATGAACAAAATGAACATAAAATTAATACGATGAAGGAAGAATATGAAGA
    TAAAATGAACACGTTGAATGAACAAAATGAAGATAAAATGAATTCGTTGA
    AGGAAGAGTATGAAAATAAGATAAATCAAATTAATAGTAATAATGAAATA
    AAAATAAAAGATGTAGTGAATGAATATATTGAAGAAGTGGACAAATTAAA
    AGTTACTTTGGATGAAAAAAAAAAACAATTTGATAAAGAAATAAATTACG
    CACATATCAAAGCTCATGAAAAGGAGCAAATATTATTAACAGAAATGGAA
    GAATTAAAATGTCAGAGGGATAATAAATATTCAGATTTATATGAGAAATA
    TATTAAACTAATAAAAAGTATTTGTATGATAATTAACATTGAATGTTGTG
    ATGATATAGAAAATGAAGATATTATAAGAAGAATTGAAGAATATATAAAT
    AATAACAAAGGCTTGAAAAAAGAAGTAGAAGAAAAAGAACATAAAAGACA
    TTCCTCCTTTAATATTTTAAAAAGTAAAGAAAAGTTTTTTAAAAATAGCA
    TAGAAGATAAAAGTCATGAATTAAAAAAAAAACATGAAAAAGATTTATTA
    TCAAAAGATAAAGAAATTGAAGAAAAGAATAAAAAAATAAAAGAACTGAA
    TAATGATATAAAAAAGTTACAAGATGAAATATTAGTATATAAAAAACAAA
    GTAATGCACAACAAGTAGATCATAAAAAGAAAAGTTGGATTCTTCTTAAA
    GATAAATCTAAAGAGAAAATAAAAGATAAAGAAAATCAAATAAATGTAGA
    AAAAAATGAAGAAAAGGATTTAAAAAAAAAAGATGATGAAATAAGAATTT
    TAAATGAAGAACTTGTAAAATATAAAACAATTTTATATAATTTAAAAAAA
    GATCCATTATTACAAAATCAAGATTTATTATCAAAAATTGACATAAATTC
    TTTAACAATAAATGAAGGAATGTGTGTAGATAAAATAGAAGAGCACATTT
    TGGATTATGATGAAGAAATAAATAAAAGCAGATCTAATTTGTTTCAACTA
    AAAAATGAAATATGTTCTTTAACAACTGAGGTTATGGAACTTAATAATAA
    GAAAAATGAATTAATTGAAGAAAATAATAAATTAAATTTAGTAGATCAAG
    GAAAGAAGAAATTAAAAAAGGATGTGGAAAAACAAAAAAAAGAAATAGAG
    AAATTAAATAAACAATTAACAAAATGTAATAAACAAATAGATGAATTAAA
    TGAAGAAGTGGAAAAATTAAATAATGAAAATATTGAATTAATTACATATT
    CAAATGATTTAAATAACAAATTTGATATGAAAGAAAATAATCTTATGATG
    AAATTAGATGAAAATGAAGATAATATAAAGAAAATGAAAAGTAAAATTGA
    TGATATGGAAAAAGAAATAAAATATAGAGAAGATGAAAAAAAAAGAAATT
    TAAATGAAATTAATAATTTAAAGAAAAAGAATGAAGATATGTGTATTAAA
    TATAATGAAATGAATATTAAGTATGGAGATATTTGTGTAAAATATGAAGA
    AATGTCTCTTACGTATAAAGAAACATCTCTTAAATATGAGCAAATTAAAG
    TGAAATATGATGAAAAGTGTTCTCAATATGACGAAATACGTTTTCAATAT
    GATGAGAAATGTTTTCAATATGATGAGATAAATAAGAAATATGGTGCTTT
    ATTAAATATAAATATTACTAATAAAATGGTTGATTCAAAAGTGGATAGAA
    ATAATAATGAAATAATTTCAGTAGATAATAAAGTAGAAGGAATTGCGAAT
    TATTTAAAACAAATATTTGAATTAAATGAAGAGATCATACGATTAAAAGG
    AGAAATAAATAAAATTAGCTTATTATATAGTAATGAATTAAATGAGAAAA
    ATAGTTATGATATAAACATGAAACATATACAAGAACAATTACTTTTTTTG
    GAAAAGACAAATAAAGAAAATGAAGAAAAAATAATTAATTTGACTAGCCA
    ATATTCTGATGCATACAAGAAGAAGAGTGATGAATCTAAATTATGTGGTG
    CACAGTTTGTTGATGATGTTAATATATATGGAAATATATCAAATAATAAT
    ATAAGAACAAATGAATATAAATATGAAGAGATGTTTGATACGAATATAGA
    AGAGAAGAATGGTATGCATTTATCTAAGTATATTCATCTATTAGAAGAAA
    ATAAATTTCGATGTATGAAAATAATTTATGAAAATGAAAATATAAAAAGT
    AGTAATAAAATAATTGGATTGTATAATTATTCAAGGTATTATGGGTTAAG
    AGAAGATTTGTGTAAAGAAGAAATCGTTCCTTCAAAAATAGGAAATATAT
    CTAATAAAAATGAAAATAATAATAAGAAGAACAACACTTGTGATGGTTAT
    GATGAGAAGGTTACAATAGTTTTATGCATTATATTAAATGAAATAATAAA
    ATTTTTATTTTTAAATGACGAATATGTATTATTATTTGAAAAGATTCATA
    AAAATGTTTGGAAACGAATGTATATCCCAGAAGAAATAAAATTTTTTATC
    CTAAAATATATTACGCTGTTAAATAACTTGAGAGATTATATAATAAGTGT
    ACATAATAATATGAAAAATGAGAAATATGATGAATGTTGGTTTTTATTTC
    AACATTATTTTGAAAGATCGAGTGATGTAAGAAAAGAGATGGTTCATTTC
    TTATTAGAAAGAAAGAGTCAAGAAAATTTAATATCTTTTAAAAGTAAATT
    AAAAAGTAAAAAAGAAAAAATATTAACAATGGACATATTGAATTTTAGTA
    AAGAACATATGCAATTAAAAACCATAGCTCATCTAAGAAAAGAAATAAAT
    TATGAAAAACTTTCTAAGGATACCTTAAATAGAGATTATAATTTATTATT
    ATATAAATATCAAGAATGTGTAAGTAAATTAAAAAGGGTAAAAAATTTAA
    TGAAAGAAATAAATCAAAATGTATTTATAGAAAAATATGATGATATAAGT
    AAAGAATTAGATAATTTTTCAGATGGATATAATGAACAAAATGAACAACA
    TGTAATGGATCCTATTTTATTAAATAATAATAAAAACAAAAATAACAAAT
    TGATAACTGAACATAATAATCCTATAATTAATAGGCTAACTAATTTTACA
    CAAAACAGAGATTCAAAATATAAAAATAAAATAATGGATGATGTAAAACA
    AAGAAAAATAAATAGTACAATGAATAATACAAATAAAAATGGTATTAATA
    TTATATATAATCATTATGAAAATTTAAATAAACCAAACTATAATGATAAT
    ATAAATAGATTAAATTCATATCATCAAAATATACATATTGCTAATTCAAT
    TCATCCTAATAGAAATCAAAATAAAAGTTTTCTTACGAATCAAGCAAATA
    GTACATATAGTGTTATGAAAAATTATATAAATTCAGATAAACCAAATTTA
    AATGGAAAAAAGAGTGTAAGAAATATTTTTAATGAAATTGTCGATGAAAA
    TGTAAATAAAACGTTTGTTCATAAAAGTGTATTTTTTTAA
  • The N-terminal part of the repetitive organellar protein, putative of Plasmodium falciparum 3D7 (GenBank: CZT98043.1) (SEQ ID NO: 6):
  • MVFTFKNKKKKKEASSDKVSKESFNEEDNENNEKREKSDSWYKKIIETKG
    KSKTKYKNDNSLDDNINEDIINNNNNNNNDNNNDNNNDNNNDNNNDNNND
    NNNENNNDNNNFNNYSDEISKNIIHKDNELENQLKDTLKSISSLSNKIVN
    YESKIEELEKELKEVKDKNIDNNDYENKLKEKEDFVKQKIDMLNEKENLL
    QEKELDINKREKKINEKEKNIIKKEETFHNIEKEYLEKNKERETISIEII
    DIKKHLEKLKIEIKEKKEDLENLNKKLLSKENVLKELKGCVKEKNETINS
    LNDNIIEKEKKYKLLEYELEEKNKQIDLLNKQEKEKEKEKEREKEKEREK
    EKEKEYDTLIKELKDEKISILEKVHSIKVREMDIEKREHNFLHMEDQLKD
    LKNSFVKNNNQLKVYKCEIKNLKTELEKKEKELKDIENVSKEEINKLINQ
    LNEKEKQILAFNKNHKEEIHGLKEELKESVKITKIETQELQEMVDIKQKE
    LDQLQEKYNAQIESISIELSKKEKEYNQYKNTYIEEINNLNEKLEETNKE
    YTNLQNNYTNEINMLNNDIHMLNGNIKTMNTQISTLKNDVHLLNEQIDKL
  • The polynucleotide strand encoding for the at least nine consecutive amino acid moieties of SEQ ID NO: 3 can be any sequence suitable for this purpose. Preferably, it is a part of or the whole sequence according to the polypeptide sequence of the full length hypothetical protein PVNG_04523 of Plasmodium vivax North Korean (GenBank: KNA01322.1; GenBank: KQ235267.1_1:98732-104326 Plasmodium vivax North Korean chromosome Unknown supercont.1.94) (SEQ ID NO: 5):
  • ATGGTATTCAAATTTAAGAAGAAAAAGAAGGAAGAAAGCTCGGACAAGTT
    AAGCAAGCAATCGCAAAACGATGAAGGAAATGCCAATGAGGAGGCAGAAA
    AAAAAGACCACAAGAGTAACTCCTGGTACAAGAAAATAATCGACAATGCA
    ATTATAACGAAGAGCAAGCATGACGATAAGGAGGAGCAGGAGGAGGAGAA
    AAATGGCGAAGGAAATGACAGCAGGGCGATGGAAAGAAATAAAGATTATC
    AATTGGAAGAGCAACTGAAGGAAACCCTAAGGTCAATCACGTCCTTGTCA
    ACCAAAATTGTGAATTACGAAACGAAGATTGAAGATTTGGAGAAAGAGTT
    AAAAATGGAAAAAGATAAACAAGTGGATAAGGCATACGAAAAGGAGTTGA
    AGGAGAAGGAGAATTTTATTAAACAAAAAATTGGCATGCTAAATGAGAAG
    GAAAATCTGCTAAATGAGAAGGAGCTGGACATAAATATGAGAGAAGAAAA
    AATTAATGACAGAGAAATGTTCATTTCGAAAAAGGAAGACAAACTGAATG
    ACATGCAGGAGCAGTACTTGGAAAAAAATAAAGAAAAAGAAAAACTCCAT
    TTTGAAATTGCAGATATTAAGATTTCCTTAGAAAAGCTAAAGTACGAAGT
    TAAAGATAAAAAGGACTGCCTAGAAAATGTCAGCAATAAGGTAATTTTGA
    AGGAAAATACTCTGAGGGAGTTAAAAGAATTTATAAGGGAAAAAAACGAA
    ATGATAGAATCGCTTAACGAGAAGATAACAGAGAAGGAGAAAATATATGA
    GCAGTTAGGGAAGGACGTGGAGGAGAAGAGAAAGATCATCGAATTGCTAG
    ACATGAAGGCAAATGAAAAGGAAAAATATTTCGAAGAAAAAATTAAAGAG
    TTAGAAAAAGAACAAAATGCGCTTCTGCAAAAGTTAAATAATGTTAAGAT
    GAGGGAGAAGGAAGTTGAGACGAGGGAAAATGACTTCCTGCACATGGAGG
    ACGAGCTGAATGATCTTCGCAGTAGCTTCTCGAAGAATGATTGTCAGCTA
    AAGATCTACAAATTGGAAATAAAAGATTTGAGCAGCGCCCTTGTGGAGAA
    GGAGAGAGAAATATTGGACTTGAAAAATACCTACGACGGGGAAATCTGCT
    CATTAAAGGATCAGATAAAGGAAAAGGAAAAGGAAATCGCCAAAGGTAGT
    TCCTCCGGTGGTGACGTGGGTGCACAAGATGAGCCAGCTAGCGAAGTTGA
    AAGTGAAGAAAAGGCGGACCCCAAAGAGGAAGGTGTGGAGAACAGCTTGA
    CCGATTTGCTCAAAATGAAGGAAAGAGAGCTGCACGAAATGAAGGAAAAA
    TACGCAAAGGAAATAGACACACTGAATAGCGAGCTGAATGAGAAAAAGAA
    AGAATTCGTGGAGGCAAAAAATAGCCACATCAACCAGATAAACAACCTAA
    ATGATGAAATTGAGGAGAGCGAAAGCAAAATGGCAGAACTGAAAAGTGGC
    TACGAAATGGAGATCAAAAAACTGCGCAGCGAAATTAATGCAGTGCACGA
    GGAGAAGTACCTCTTGAGCAACGAAAAACAAACACTCAGTGGAGAGATAG
    ACAAGCTGAATGAAGAGAAGAAGTCCCTGGCCAGCGAGAAGGAGGAGCTA
    CATAACAAAATAACCACGTTGAACAGCGAAATTGGGACGTTACATGTGGA
    GAAACAGGCACTCACTGGAGAAATAAACACCTTAAACGATCTGATTCACA
    CCCTGAAGAATGAAATCAGCTCGTCGGATAACCTGATTAGCAAATTGAAA
    GAGCAAATGAACGCCATCAACGAAGAAAAGGAAGGAAAGGAAAAACTCAT
    CACAGAGATAGAAAATAATTATAAAAATGAAATAAACGCGCTGAAGGAAA
    AATTAAAAGACACGGACAATCAAGTGAGCATAAGTATTAGGGAAGAGATG
    GACCACCTCAAATGCGTCCTTGGCGAAACGGAAAAGGAAAACAAACAGAT
    GAAGGAGGACTACCACAAAAAGATAAAACAGTATGATGAAGAATTGCTAT
    CGAAGCAGCAATATTTTGAAGAAGAATTAAATAACATACGCATCAAATCG
    CACGAAAAGGAACAAATTTTGATTTTAAAAAATGACGAGTTGAAGGAGTC
    GAAGCTAAAGACGGAGGAGAAGTACCTCAAGCTGTACGATGACAAAATGA
    GTCTCCTCAGGAATATGTGCTCCAAAGTGGGGCTCCCCTACAGCGATGAA
    GTTTCGGTGGAGGAGCTTCTCGAGCGGGTGGGCAACTACGTAAGTGGGAT
    GGGTGAACCGGGGGGTGCGGCACACAGGGGGGAGCAAAGTGAGGAGCCGC
    ATGAGGGGCAGTCGATTGTGGAGGAGACGAATGAACCCCTTTTGAGCGCC
    CAAACGGCCGACAATGCTAATAGCCTAGAGGACAAGACAACCCTACAGGC
    GCTACAGAAAGAACTGGAAAGTGTGCAAGAAGAGTACAGAGAAGAGGTAG
    CCAAAATGAAGAGCTATTTGGCGATGAAGGAAAAAACGATAGAGGAGTCG
    AACCACACAATCGCCGAGTTGACCGGAAAGATAAACAGCCTGAATGATAC
    CATTTCGTTTTTTAAGGTTAACCACTCTGAGGAGAAAATTAATTCCTATA
    TGGACGAAATTAACAGCTTGAGCTTGACGCTTAGCGAGCTGAAGGCTAAT
    AATGAACAGGAGCAGTTGGAGAATCGAAACGAAATTGCCAGGCTGTCGGA
    AGAGCTCAGCGGGTATAAGCGGCGTGCTGATGAGCAATGTAGAAAGAGGA
    GCAGCGAGAAGGAGAGAAGCGAGTCCAAGAGGGGAGACACAAGAGGTGAC
    TCCGAGAAGGAACAAATCTCCGAGTCGGACGTGGAAGGGGGGGGCAATTT
    AAAATCCTTTTTACACTTTCCCCTTCGAAAAATAAAAGGGAAAAAAAGAA
    AGGCCTCTAAAACTGAGAAGGAAATACAAACGGAGCTTAGGAGAAACGAG
    CCAGAGAATGAACAGAGTGAGAAAAATGAGAAGGCGCCTAGAGGAGACAG
    CCTGGAGGTGGACCAGTACAAAAAGGAATTGGAGGAAAAGGCGAAGATTA
    TTGAGGACTTGAAGGACAAAATATGCACCCTGACGAATGAGGTTATGGAT
    TTGAAGAATTTGAAGAACGAGCTGGCTGAGCGGGATAGCAGCTTGGCGAA
    GGCGGGCGAGGAGGCGGAAAGGCAAAGAGAGCAGTTGGACACGCTGAGCG
    CCCAACTGGGGGGTGCAAACGGAGAGGTGGAGAGACTCAGCGAAGAGGTG
    GAGAGGCTCAACGAAGAGGTGGAGAAGCTGAAGGAGGGAGAGGCACAATC
    GTGGGGGGAAGCGGAGAAGTGGAAAGGGGAAGCAGAGAAGTGGAAAGAAG
    ACGCAGCGAAGTGGGAAGCGGACACAGTGAAATTGAAAGAGGACGCAGCG
    AAATGGGAATCGGACGCAGTGAAGTGGGAATCGGACGCCGAGACGTGGAG
    GAAAGAAGCGGAGGAACTGAGAAGCAGCGCGAATCAATTGAACGAAGAAT
    TATGCTCGAAGGAAAATAACTACGTGTTGAAGCTGAACGAAAATGTGGGA
    GTTATACAAAAAATGAAGGACTCAATTGATGCACGTGAAAAGGAGAAGGA
    GAATTACGTTCGCGAGATAAACGATTTGAGAAACGAACTCGAAGGGTTGA
    AATTGAAGCATGATGCGTTGAGTGAGACGTATAAGCAGTTGGAGGGGAAG
    AGCAGCCCCCCCAGTGGAGATGACCCCCCTGGTGGAGATAACTACACCAG
    TGAGGGAGAGAATAAATTAAGCATCCCAAATGAGAATTGCGAAATGGACC
    AAGCGGAGGAAGCGAATGCCAACCCAGGTGTTCCCAAGAGCGAAATTGCC
    ACCGAGGGGGGTGTCTCCTCATTGGCAGTGAACGATTACATAAGCGAAAT
    AGCGCACCTGAAGGAAGAAATAAACAGACTAACCCTACTGTATAGCAACG
    AACTGAACGAAAAAAACAGCTCTGATATTAGGACCAAAGAGCTGCTGAGC
    CAGTTGAAGGAACTCGAAGTGAGGGATAAAGAAAATGAGGAAAAGATTGC
    TGCGCTGAGCAAAATGAATGAGAAAATGAAAGCGAAAAATGAAAAGCTGA
    AATCGGGGAAGTGGTTATCTAGGAAGGACCACGCGCCGAATGAAGAGGTA
    GATATCGCAGGGGAGGAGCGTAAGAAGAAGGAGAAGGAGAAAGTGCCTCA
    CCCGGATGTGAAAGAGGAGAGTCTGTCTTCAGAGCATGTGAACACACTGG
    AAGGAAACACCTACCGCGTGATGAGAATAGTTGATGAAAGTAGCCCCGCG
    GGAGGAGGCCAAATAATAGGGTCCTACTTGTACACCAAAAAGGTGGAAGA
    TTTACACGCAGTAAATGGAGCAAATGTGGCAGATGCACAGCTGGCTGAGA
    AAAACGCAATCACAGTTGTGTGTCTAATTCTAAGCGAAATCTTAAGCCTC
    CTATTTTTGAACGATCAATTTGTTAACGCCTTTGAACGGATAAACAAAAG
    TCTGTGGAAGCTTATGTACATGCCTGAAGAGATTAAAGCGCTGCTTCTGA
    GGTATTTTTCCTTTATGAGTAAGCTCAGGGATTATGCCAAGGAGGTGCAC
    GGGAGGGTGGAAAATGAGAGGTATGAAGACAGCCAAAGGCAGGACAACCA
    ACGGTACGACGATTCGTGGTTACTTTTTCAAAACTATTTGGAGACGTCGA
    GTAGTATCAAGAGGGACCTGGTGTGCTTCATTTTGGAAGAGAAGGAAAAT
    GAACTAGCCGAGCTGGGCGAGCACTATGGTGGTGGAATGAGAAAGGGAGA
    GGAAGTAATCGGGGGAGTACGCGGAGTGCGCGGGGGAAAAATCGCCGACA
    TCATAAACCTTTCAAAGGACGAAATGAGATTGAAGACCATAGCACAGTTA
    AGAAGAGACCTAGATTTTGAAAAGAAATCGAAAACATTGCTAAGCAGGGA
    TTATCAGTTGTTACTTTATAAGTACCAGGAATGCGTGAGGAAGCTAAAGA
    GGGTAAAAAATATGATAAGGCAGCTAAATCTGAACGACCATTCAAATAGA
    GGCAGTTTCGCCTTAAACAGGGAGCTGGACAGGTGTTCCGAAGTGAGCAA
    CGAGCGAGGTTTTAACGAGGAGGGGGGTGATGAAGATTCGCACGGAAATT
    ACAAAAACTGCATTCTGCAAGACAATAATAATAATAGCAGTGTAAATAAC
    TATAATAGTAGTAACACCAAATTGGAGAGTCGGGAAAATGTTCTAATCAA
    GGACCTAATCAATTTGAGGAGGGCGCAAAAGGTGAAGGGAAATAATTTGA
    TCCACTGGGGCCGTCCCAGCATGATGATGGGGGGCAGGTGTCACCAAGAC
    GCTTCCCATGTGGTAAGGGCGATGGTAAATGGACCCAAAATAAGCAGCCA
    GAATATCTTCGCACACATGAACAGGCTGAGCAATGCGCCCAAAATTAGCG
    ACCACTTGGATGACATGAAAAAAATGAAAAATATTTTTAACGAATTTGTT
    GAAACCAGAGGGGACGTTACGTTTGTGCACAGGAGTCCCTTCTGCGAAAC
    GTGA
  • As used herein, the term “isolated” may be understood in the broadest sense as any increase in purity. The isolated polypeptide strand P does not form part of a plasmodium or larger sized fragments of 100 or more nm thereof. Preferably, the isolated polypeptide strand P represents at least 5% by weight, more preferably at least 10% by weight, even more preferably at least 25% by weight, even more preferably at least 50% by weight, even more preferably at least 75% by weight, in particular at least 90% by weight of the whole polypeptide content of the vaccine V.
  • The terms “polypeptide”, “protein” and “peptide” may be understood interchangeably throughout the invention in the broadest sense as any chemical entity mainly composed of amino acid residues and comprising at least nine amino acid residues consecutively linked with another via amide bonds. It will be understood that a protein in the sense of the present invention may or may not be subjected to one or more posttranslational modification(s) and/or be conjugated with one or more non-amino acid moiety/moieties. The termini of the protein may, optionally, be capped by any means known in the art, such as, e.g., amidation, acetylation, methylation, acylation. Posttranslational modifications are well-known in the art and may be but may not be limited to lipidation, phosphorylation, sulfatation, glycosylation, truncation, oxidation, reduction, decarboxylation, acetylation, amidation, deamidation, disulfide bond formation, amino acid addition, cofactor addition (e.g., biotinylation, heme addition, eicosanoid addition, steroid addition) and complexation of metal ions, non-metal ions, peptides or small molecules and addition of iron-sulphide clusters. Moreover, optionally, co-factors, in particular cyclic guanidinium monophosphate (cGMP), but optionally also such as, e.g., ATP, ADP, NAD+, NADH+H+, NADP+, NADPH+H+, metal ions, anions, lipids, etc. may be bound to the protein, irrespective on the biological influence of these co-factors. In the context of Glu-plasminogen in particular glycosylation may play a role. In a particularly preferred embodiment, the isolated polypeptide strand P does not bear posttranslational modifications, i.e., consists of the plain polypeptide sequence consisting of consecutive amino acid moieties.
  • As used herein, the terms “pharmaceutically acceptable carrier”, “pharmaceutically acceptable excipient”, “carrier” and “excipient” may be understood interchangeably in the broadest sense as any substance that may support the pharmacological acceptance of the vaccine V. A pharmaceutically acceptable carrier may exemplarily be selected from the list consisting of an aqueous buffer, saline, water, dimethyl sulfoxide (DMSO), ethanol, vegetable oil, paraffin oil or combinations of two or more thereof. Furthermore, the pharmaceutically acceptable carrier may optionally contain one or more detergent(s), one or more foaming agent(s) (e.g., sodium lauryl sulfate (SLS), sodium doceyl sulfate (SDS)), one or more coloring agent(s) (e.g., food coloring), one or more vitamin(s), one or more salt(s) (e.g., sodium, potassium, calcium, zinc salts), one or more humectant(s) (e.g., sorbitol, glycerol, mannitol, propylenglycol, polydextrose), one or more enzyme(s), one or more preserving agent(s) (e.g., benzoic acid, methylparabene, one or more antioxidant(s), one or more herbal and plant extract(s), one or more stabilizing agent(s), one or more chelating agents (e.g., ethylenediaminetetraacetic acid (EDTA), and/or one or more uptake mediator(s) (e.g., polyethylene imine (PEI), a cell-penetrating peptide (CPP), a protein transduction domain (PTD), an antimicrobial peptide, etc.). In addition, the excipient of the vaccine V may or may not comprise one or more adjuvants. An excipient may be an adjuvant such as, e.g., alum.
  • As used herein, the term “adjuvant” may be understood in the broadest sense as any that supports immunologic stimulation. It may be a solution or emulsion of antigen emulsified in mineral oil and used as an immunopotentiator (booster). An adjuvant may also comprise muramyl peptide (MDP). An adjuvant typically significantly enhances the immune response to the isolated polypeptide strand P. Typically, immune response is at least two-fold higher compared to a comparable vaccine lacking the adjuvant.
  • In a preferred embodiment, the vaccine V comprises an adjuvant supporting immunologic stimulation. In a preferred embodiment, the excipient of the vaccine V comprises at least one adjuvant, such as, e.g., alum. In a preferred embodiment, the vaccine V comprises at least one adjuvant supporting immunologic stimulation selected from the group consisting of alum and an immunostimulatory peptide, and optionally one or more further pharmaceutically acceptable carriers. The combination of an adjuvant such as alum and one or more immunostimulatory peptides may also be designated as composite adjuvant. In a preferred embodiment, an adjuvant in the sense of the present invention is a composite adjuvant. In a preferred embodiment, such composite adjuvant includes alum and one or more further immunostimulatory agents. In a preferred embodiment, such composite adjuvant includes alum and one or more immunostimulatory peptides (e.g., muramyl peptide (MDP) and/or monophosphoryl lipid A (MPL), a modified bacterial coat molecule). In a preferred embodiment, an immunostimulatory peptide is selected from the group consisting of muramyl peptide (MDP) and/or monophosphoryl lipid A (MPL).
  • It is known that alum bears several technical disadvantages such as rather unsatisfactory enlisting of cytotoxic T cells that may be helpful for fighting malaria (Leslie, 2013). It will be, thus, understood that alum may be replaced by another adjuvant that at least partly overcomes these drawbacks. Such replacement of an adjuvant would not alter the sense of the present invention.
  • As experimentally evidenced, peptides derived from SEQ ID NO: 2 or SEQ ID NO: 3 are particularly strong immunogens. Therefore, in some preferred embodiments, an adjuvant such as, e.g., muramyl peptide (MDP), may also be omitted. This may avoid undesired side effects and/or may be more cost-efficient. Accordingly, in another preferred embodiment, the vaccine V does (essentially) not comprise (thus, is (essentially) free of) muramyl peptide (MDP).
  • In an alternative preferred embodiment, the vaccine V does (essentially) not comprise (thus, is (essentially) free of) an adjuvant.
  • In a preferred embodiment, the vaccine V comprises at least one isolated polypeptide strand P comprising or consisting of at least nine consecutive amino acid moieties of the repetitive organellar protein, putative of Plasmodium falciparum 3D7 (SEQ ID NO: 2) or the hypothetical protein PVNG_04523 of Plasmodium vivax North Korean (SEQ ID NO: 3) or a polynucleotide strand encoding for said polypeptide strand P.
  • In a preferred embodiment, the vaccine V comprises or consists of:
      • (A) at least one isolated polypeptide strand P comprising or consisting of at least nine consecutive amino acid moieties of SEQ ID NO: 2; and
      • (B) at least one pharmaceutically acceptable carrier or excipient.
  • In another preferred embodiment, the vaccine V comprises or consists of:
      • (A) at least one isolated polypeptide strand P comprising or consisting of at least nine consecutive amino acid moieties of SEQ ID NO: 3; and
      • (B) at least one pharmaceutically acceptable carrier or excipient.
  • In another preferred embodiment, the vaccine V comprises or consists of:
      • (A) at least one polynucleotide strand encoding for a polypeptide strand P comprising or consisting of at least nine consecutive amino acid moieties of SEQ ID NO: 2; and
      • (B) at least one pharmaceutically acceptable carrier or excipient.
  • In another preferred embodiment, the vaccine V comprises or consists of:
      • (A) at least one polynucleotide strand encoding for a polypeptide strand P comprising or consisting of at least nine consecutive amino acid moieties of SEQ ID NO: 3; and
      • (B) at least one pharmaceutically acceptable carrier or excipient.
  • Preferably, the isolated polypeptide strand P is not too short in order to provide various antigens and to improve antigenicity. In a preferred embodiment, the isolated polypeptide strand P comprises or consists of at least 10, at least 15, at least 20, at least 50 or at least 100 consecutive amino acid moieties of SEQ ID NO: 2 and/or SEQ ID NO: 3.
  • In a preferred embodiment, the polypeptide strand P comprises or consists of from 10 to 250, from 10 to 100, from 15 to 75, from 20 to 50, or from 25 to 45 consecutive amino acid moieties of SEQ ID NO: 2. In another preferred embodiment, the polypeptide strand P comprises or consists of from 10 to 250, of from 11 to 100, from 15 to 75, from 20 to 50, or from 25 to 45 consecutive amino acid moieties of SEQ ID NO: 3. In another preferred embodiment, the polypeptide strand P comprises or consists of from 10 to 250, of from 11 to 100, from 15 to 75, from 20 to 50, or from 25 to 45 consecutive amino acid moieties of SEQ ID NO: 6.
  • In a preferred embodiment, the isolated polypeptide strand P comprises (or consists of) at least 100, at least 200, at least 500, at least 1000, or at least 1800 consecutive amino acids having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the polypeptide sequence of SEQ ID NO: 2 and/or SEQ ID NO: 3.
  • In a preferred embodiment, the isolated polypeptide strand P comprises or consists of an amino acid sequence having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the polypeptide sequence of SEQ ID NO: 2.
  • In another preferred embodiment, the isolated polypeptide strand P comprises or consists of an amino acid sequence having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the polypeptide sequence of SEQ ID NO: 3.
  • In a highly preferred embodiment, the isolated polypeptide strand P consists of an amino acid sequence having the polypeptide sequence of SEQ ID NO: 2. In a highly preferred embodiment, the isolated polypeptide strand P consists of an amino acid sequence having the polypeptide sequence of SEQ ID NO: 2 obtained from heterologous expression. In a highly preferred embodiment, the isolated polypeptide strand P consists of an amino acid sequence having the polypeptide sequence of SEQ ID NO: 2 obtained from heterologous expression in a bacterium, in particular in Escherichia coli (E. coli).
  • In a highly preferred embodiment, the isolated polypeptide strand P consists of an amino acid sequence having the polypeptide sequence of SEQ ID NO: 3. In a highly preferred embodiment, the isolated polypeptide strand P consists of an amino acid sequence having the polypeptide sequence of SEQ ID NO: 3 obtained from heterologous expression. In a highly preferred embodiment, the isolated polypeptide strand P consists of an amino acid sequence having the polypeptide sequence of SEQ ID NO: 3 obtained from heterologous expression in a bacterium, in particular in Escherichia coli (E. coli).
  • In a highly preferred embodiment, the vaccine V comprises two isolated polypeptide strands P consisting of amino acid sequence having the polypeptide sequences of SEQ ID NO: 2 and SEQ ID NO: 3. In a highly preferred embodiment, the vaccine V comprises two isolated polypeptide strands P consisting of amino acid sequence having the polypeptide sequences of SEQ ID NO: 2 and SEQ ID NO: 3 each obtained from heterologous expression, preferably obtained from heterologous expression in a bacterium, in particular in Escherichia coli (E. coli).
  • In another preferred embodiment, the isolated polypeptide strand P comprises or consists of an amino acid sequence having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the polypeptide sequence of SEQ ID NO: 6.
  • In an alternative preferred embodiment, the isolated polypeptide strand P comprises or consists of an amino acid sequence having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the polypeptide sequence of SEQ ID NO: 1.
  • In an alternative preferred embodiment, the isolated polypeptide strand P comprises or consists of an amino acid sequence having amino acids of positions 1283-1516 (234 amino acids) of SEQ ID NO: 1.
  • In a preferred embodiment, the nucleotide sequence usable as antigen encodes for such polypeptide sequence of SEQ ID NO: 2 and/or SEQ ID NO: 3. In a preferred embodiment, the nucleotide sequence usable as antigen (including an indirect antigen encoding the polypeptide strand P (that may serve as antigen)) bears at least 300, at least 600, at least 1500, at least 3000, or at least 5000 consecutive nucleotides having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the sequence of SEQ ID NO: 4 and/or SEQ ID NO: 5. As indicated herein, such sequence may optionally be embedded in a larger polynucleotide sequence. In a preferred embodiment, the nucleotide sequence encodes for the polypeptide strand P (that may serve as antigen).
  • In a preferred embodiment, the nucleotide sequence encoding the polypeptide strand P bears at least 300, at least 600, at least 1500, at least 3000, or at least 5000 consecutive nucleotides having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the sequence of SEQ ID NO: 4. In a preferred embodiment, the nucleotide sequence encoding the polypeptide strand P bears at least 300, at least 600, at least 1500, at least 3000, or at least 5000 consecutive nucleotides having at least 80%, more preferably at least 90%, even more preferably at least 95%, more preferably at least 98% or homology to or even sequence identity with the sequence of SEQ ID NO: 5.
  • In a preferred embodiment, the at least one isolated polypeptide strand P consists of or comprises a peptide strand having at least 80% sequence homology to sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression. In a preferred embodiment, the at least one isolated polypeptide strand P consists of or comprises a peptide strand having at least 90% sequence homology to sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression. In a preferred embodiment, the at least one isolated polypeptide strand P consists of or comprises a peptide strand having at least 95% sequence homology to sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression. In a preferred embodiment, the at least one isolated polypeptide strand P consists of or comprises a peptide strand having at least 98% sequence homology to sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression. In a preferred embodiment, the at least one isolated polypeptide strand P consists of or comprises a peptide strand having a sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression.
  • The person skilled in the art will notice that the isolated polypeptide strand P as used in a vaccine V of the present invention may also form part of a fusion protein, a multi-antigen vaccine, or a viral vector.
  • In a highly preferred embodiment, the isolated polypeptide strand P comprises (essentially) the whole polypeptide sequence of SEQ ID NO: 2 and/or SEQ ID NO: 3. In a highly preferred embodiment, the nucleotide sequence usable as antigen comprises (essentially) the whole polypeptide sequence of SEQ ID NO: 4 and/or SEQ ID NO: 5.
  • The isolated polypeptide strand P may be obtained by any means. In order to obtain a rather pure and polypeptide one effective mean is to employ an effective (over)expression technique. The person skilled in the art is aware of numerous methods suitable for this purpose. In a preferred embodiment, the isolated polypeptide strand P is obtained from heterologous expression.
  • As used herein, “heterologous expression” may be understood in the broadest sense as expression of a gene encoding for the polypeptide according to SEQ ID NO: 2 and/or SEQ ID NO: 3 or fragment thereof in a host organism, which does not naturally have this gene or gene fragment. Insertion of the gene in the heterologous host may be performed by recombinant DNA technology which is well-known to the person skilled in the art. For this purpose, the gene may be inserted to the host, wherein it may be integrated into the host DNA causing permanent expression, or may not be integrated causing transient expression of this gene.
  • In a more preferred embodiment, the isolated polypeptide strand P is obtained from heterologous expression in bacterial or eukaryotic cells. In a highly preferred embodiment, the isolated polypeptide strand P is obtained from heterologous expression in bacterial cells such as E. coli. Such host organism comprising the gene encoding for the polypeptide strand P may also be designated as “expression system”. An advantage of such heterologous expression is that high amounts of the peptide are obtainable in good purity. In contrast to peptide synthesis, toxic agents can be avoided. On the other hand, in particular when expressed in bacteria, the peptides are typically not glycosylated.
  • In a preferred embodiment, the vaccine V of the present invention comprises or consists of:
      • (A) at least one isolated polypeptide strand P comprising or consisting of a peptide strand having at least 80% sequence homology to a sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression; and
      • (B) at least one adjuvant supporting immunologic stimulation and optionally one or more further pharmaceutically acceptable carriers.
  • In view of the further preferred embodiments described herein, it will be understood that the isolated polypeptide strand P may also have a higher homology and/or any of the other properties described herein. Further, in view of the further preferred embodiments described herein, the pharmaceutically acceptable carrier and/or the adjuvant may bear more specific properties.
  • In a preferred embodiment, the vaccine V comprises or consists of:
      • (A) at least one isolated polypeptide strand P comprising or consisting of a peptide strand of sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression; and
      • (B) at least one adjuvant supporting immunologic stimulation selected from the group consisting of alum and an immunostimulatory peptide, and optionally one or more further pharmaceutically acceptable carriers.
  • In a preferred embodiment, the vaccine V comprises or consists of:
      • (A) at least one isolated polypeptide strand P comprising or consisting of a peptide strand of sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression in bacteria; and
      • (B) a composite adjuvant supporting immunologic stimulation comprising alum and at least one immunostimulatory peptide, and optionally one or more further pharmaceutically acceptable carriers.
  • Alternatively, the polypeptide strand P may also be isolated from Plasmodium falciparum or Plasmodium vivax. This may optionally be performed by chromatographic means.
  • In a preferred embodiment, the at least one isolated polypeptide strand P comprises or consists of at least nine consecutive amino acid moieties of a sequence having at least 80% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • The sequences of SEQ ID Nos 7 to 15 are the following:
  • (SEQ ID NO: 7)
    KQEKEKEKEKEREKEKEREKEKEKEY
    (SEQ ID NO: 8)
    KNLKTELEKKEKELKDIENVSKEEINKL
    (SEQ ID NO: 9)
    SKKEKEYNQYKNTYIEEINNLNEKLEETNKEYTNLQNNYTN
    (SEQ ID NO: 10)
    KEEYEDKMNTLNEQNEDKMNSLKEEYENK
    (SEQ ID NO: 11)
    KGLKKEVEEKEHKRHSSFNILKSKEKFFKNSIEDKSHELKKKHE
    (SEQ ID NO: 12)
    KDKSKEKIKDKENQINVEKNEEKDLKKKDD
    (SEQ ID NO: 13)
    EDEKKRNLNEINNLKKKNEDMCIKYNEMN
    (SEQ ID NO: 14)
    KTNKENEEKIINLTSQYSDAYKKKSDES
    (SEQ ID NO: 15)
    SNNNIRTNEYKYEEMFDTNIEEKNG
    (SEQ ID NO: 16)
    GNISNKNENNNKKNNTCDGYDEKVT
  • These sequences are described in more detail in the experimental part below.
  • In a preferred embodiment, the at least one isolated polypeptide strand P comprises or consists of at least nine consecutive amino acid moieties of a sequence having at least 90% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • In a preferred embodiment, the at least one isolated polypeptide strand P comprises or consists of at least nine consecutive amino acid moieties of a sequence having at least 95% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • In a preferred embodiment, the at least one isolated polypeptide strand P comprises or consists of at least nine consecutive amino acid moieties of a sequence having at least 98% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • In a preferred embodiment, the at least one isolated polypeptide strand P comprises or consists of at least nine consecutive amino acid moieties of a sequence having a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • In a preferred embodiment, the above peptides may also be at least ten consecutive amino acid moieties, at least 15 consecutive amino acid moieties, at least 20 consecutive amino acid moieties, at least 25 consecutive amino acid moieties, at least 50 consecutive amino acid moieties, or at least 100 consecutive amino acid moieties as far as the respective sequence provided above is long enough.
  • In another preferred embodiment, the at least one isolated polypeptide strand P comprises or consists of a sequence having at least 80% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • In a preferred embodiment, the at least one isolated polypeptide strand P comprises or consists of a sequence having at least 90% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • In a preferred embodiment, the at least one isolated polypeptide strand P comprises or consists of a sequence having at least 95% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • In a preferred embodiment, the at least one isolated polypeptide strand P comprises or consists of a sequence having at least 98% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • In a preferred embodiment, the at least one isolated polypeptide strand P comprises or consists of a sequence having a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • In a preferred embodiment, the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising at least one peptide sequence selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • In a preferred embodiment, the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising at least two peptide sequences selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16. In a preferred embodiment, the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising at least sequences of SEQ ID NO: 7 and 8, SEQ ID NO: 7 and 9, SEQ ID NO: 7 and 10, SEQ ID NO: 7 and 11, SEQ ID NO: 7 and 12, SEQ ID NO: 7 and 13, SEQ ID NO: 7 and 14, SEQ ID NO: 7 and 15, SEQ ID NO: 7 and 16, SEQ ID NO: 8 and 9, SEQ ID NO: 8 and 10, SEQ ID NO: 8 and 11, SEQ ID NO: 8 and 12, SEQ ID NO: 8 and 13, SEQ ID NO: 8 and 14, SEQ ID NO: 8 and 15, SEQ ID NO: 8 and 16, SEQ ID NO: 9 and 10, SEQ ID NO: 9 and 11, SEQ ID NO: 9 and 12, SEQ ID NO: 9 and 13, SEQ ID NO: 9 and 14, SEQ ID NO: 9 and 15, SEQ ID NO: 9 and 16, SEQ ID NO: 10 and 11, SEQ ID NO: 10 and 12, SEQ ID NO: 10 and 13, SEQ ID NO: 10 and 14, SEQ ID NO: 10 and 15, SEQ ID NO: 10 and 16, SEQ ID NO: 11 and 12, SEQ ID NO: 11 and 13, SEQ ID NO: 11 and 14, SEQ ID NO: 11 and 15, SEQ ID NO: 11 and 16, SEQ ID NO: 12 and 13, SEQ ID NO: 12 and 14, SEQ ID NO: 12 and 15, SEQ ID NO: 12 and 16, SEQ ID NO: 13 and 14, SEQ ID NO: 13 and 15, SEQ ID NO: 13 and 16, SEQ ID NO: 14 and 15, SEQ ID NO: 14 and 16, or SEQ ID NO: 15 and 16.
  • As used throughout the present invention, the term “truncated version of SEQ ID NO: 2” means a peptide comprising or consisting of a fraction of SEQ ID NO: 2 truncated by at least one amino acid moiety in length. In other words, in a t truncated version of SEQ ID NO: 2, at least one amino acid moiety is missing. Even if the truncated peptide sequence is extended by one or more amino acid moieties, these are not the same as those of SEQ ID NO: 2.
  • In a preferred embodiment, a truncated version of SEQ ID NO: 2 comprises or consists of a fraction of SEQ ID NO: 2 truncated by at least two, at least three, at least five, at least ten, at least 20, at least 50, at least 75, at least 100, at least 200, at least 500, or at least 1000 amino acid moieties in length. In a preferred embodiment, the polypeptide strand P which is a truncated version of SEQ ID NO: 2 comprises or consists of from 32 to 1900, from 40 to 1800, from 50 to 1700, from 60 to 1600, from 70 to 1500, from 80 to 1400, from 90 to 1300, from 100 to 1200, from 200 to 1100, from 300 to 1000 amino acid moieties in length.
  • In a preferred embodiment, a truncated version of SEQ ID NO: 2 comprises or consists of a fraction of SEQ ID NO: 2 truncated by at least 100 amino acid moieties in length.
  • In a preferred embodiment, the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising or consisting of a fraction of SEQ ID NO: 2 truncated by at least 100 amino acid moieties in length and comprising at least two peptide sequences selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • In a preferred embodiment, the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising or consisting of a fraction of SEQ ID NO: 2 truncated by at least 100 amino acid moieties in length and comprising all of the peptide sequences SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • In a preferred embodiment, the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising at least three peptide sequences selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • In a preferred embodiment, the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising at least four peptide sequences selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16. In a preferred embodiment, the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising at least five peptide sequences selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16. In a preferred embodiment, the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising at least six, at least seven, at least eight, or least nine, peptide sequences selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • In a preferred embodiment, the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising all of the peptide sequences SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • As indicated herein, the polypeptide strand P may also be obtained by means of heterologous expression. It has been surprisingly found that also polypeptide strands P which were obtained from heterologous expression in bacteria led to a remarkable effectivity. Therefore, also non-glycosylated peptide strands P showed activity as experimentally evidenced.
  • Accordingly, in a preferred embodiment, the polypeptide strand P does not comprise (i.e., is (essentially) free of glycosylation.
  • Alternatively, it will be understood that glycosylated peptides are, however, also suitable. Therefore, in an alternative preferred embodiment, the polypeptide strand P comprises one or more glycosylated sides.
  • In a preferred embodiment, the polypeptide strand P comprises or consists of an easily assessable epitope. Accordingly, it preferably bears a sequence as laid out above. This is evidenced further by bioinformatic means as laid out in the experimental section below. In a preferred embodiment, the polypeptide strand P is not a hardly assessable sequence.
  • Thus, in a preferred embodiment, in particular when the polypeptide strand P is not the full-length protein (of any of SEQ ID NOs: 2 or 3), the polypeptide strand P is not and/or comprises no peptide strand that has a length of nine or more consecutive amino acid moieties of at least 80% homology of any of SEQ ID NOs: 17-24. In a preferred embodiment, in particular when the polypeptide strand P is not the full-length protein (of any of SEQ ID NOs: 2 or 3), the polypeptide strand P is not and/or comprises no peptide strand that has a length of nine or more consecutive amino acid moieties of at least 80% homology of any of SEQ ID NOs: 17-21.
  • In a preferred embodiment, in particular when the polypeptide strand P is not the full-length protein (of any of SEQ ID NOs: 2 or 3), the polypeptide strand P is not and/or comprises no peptide strand that has a length of nine or more consecutive amino acid moieties of at least 98% homology of any of SEQ ID NOs: 17-21. In a preferred embodiment, in particular when the polypeptide strand P is not the full-length protein (of any of SEQ ID NOs: 2 or 3), the polypeptide strand P is not and/or comprises no peptide strand that has a length of nine or more consecutive amino acid moieties of at least 98% homology of any of SEQ ID NOs: 17-24. In a preferred embodiment, in particular when the polypeptide strand P is not the full-length protein (of any of SEQ ID NOs: 2 or 3), the polypeptide strand P is not and/or comprises no peptide strand that has a length of nine or more consecutive amino acid moieties of any of SEQ ID NOs: 17-21. In a preferred embodiment, in particular when the polypeptide strand P is not the full-length protein (of any of SEQ ID NOs: 2 or 3), the polypeptide strand P is not and/or comprises no peptide strand that has a length of nine or more consecutive amino acid moieties of any of SEQ ID NOs: 17-24.
  • In a preferred embodiment, the polypeptide strand P is not a sequence having at least 80% sequence homology to a peptide sequence selected from the group consisting of one or more of SEQ ID NOs: 17-24. In a preferred embodiment, the polypeptide strand P is not a peptide strand of a sequence having at least 80% sequence homology to all of SEQ ID NOs: 17-21. In a preferred embodiment, the polypeptide strand P is not a peptide strand of a sequence having at least 80% sequence homology to all of SEQ ID NOs: 17-24.
  • SEQ ID NOs: 17-24 refer to the following sequences:
  • (SEQ ID NO: 17)
    IKTMNTQISTLKNDVHLLNEQDKLNNEKGTLNSKISELNVQI
    MDL
    (SEQ ID NO: 18)
    LLSKDKEIEEKNKKIKELNNDIKKL
    (SEQ ID NO: 19)
    ICSLTTEVMELNNKKNELIEENNKLNLVDQGKKKLKKDVEK
    QKKEIEKL
    (SEQ ID NO: 20)
    VDKIEEHILDYDEEINKSRSNLFQLKNEICSLTTEVMELNNKK
    NELIEENNKLNLVDQGKKKLKKDVEKQKKEIEKL
    (SEQ ID NO: 21)
    LDENEDNIKKMKSKIDDMEKEIKYR
    (SEQ ID NO: 22)
    TISSLSNKIVNYESKIEELEKELKEVK
    (SEQ ID NO: 23)
    IIDIKKHLEKLKIEIKEKKEDLENL
    (SEQ ID NO: 24)
    IKTMNTQISTLKNDVHLLNEQDKLNNEKGTLNSKISEL
  • In a preferred embodiment, the polypeptide strand P does not have a sequence of SEQ ID NOs: 17-21. In a preferred embodiment, the polypeptide strand P does not have a sequence of SEQ ID NOs: 17-24.
  • A vaccine V based on a polynucleotide strand encoding for the polypeptide strand P may be any kind of polynucleotide. In a preferred embodiment, the polynucleotide strand encoding for said polypeptide strand P is double or single stranded DNA or double or single stranded RNA, or an analogue of double or single stranded DNA or double or single stranded RNA.
  • Such nucleotide analogue may exemplarily comprise or even consist of nucleotide analogues such as, e.g., peptide nucleic acid (PNA), Morpholino and locked nucleic acid (LNA), as well as glycol nucleic acid (GNA) and threose nucleic acid (TNA).
  • In a preferred embodiment, the polynucleotide strand encoding for said polypeptide strand P is a plasmid.
  • In a preferred embodiment, the at least one polynucleotide strand encoding for the polypeptide strand P is a polynucleotide strand encoding for a sequence having at least 80% sequence homology, at least 90% sequence homology, at least 95% sequence homology, or at least 98% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • In a preferred embodiment, the at least one polynucleotide strand encoding for the polypeptide strand P is a polynucleotide strand encoding for a sequence having a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
  • As indicated above, it will be understood that the vaccine V of the present invention is well suitible for preventing malaria in a patient.
  • Accordingly, as indicated above, a further aspect of the present invention relates to the vaccine V of the present invention for use in a method for preventing malaria in a patient.
  • In other words, the present invention relates to a method for preventing malaria in a patient, wherein a sufficient amount of the vaccine V is administered to said patient. This method may be understood as active immunization.
  • As throughout the present invention, malaria is preferably malaria caused by Plasmodium falciparum or Plasmodium vivax. The person skilled in eth art will notice that the respective vaccine V (comprising at least one isolated polypeptides P or at least one nucleotide encoding for such) will provide at least partly protection against Plasmodia of the respective type. A vaccine V comprising at least one isolated polypeptides P of a sequence homolog to Plasmodium falciparum or at least one nucleotide encoding for such will particularly prevent the patient from malaria caused by Plasmodium falciparum.
  • A vaccine V comprising at least one isolated polypeptides P of a sequence homolog to Plasmodium vivax or at least one nucleotide encoding for such will particularly prevent the patient from malaria caused by Plasmodium vivax.
  • As used herein, “preventing” does not necessarily means that the disease is completely prohibited, but means, in the broadest sense, that the symptoms of malaria are diminish, when the patient is exposed to the respective plasmodia (e.g., Plasmodium falciparum and/or Plasmodium vivax (and/or, regarding rodents, Plasmodium chabaudi)). The effect of diminishing symptoms is the reduction in comparison to a comparable patient exposed to a comparable amount of the respective plasmodia (e.g., Plasmodium falciparum and/or Plasmodium vivax (and/or, regarding rodents, Plasmodium chabaudi)) without administration of the vaccine V.
  • As used herein, a “sufficient amount” may be understood in the broadest sense as an amount that is suitible to provoke an immune response against plasmodia. Preferably, a sufficient amount vaccine V is suitible to diminish the quantity of the respective plasmodia (e.g., Plasmodium falciparum and/or Plasmodium vivax (and/or, regarding rodents, Plasmodium chabaudi)) when the patient is exposed to plasmodia.
  • In a preferred embodiment, at each administration, the patient is administered with an amount of an isolated polypeptide strand P in the range of from 0.1 μg/kg to 1 g/kg, of from 1 μg/kg to 100 mg/kg, of from 5 μg/kg to 50 mg/kg, of from 10 μg/kg to 25 mg/kg, of from 25 μg/kg to 20 mg/kg, of from 50 μg/kg to 10 mg/kg, or of from 100 μg/kg to 5 mg/kg.
  • In a preferred embodiment, the patient is administered with a sufficient amount of the vaccine V to diminish the symptoms of malaria or even to prohibit the outbreak of malaria completely. In other words, this may be sufficient to confer protection.
  • The patient may be administered with the vaccine V of the present invention once, twice, three times of more often. The patient may also be administered on a regular basis (e.g., once per month, once per two months, one per three months, once per half a year, once per year or once every five years).
  • In a preferred embodiment, the patient is administered only once. This may already be sufficient for developing an immune response to diminish the quantity of the respective plasmodia (e.g., Plasmodium falciparum and/or Plasmodium vivax (and/or, regarding rodents, Plasmodium chabaudi)) when the patient is exposed to plasmodia. This may diminish the symptoms of malaria or even to prohibit the outbreak of malaria completely. In other words, this may be sufficient to confer protection.
  • In a preferred embodiment, the vaccine V is administered at least once to the patient before exposure to the respective plasmodia (e.g., Plasmodium falciparum and/or Plasmodium vivax (and/or, regarding rodents, Plasmodium chabaudi)). In another preferred embodiment, the vaccine V is administered to the patient during and/or after exposure to the respective plasmodia. In a preferred embodiment, the vaccine V is administered at least once to the patient before exposure to the respective plasmodia and during and/or after exposure to the respective plasmodia
  • As indicated above, the vaccine V of the present invention may be very well be used for generating antibodies binding to Plasmodium falciparum or Plasmodium vivax. As used in the context of antibodies, terms like “binding to”, “directed to”, “targeted” or the like may be understood interchangeably in the broadest sense as interacting selectively and non-covalently with a binding affinity of a dissociation constant (Kd) of 1000 nM or less. Accordingly, the present invention also refers to the generation of antibodies by means of a vaccine V of the present invention. In other words, the present invention also refers to the use of a vaccine V of the present invention for preparing an antibody AB binding to Plasmodium falciparum or Plasmodium vivax , in particular, binding to the repetitive organellar protein, putative of Plasmodium falciparum 3D7 or the hypothetical protein PVNG_04523 of Plasmodium vivax North Korean.
  • Accordingly, a further aspect of the present invention refers a method for preparing an antibody AB, binding to Plasmodium falciparum or Plasmodium vivax comprising the following steps:
      • (i) providing:
        • (a) a vaccine V according to the present invention, and
        • (b) an organism O suitible for generating antibodies;
      • (ii) administering the organism O with the vaccine V;
      • (iii) waiting until the subjected organism of step (ii) shows an immune response against the antigens of the vaccine V;
      • (iv) obtaining antibody-generating cells C of the organism O of step (iii);
      • (v) optionally hybridizing the antibody-generating cells of step (iv) with myeloma cells obtaining immortalized antibody-generating cells C1;
      • (vi) optionally isolating the nucleotide encoding for the antibody AB of interest and transfer it to another antibody-generating cell type C2 suitible for expressing the antibody AB;
      • (vii) cultivating the antibody-generating cells C, C1 or C2 of any of steps (iv) to
      • (vi) under conditions enabling the production of the antibody AB; and
      • (viii) isolating the antibody from step (vii).
  • In the context of such method, the organism O may be any organism suitible for generating antibodies. Preferably, the organism is a (typically non-human) mammalian. Exemplarily, the organism O may be selected from the group consisting of a mouse, a rat, a rabbit, a goat, a hamster, a donkey, a cow, a pig, or a camel.
  • Preferably, administration of step (ii) is systemic administration (e.g., intravenously (i.v.), intraarterially (i.a.), intraperitoneally (i.p.), intramusculary (i.m.), subcutaneously (s.c.), transdermally, nasally). Alternatively, administration may also be local administration (e.g., intrathecally or intravitreally). Preferably, administration is systemic administration, in particular intravenous injection.
  • The waiting time depends on the vaccine V, the species of the organism O and the like. Typically it will take several days up to several weeks until an immune response is obtained. Exemplarily, the waiting time may be between 1 day and twenty weeks, preferably between 2 days and ten weeks, more preferably between 3 days and five weeks, exemplarily between 4 days and three weeks. During this time, the animals are kept under suitable conditions for maintain health for this species.
  • Obtaining antibody-generating cells C of the organism O of step (iii) may be performed by any means known for this purpose in the art. Optionally, the cells C may be obtained from the blood of the immunized organism O. Alternatively, the cells C may be obtained from the lymph and/or spleen of the immunized organism O. Depending on the method used the organism O may be kept alive or may be sacrificed.
  • The antibodies AB of interest may be directly obtained from the cells C or may be obtained from further processed cells.
  • In a preferred embodiment, the method comprised the further step (v) of hybridizing the antibody-generating cells of step (iv) with myeloma cells obtaining immortalized antibody-generating cells C1. This may enable obtaining monoclonal antibodies. The person skilled in the art is well aware of methods usable for preparing such hybridoma cells C1.
  • In a preferred embodiment, the method comprised the further step (vi) of isolating the nucleotide encoding for the antibody AB of interest and transfer it to another antibody-generating cell type C2 suitible for expressing the antibody AB. This may enable heterologous expression of the antibody AB or fragments thereof. Optionally, also the Fc part of the antibody may be altered. Exemplarily, the antibody AB may than be humanized. The person skilled in the art is well aware of methods usable for this purpose. Further, the person skilled in the art will notice that the optional steps (v) and (vi) may optionally also be combined with another.
  • In any case, the antibody-generating cells C, C1 or C2 of any of steps (iv) to (vi) are cultivated under conditions enabling the production of the antibody AB followed by isolating the antibody AB. Such isolation step may be performed by any means such as, e.g., by means of chromatographic means such as affinity chromatography using a stationary phase comprising the antigen of the antibody AB.
  • In a further aspect, the present invention also embraces each of the antibody-generating cells C, C1 or C2.
  • It will be noted that the present invention also refers to an anti-repetitive organellar protein, putative of Plasmodium falciparum 3D7 or anti-hypothetical protein PVNG_04523 of Plasmodium vivax North Korean antibody.
  • Accordingly, a still further aspect of the present invention refers to an antibody or antibody fragment AB binding a polypeptide strand P of SEQ ID NO: 2 or SEQ ID NO: 3 or a polynucleotide strand encoding for said polypeptide strand P with a dissociation constant of not more than 1000 nM.
  • Preferably, the antibody or antibody fragment AB binding a polypeptide strand P of SEQ ID NO: 2 or SEQ ID NO: 3 or a polynucleotide strand encoding for said polypeptide strand P with a dissociation constant of not more than 100 nM, more preferably not more than 100 nM, even more preferably not more than 50 nM or not more than 20 nM.
  • As used in the context of the present invention, the term “antibody” may be understood in the broadest sense as any type of immunoglobulin or antigen-binding fraction or mutant thereof known in the art.
  • Exemplarily, the antibody of the present invention may be an immunoglobulin A (IgA), immunoglobulin D (IgD), immunoglobulin E (IgE), immunoglobulin G (IgG), immunoglobulin M (IgM), immunoglobulin Y (IgY) or immunoglobulin W (IgW). Preferably, the antibody is an IgA, IgG or IgD. More preferably, the antibody is an IgG. However, it will be apparent that the type of antibody may be altered by biotechnological means by cloning the gene encoding for the antigen-binding domains of the antibody of the present invention into a common gene construct encoding for any other antibody type.
  • The binding between the antibody and its molecular target structure (i.e., its antigen based on the polypeptide strand P of SEQ ID NO: 2 or SEQ ID NO: 3 or a polynucleotide strand encoding for said polypeptide strand P) typically is a non-covalent binding. Preferably, the binding affinity of the antibody to its antigen has a dissociation constant (Kd) of less than 1000 nM, less than 500 nM, less than 200 nM, less than 100 nM, less than 50 nM, less than 40 nM, less than 30 nM or even less than 20 nM.
  • The term “antibody” as used herein may be understood in the broadest sense and also includes what may be designated as an antibody mutant. As used in the context of the present invention, an antibody mutant may be understood in the broadest sense as any antibody mimetic or antibody with altered sequence known in the art. The antibody mutant may have at least 10%, at least 20%, at least 50%, at least 60%, at least 70%, at least 80%, at least 85%, at least 90% or at least 95% of the binding affinity of a corresponding antibody, i.e., bear a dissociation constant (Kd) of less than 10 μM, less than 1 μM, less than 500 nM, less than 200 nM, less than 100 nM, less than 50 nM, less than 40 nM, less than 30 nM or even less than 20 nM.
  • As used herein, the term “antibody fragment” may be understood in the broadest sense as any fragment of an antibody that still bears binding affinity to its molecular target (i.e., its antigen based on the polypeptide strand P of SEQ ID NO: 2 or SEQ ID NO: 3 or a polynucleotide strand encoding for said polypeptide strand P). Exemplarily, the antibody fragment may be a fragment antigen binding (Fab fragment), Fc, F(ab′)2, Fab′, scFv, a truncated antibody comprising one or both complementarity determining region(s) (CDR(s)) or the variable fragment (Fv) of an antibody. Variable domains (Fvs) are the smallest fragments with an intact antigen-binding domain consisting of one VL and one VH. Such fragments, with only the binding domains, can be generated by enzymatic approaches or expression of the relevant gene fragments, e.g. in bacterial and eukaryotic cells. Different approaches can be used, e.g. either the Fv fragment alone or ‘Fab’-fragments comprising one of the upper arms of the “Y” that includes the Fv plus the first constant domains. These fragments are usually stabilized by introducing a polypeptide link between the two chains which results in the production of a single chain Fv (scFv). Alternatively, disulfide-linked Fv (dsFv) fragments may be used. The binding domains of fragments can be combined with any constant domain in order to produce full length antibodies or can be fused with other proteins and polypeptides. A recombinant antibody fragment is the single-chain Fv (scFv) fragment. Dissociation of scFvs results in monomeric scFvs, which can be complexed into dimers (diabodies), trimers (triabodies) or larger aggregates such as TandAbs and Flexibodies. The antibody may be a Fab, a Fab′, a F(ab′)2, a Fv, a disulfide-linked Fv, a scFv, a (scFv)2, a bivalent antibody, a bispecific antibody, a multispecific antibody, a diabody, a triabody, a tetrabody or a minibody.
  • As mentioned above, the term “antibody” may also include an antibody mimetic which may be understood in the broadest sense as organic compounds that, like antibodies, can specifically bind antigens and that typically have a molecular mass in a range of from approximately 3 kDa to approximately 25 kDa. Antibody mimetics may be, e.g., affibody molecules (affibodies), affilins, affitins, anticalins, avimers, DARPins, Fynomers, Kunitz domain peptides, single-domain antibodies (e.g., VHH antibodies or VNAR antibodies, nanobodies), monobodies, diabodies, triabodies, flexibodies and tandabs. The antibody mimetics may be of natural origin, of gene technologic origin and/or of synthetical origin. The antibody mimetics may also include polynucleotide-based binding units. Optionally, the antibody may also be a CovX-body. Optionally, the antibody may also be a cameloid species antibody.
  • In a preferred embodiment, the antibody or antibody fragment AB binds to its antigen based on the polypeptide strand P of SEQ ID NO: 2 or SEQ ID NO: 3 or a polynucleotide strand encoding for said polypeptide strand P with an at least 10-fold, even more preferably at least 100-fold, even more preferably at least 1000-fold higher binding affinity than to the corresponding (i.e., sequence homologue) antigen of a polypeptide SEQ ID NO: 2 or SEQ ID NO: 3 or the respective polynucleotide strand encoding for said polypeptide.
  • The antibody or antibody fragment AB may be obtained by any means. In a preferred embodiment, the antibody or antibody fragment AB is obtained from a method of the present invention.
  • Optionally, the antibody of the present invention may be a monoclonal antibody, a chimeric antibody and/or a humanized antibody. Monoclonal antibodies are monospecific antibodies that are identical because they are produced by one type of immune cell that are all clones of a single parent cell. A chimeric antibody is an antibody in which at least one region of an immunoglobulin of one species is fused to another region of an immunoglobulin of another species by genetic engineering in order to reduce its immunogenicity. For example murine VL and VH regions may be fused to the remaining part of a human immunoglobulin. A particularly preferred type of chimeric antibodies are humanized antibodies. Humanized antibodies are produced by merging the DNA that encodes the CDRs of a non-human antibody with human antibody-producing DNA. The resulting DNA construct can then be used to express and produce antibodies that are usually not as immunogenic as the non-human parenteral antibody or as a chimeric antibody, since merely the CDRs are non-human.
  • In a preferred embodiment, the antibody or antibody fragment AB is a monoclonal humanized antibody.
  • Preferably, the antibody or antibody fragment AB bears a high affinity to the polypeptide strand P of SEQ ID NO: 2 or SEQ ID NO: 3. In a preferred embodiment, the antibody or antibody fragment AB bears a binding affinity to a polypeptide strand P of SEQ ID NO: 2 or SEQ ID NO: 3 of a dissociation constant of not more than 100 nM, not more than 50 nM, not more than 20 nM, not more than 10 nM, or not more than 5 nM.
  • The antibody or antibody fragment AB may be provided and stored in any suitable form. The antibody or antibody fragment AB, independent on its chemical nature, may optionally be dissolved in any medium suitable for storing said antibody such as, e.g., water, an aqueous buffer (e.g., a Hepes, Tris, or phosphate buffer (e.g. phosphate buffered saline (PBS)), an organic solvent (e.g., dimethyl sulfoxide (DMSO), dimethylformide (DMF)) or a mixture of two or more thereof. The antibody or mutant thereof according to the present invention may be of any species or origin. It may bind to any epitope(s) comprised by its molecular target structure (e.g., linear epitope(s), structural epitope(s), primary epitope(s), secondary epitope(s), e.g., based on SEQ ID NO: 2 or SEQ ID NO: 3 or a nucleotide encoding therefor). Preferably, the antibody or mutant thereof may recognize the naturally folded molecular target structure or a domain or fragment thereof (e.g., SEQ ID NO: 2 or SEQ ID NO: 3 or a nucleotide encoding therefor in its natural environment inside the plasmodia). The antibody or mutant thereof may be of any origin an antibody may be obtained from such as, e.g., natural origin, a gene technologic origin and/or a synthetic origin. Optionally, the antibody may also be commercially available. The person skilled in the art will understand that the antibody may further comprise one or more posttranscriptional modification(s) and/or may be conjugated to one or more further structures such as label moieties or cell-penetrating peptides (CPPs). Optionally, the antibody or antibody fragment may be added to a support, particularly a solid support such as an array, bead (e.g. glass or magnetic), a fiber, a film etc. The skilled person will be able to adapt the antibody of the present invention and a further component to the intended use by choosing a suitable further component.
  • Optionally, the antibody or antibody fragment AB may be conjugated with any kind of detectable label moiety. Then, the antibody or antibody fragment AB and the label moiety may be covalently or non-covalently conjugated with another, either directly of via a spacer.
  • As used throughout the present invention, the term “conjugated with” may be understood in the broadest sense as any kind of covalent or non-covalent attachment or linkage of one component with another component. Such conjugate can be obtained by chemical means and/or by genetic engineering and biotechnological means. The label moiety may be any moiety that is detectable, preferably detectable by an imaging method.
  • Exemplarily, the label moiety may be a fluorescent moiety. Exemplarily, a fluorescent moiety may be a fluorescent polypeptide moiety (e.g., cyan fluorescent protein (CFP), green fluorescent protein (GFP) or yellow fluorescent protein (YFP), red fluorescent protein (RFP), mCherry, etc.), a small-molecule dye moiety (e.g., an Atto dye moiety (e.g., ATTO 390, ATTO 425, ATTO 465, ATTO 488, ATTO 495, ATTO 520, ATTO 532, ATTO 550, ATTO 565, ATTO 590, ATTO 594, ATTO 610, ATTO 611X, ATTO 620, ATTO 633, ATTO 635, ATTO 637, ATTO 647, ATTO 647N, ATTO 655, ATTO 665, ATTO 680, ATTO 700, ATTO 725, ATTO 740), a Cy dye moiety (e.g., Cy3, Cy5, Cy5.5, Cy 7), an Alexa dye moiety (e.g., Alexa Fluor 488, Alexa Fluor 546, Alexa Fluor 647, Alexa Fluor 680, Alexa Fluor 750), a VisEn dye moiety (e.g. VivoTag680, VivoTag750), an S dye (e.g., S0387), a DyLight fluorophore moiety (e.g., DyLight 750, DyLight 800), an IRDye moiety (e.g., IRDye 680, IRDye 800), a fluorescein dye moiety (e.g., fluorescein, carboxyfluorescein, fluorescein isothiocyanate (FITC)), a rhodamine dye moiety (e.g., rhodamine, tetramethylrhodamine (TAMRA)), a HOECHST dye moiety, a quantum dot moiety or a combination of two or more thereof. Such fluorescent label moiety may be used in fluorescence microscopy.
  • Alternatively or additionally, the label moiety may be metal atom, metal ion or metal bead (e.g., a (colloidal) gold such as a gold bead). Such metal bead may be used in electron microscopy.
  • Alternatively or additionally, the label moiety may be radioactive label such as, e.g., 3H, 14C, 123I, 124I, 131I, 32P, 99mTc or lanthanides (e.g., 64Gd). In this context, a radioactive label may or may not be suitable for scintillation assays, computer tomography (CT), single-photon emission computed tomography (SPECT) or as a label suitable for Positron Emission Tomography (PET) (e.g., 11 C, 13 N, 15O, 18F, 82Rb).
  • As indicated above, the present invention also refers to the medicinal and non-medicinal uses of the antibody or antibody fragment AB of the present invention.
  • Accordingly, a still further aspect of the present invention refers to an antibody or antibody fragment AB of the present invention for use in a method for treating or preventing malaria in a patient.
  • The present invention refers to a method for treating or preventing malaria in a patient, said method comprising administration of antibody or antibody fragment AB of the present invention to said patient in an amount suitible for treating or preventing malaria in said patient.
  • Preferably, administration is systemic administration (e.g., intravenously (i.v.), intraarterially (i.a.), intraperitoneally (i.p.), intramusculary (i.m.), subcutaneously (s.c.), transdermally, nasally). Alternatively, administration may also be local administration (e.g., intrathecally or intravitreally). Preferably, administration is systemic administration, in particular intravenous injection. The administration frequency may be adapted to the individual patient. Administration may be performed once, twice, or more often or continuously (e.g., via drip). Exemplarily, administration may be performed three times daily, twice daily, or every two days or less often.
  • A still further aspect of the present invention refers to an antibody or antibody fragment AB for use in a method for diagnosing malaria in a patient.
  • In other words, the present invention refers to a method for diagnosing malaria in a patient, said method comprising administration of antibody or antibody fragment AB of the present invention to said patient in an amount suitible for detecting Plasmodium falciparum or Plasmodium vivax in said patient.
  • For detecting the antibody or antibody fragment AB used for diagnostic purposes, the antibody or antibody fragment AB is preferably labelled, i.e., conjugated to a label moiety, as described above.
  • The antibody or antibody fragment AB according to the present invention may also be used for staining in vitro.
  • For example, detection may be performed by enzyme-linked immunosorbent assay (ELISA), flow cytometry, fluorescence activated cell sorting (FACS), magnetic activated cell sorting (MACS), antibody-based dipsticks, etc.
  • Accordingly, a further aspect of the present invention relates to a method for staining Plasmodium falciparum or Plasmodium vivax in vitro, said method comprising the steps of:
      • (i) providing
        • (a) an optionally fixed sample S containing Plasmodium falciparum or Plasmodium vivax, and
        • (b) an optionally stained antibody or antibody fragment AB according to the present invention;
      • (ii) contacting the sample S with the optionally stained antibody or antibody fragment AB; and
      • (iii) optionally contacting the treated sample of step (ii) with a second antibody or antibody fragment AB2 which is stained and selectively binds to the antibody or antibody fragment AB.
  • When at least one of AB1 or AB2 is fluorescently labelled, the sample may be investigated by fluorescence microscopy of flow cytometry. Exemplarily, fluorescence microscopy may comprise one or more of the following methods: laser scanning microscopy (LSM), two-photon fluorescence microscopy, fluorescence molecular imaging (FMI), fluorescence energy transfer (FRET), fluorescence correlation spectroscopy (FCS), and/or fluorescence cross-correlation spectroscopy (FCCS). All these techniques as such are well-known to those skilled in the art. In the imaging step, the polypeptide strand P of SEQ ID NO: 1, SEQ ID NO: 2, SEQ IND NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16 or nucleotide encoding for at least one thereof may be fluorescently stained by a fluorescently labeled antibody AB1 or unlabeled AB1 in combination with labelled AB2. The excess fluorescently labeled antibody may be washed away and the localization and intensity is determined spatially resolved in the sample.
  • A still further aspect of the present invention refers to the (preferably in vitro) use of any of the polypeptides of the present invention as described herein (e.g. a polypeptide selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and/or SEQ ID NO: 16) for detecting Plasmodium falciparum or Plasmodium vivax in a patient's body fluid.
  • In a preferred embodiment, patient's body fluid is blood, blood plasm, blood serum or a fraction thereof.
  • A still further aspect of the present invention refers to a method of detecting Plasmodium falciparum or Plasmodium vivax (preferably in vitro), said method comprising the step of detecting the presence of any of the polypeptides of the present invention as described herein (e.g. a polypeptide selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and/or SEQ ID NO: 16) in a patient's body fluid. A further step may optionally be treating malaria in the patient accordingly.
  • A still further aspect of the present invention refers to a polypeptide of the present invention as described herein (e.g. a polypeptide selected from the group consisting of (SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and/or SEQ ID NO: 16) for use in a method for diagnosing a Plasmodium falciparum or Plasmodium vivax infection in a patient. Optionally the patient is further subjected to a malaria treatment.
  • A still further aspect of the present invention refers to a method of detecting Plasmodium falciparum or Plasmodium vivax (preferably in vitro), said method comprising the step of detecting the presence of an antibody specific for any of the polypeptides of the present invention as described herein (e.g. a polypeptide selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and/or SEQ ID NO: 16) in a patient's body fluid. A further step may optionally be treating malaria in the patient accordingly.
  • A still further aspect of the present invention refers to an antibody specific for any of the polypeptides of the present invention as described herein (e.g. a polypeptide selected from the group consisting of (SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and/or SEQ ID NO: 16) for use in a method for diagnosing a Plasmodium falciparum or Plasmodium vivax infection in a patient. Optionally the patient is further subjected to a malaria treatment.
  • A still further aspect of the present invention refers to a primer that is complementary to a fraction of any of SEQ ID NO: 4, SEQ ID NO: 5, or SEQ ID NO: 6 for detecting Plasmodium falciparum or Plasmodium vivax in a patient's body fluid.
  • In a preferred embodiment, the primer is a single chain nucleotide strand of a length of between five to 50 nucleotide moieties in length, between seven to 40 nucleotide moieties in length, between eight to 35 nucleotide moieties in length, or between nine to 30 nucleotide moieties in length.
  • A still further aspect of the present invention refers to a pair of primers that are both complementary to a fraction of any of SEQ ID NO: 4, SEQ ID NO: 5, or SEQ ID NO: 6 for detecting Plasmodium falciparum or Plasmodium vivax in a patient's body fluid.
  • A still further aspect of the present invention refers to a method of detecting Plasmodium falciparum or Plasmodium vivax (preferably in vitro) in a patient, said method comprising the steps of
      • (i) contacting the nucleotides contained in a patient's body fluid with a pair of primers of which at least one is complementary to a fraction of any of SEQ ID NO: 4, SEQ ID NO: 5, or SEQ ID NO: 6;
      • (ii) conducting polymerase chain reaction (PCR) with the sample obtained in step (i); and
      • (iii) detecting the presence or absence of amplification product comprising the primers; and
      • (iv) optionally treating malaria in the patient accordingly.
  • A still further aspect of the present invention refers to a method of detecting Plasmodium falciparum or Plasmodium vivax (preferably in vitro) in a patient, said method comprising the steps of
      • (i) contacting the nucleotides contained in a patient's body fluid with a pair of primers which are both complementary to a fraction of any of SEQ ID NO: 4, SEQ ID NO: 5, or SEQ ID NO: 6;
      • (ii) conducting polymerase chain reaction (PCR) with the sample obtained in step (i); and
      • (iii) detecting the presence or absence of amplification product comprising the primers; and
      • (iv) optionally treating malaria in the patient accordingly.
  • The Examples provided below and the claims illustrate further embodiments of the present invention.
  • EXAMPLES Example 1 Protective Effect of the Immunization with Pch ROPE
      • Immunization experiment with Pch ROPE in Balb/c mice.
  • A cDNA library of a Plasmodium chabaudi 96V was constructed in pBluescript SK (Werner et al. 1998). From the cDNA library the recombinant plasmid pBluescript SK (-) 70 was isolated by screening. It contains part of the Pch ROPE sequence coding for amino acids 1283-1516 (234 amino acids) as an insert.
  • The insert was sub cloned in the pGEX-1T vector, transformed in E. coli JM 109 bacteria and expressed as a glutathione-S-transferase fusion protein in E. coli JM 109. The fusion protein was isolated using glutathione-agarose beads, followed by thrombin cleavage to obtain the 234 amino acid Pch ROPE protein fragment in a pure form, called rec 700.
  • Mice were immunized at day 0 and day 21 with:
  • 7.5 μg rec 700
  • 60 μg Alum
  • in 200 μl PBS pH 7.4
  • At day 35, mice were infected with 5×106 Plasmodium chabaudi 96V parasitized erythrocytes. Balb/c mice were immunized only two times and without any supplementation of an adjuvant such as muramyl peptide (MDP).
  • Results
  • All of the non-immunized mice died. 50% of the immunized Balb/c survived and 50% died. The fact that a high number of Balb/c mice survived an otherwise deadly infection with this extremely virulent Plasmodium chabaudi 96V strain using a weak immunization protocol, was surprising.
  • It is noteworthy in this context to mention that in other experiments, not a single untreated mouse survived an infection with the Plasmodium chabaudi 96V strain over a period of about four years of conducting regular experiments with this strain (Watier et al., 1992).
  • Particularly surprising was the finding that a protective effect of the immunization could still be observed when adjuvants like muramyl peptide (MDP) were omitted. Such adjuvants are typically considered to be a relevant component to illicit a stronger immune response against an antigen used as a vaccine.
  • Furthermore it was recently shown that, in the case of malaria, alum (used in above immunization) is a comparably poor adjuvant for fighting diseases like malaria (Leslie, 2013). This shows that ROPE is a particularly effective immunogen. It is indicating a very high protection capacity of the ROPE protein when used as a malaria vaccine. Moreover, ROPE is very effective for generating specific antibodies.
  • It will be understood that an infection with Plasmodium chabaudi in rodents is a well-established model system for infections with Plasmodium falciparum or Plasmodium vivax in humans. For this reason, it is reasonable to assume that ROPE has a protective effect for mammals (including humans) immunized with Plasmodium falciparum or Plasmodium vivax recombinant ROPE against infection with these parasites. As shown immunization was highly efficient. Ideally a single immunization step using a strong immunization protocol would be sufficient to confer protection.
  • Example 2 Further Development of a Pf ROPE or Pv ROPE Malaria Vaccine
  • Steps towards the development of a Pf ROPE or Pv ROPE malaria vaccine include generation and testing of antibodies against ROPE fragments, testing of antibodies against ROPE peptide-microarrays covering the entire ROPE sequence, and preclinical and clinical trials.
  • a.) Generation and Testing of Antibodies Against ROPE Fragments
  • Synthesizing parts of Plasmodium falciparum (Pf) ROPE as peptides, raising soluble scFvs (single chain fragment variable) antibodies against these peptides and testing the capacity to block invasion of human red blood cells by Plasmodium falciparum in vitro in culture. Antibodies showing the strongest inhibition will be used to produce antibodies that can be used for passive immunization in humans, similar to an anti-malaria drug.
  • Several peptides are produced from the Pf ROPE amino acid sequence. To analyze the Pf B cell epitopes, the Pf ROPE sequence was analyzed using the PROTEAN subroutine in the DNASTAR package. This subroutine uses (Wang et al., 2016):
      • Predicted alpha-regions (Gamier and Robson, 1989; Chou and Fasman, 1978)
      • Hydrophilicity (Kyte and Doolittle, 1982)
      • Flexibility (Karpus and Schulz, 1985)
      • Surface probability (Emini et al., 1985)
      • Antigenicity (Jameson and Wolf, 1988).
  • Based on this analysis the following peptides with good hydrophilicity, high accessibility, high flexibility, and strong antigenicity were selected as the antigen epitopes as shown in Table 1.
  • TABLE 1
    Epitopes derived from ROPE
    Amino
    acid
    position Length
    in (amino Sequence
    ROPE Sequence acids) No.
    331-356 KQEKEKEKEKEREKEKEREKEKEKEY 26 SEQ ID NO: 7
    420-447 KNLKTELEKKEKELKDIENVSKEEINKL 28 SEQ ID NO: 8
    520-560 SKKEKEYNQYKNTYIEEINNLNEKLEE 41 SEQ ID NO: 9
    TNKEYTNLQNNYTN
    812-840 KEEYEDKMNTLNEQNEDKMNSLKEEY 29 SEQ ID NO: 10
    ENK
    953-996 KGLKKEVEEKEHKRHSSFNILKSKEKFF 44 SEQ ID NO: 11
    KNSIEDKSHELKKKHE
    1050-1079 KDKSKEKIKDKENQINVEKNEEKDLKKK 30 SEQ ID NO: 12
    DD
    1277-1305 EDEKKRNLNEINNLKKKNEDMCIKYNEMN 29 SEQ ID NO: 13
    1452-1479 KTNKENEEKIINLTSQYSDAYKKKSDES 28 SEQ ID NO: 14
    1497-1521 SNNNIRTNEYKYEEMFDTNIEEKNG 25 SEQ ID NO: 15
    1581-1605 GNISNKNENNNKKNNTCDGYDEKVT 25 SEQ ID NO: 16
  • It is reasonable that these peptides or fragments thereof are particularly suitable as a polypeptide strand P usable in a vaccine V.
  • Human antibodies are generated against above Pf ROPE peptides by phage display using human antibody gene libraries (Kugler et al. 2015).
  • In brief, biotinylated Pf ROPE peptides are immobilized on streptavidin coated microtiter plates. The libraries are incubated with the peptides, non-binding antibody phage particles removed by rigid washing steps. The bound antibody phages are eluted by trypsin and re-amplified using E. coli XL1-Blue and the M13-K07 helper phage. Subsequently, two further panning rounds are performed. Monoclonal antibodies are produced as soluble scFvs (single chain fragment variable) antibodies, using the phage display vector pHAL30 and identified by screening ELISA on the immobilized Pf ROPE peptides. This step is needed to discard non-specific binders of the corresponding antibody phage particles.
  • For further tests the monoclonal scFvs—single chain fragment variable-antibodies are re-cloned into the bivalent scFv-Fc format and produced in mammalian cells. The mammalian vector pCSE2.6-hIgG-Fc-XP and HEK293 6E cells are used. This is an IgG like bivalent molecule and also effector functions are established. (Jager et al., 2013). These IgG-like antibodies are used to test for their capacity to block invasion of human red blood cells by Plasmodium falciparum in vitro in culture. An in vitro growth inhibition activity assay (GIA) is used to measure the efficacy of the soluble scFvs antibodies directed against our peptides in blocking merozoite invasion (Kennedy et al., 2002).
  • Peptides identified as protective in the context of this in vitro inhibition can be used for the production of chemically synthesized vaccines, either as single peptides or as a fusion of several peptides or can be used as diagnostics.
  • b.) Testing of Antibodies Against ROPE Fragments in Microarrays
  • Peptide microarrays covering the entire Pf ROPE sequence are prepared. Sera from malaria patients are used to identify the immunodominant parts of the Pf ROPE protein during the course of an infection with Plasmodium falciparum.
  • The whole amino acid sequence of the target protein is retrieved from a public database and translated into 15-mer peptides with a peptide-peptide overlap of e.g. 12 amino acids. The peptide arrays with the corresponding peptides are produced by the company PEPperPRINT GmbH (Heidelberg, Germany) in a laser printing process on glass slides, coated with a PEGMA/PMMA graft copolymer, which are functionalized with a ßAla-ßAla-linker.
  • A layer of amino acid particles, containing Fmoc-amino acid pentafluorophenyl esters, is printed layer after layer onto the functionalized glass slides, with intermittent melting (i.e. coupling) steps at 90° C. and chemical washing and capping steps (Stadler et al., 2008), based on the same principle as Merrifield's solid-phase peptide synthesis. Peptides are generated in duplicates on the arrays, which are screened for IgG and IgM responses in human sera.
  • Therefore, peptide microarrays are placed in incubation trays (PEPperPRINT GmbH, Heidelberg, Germany) and blocked for 30 min at room temperature with western blot blocking buffer MB-070 (Rockland, USA). Then, sera are diluted 1:1000 in PBS buffer with 0.05% Tween 20 pH 7.4 (PBS-T) and 10% blocking buffer, incubating the sera for 16 h at 4° C. and 50 RPM orbital shaking. Peptide microarrays are washed three times shortly with PBS-T, followed by an incubation with a 1:2500 dilution of the secondary fluorescently labeled antibody, together with a control antibody for 30 min at room temperature. The peptide microarrays are washed with PBST and rinsed with deionized water. After drying in a stream of air, fluorescent images are acquired using an Odyssey Imaging System (LI-COR, USA) at 700 nm. Image analysis and quantification is performed with the PepSlide Analyzer software (Sicasys Software GmbH, Heidelberg, Germany).
  • c.) Further Preclinical and Clinical Trials with Recombinant Pf ROPE Protein
  • The result of this mapping will determine which part of the long Pf ROPE protein (1979 amino acids) will be expressed as a recombinant protein in E. coli and be used as an anti-malaria vaccine in Aotus monkeys and once proven to be efficient in human trials.
  • The DNA sequence coding for the entire Pf ROPE protein or parts of it is amplified by PCR of genomic Pf DNA and cloned into the pET-21a (+)-plasmid at the multiple cloning site (MCS). The MCS is under the control of a T7 promoter and flanked by a T7- and a HIS-tag. The recombinant pET-21a (+)-plasmid containing a sequence coding for Pf ROPE is transformed into E. coli BL21(DE3), a strain that allows high-efficiency protein expression of any gene that is under the control of a T7 promoter. The expressed recombinant Pf ROPE protein carries a histidine-tag at its C-terminus and is purified on a Nickel-column.
  • The recombinant Pf ROPE protein is mixed with a pharmaceutically acceptable carrier or excipient. A vaccine is obtained. This is applied to Aotus monkeys and once proven to be efficient in human trials.
  • REFERENCES
  • U.S. Pat. No. 8,716,443
  • Chou P Y, Fasman G D (1978) Prediction of the secondary structure of proteins from their amino acid sequence. Adv Enzymol 47:45-148.
  • Emini E A, Hughes J V, Perlow D S, Boger J (1985) Induction of hepatitis A virus-neutralizing antibody by virus specific synthetic peptide. J Virol 55:836-839
  • Gamier J, Robson B (1989) The GOR method for predicting secondary structures in proteins. In Fasman G D ed., Prediction of protein structure and the principles of protein conformation. New York, USA. Plenum Press. pp 417-465.
  • Jager V, Büssow K, Wagner A, Weber S, Hust M, Frenzel A and Schirrmann (2013) High level transient production of recombinant antibodies and antibody fusion proteins in HEK293 cells. BMC Biotechnology 13:52, 1472-6750/13/52
  • Jameson B A, Wolf (1988) The antigenic index: a novel algorithm for predicting antigenic determinants. Comput Appl Biosci 4(1):181-186
  • Kulangara, C., A. V. Kajava, G. Corradin, and I. Felger (2009) Sequence conservation in Plasmodium falciparum alpha-helical coiled coil domains proposed for vaccine development. PLoS One 4:e5419.
  • Karplus P A, Schulz G E. (1985) Prediction of chain flexibility in proteins: tool for the selection of peptide antigens. Naturwissenschaften 72:212-213.
  • Kennedy M C, Jin Wang, Yanling Zhang, Miles A P, Chitsaz F, Saul A, Long C A, Miller L H, Stowers A W (2002) In vitro studies with recombinant Plasmodium falciparum apical membrane antigen 1 (AMA1): Production and activity of an AMA1 vaccine and generation of a multiallelic response. Infect Immun 70(12):6948-6960.
  • Kugler J, Wilke S, Meier D, Tomszak F, Frenzel A, Schirrmann T, Dübel S, Garritsen H, Hock B, Toleikis L, Schutte M, Hust M (2015) Generation and analysis of the improved human HAL9/10 antibody phage display libraries Biotechnology 15:10; DOI 10.1186/s12896-015-0125-0
  • Kyte J, Doolittle R F. (1982) A simple method for displaying the hydropathic character of a protein. J Mol Biol 157:105-132.
  • Leslie, M. (2013) Solution to Vaccine Mystery Starts to Crystallize. Science 341:26-27
  • Stadler V et al. (2008) Combinatorial synthesis of peptide arrays with a laser printer. Angew. Chem. Int. Ed. Engl. 47(37):7132-7135
  • Topolska A E, Richie T L, Nhan D H, Coppel, R L. (2004), Associations between responses to the rhoptry-associated membrane antigen of Plasmodium falciparum and immunity to malaria infection. Infect Immun 72(6):3325-3330
  • Villard V, Agak G W, Frank G, Jafarshad A, Servis C, Servis C, Nébié I, Sirima S B, Felger I, Arevalo-Herrera M, Herrera S, Heitz F, Backer V, Druilhe P, Kajava A V, Corradin G (2007) Rapid identification of malaria vaccine candidates based on alpha-helical coiled coil protein motif. PLoS ONE, Issue 7, e645.
  • Wang Y, Wang G, Cai J P (2016) Identifying Novel B Cell Epitopes within Toxoplasma gondii GRA6. Korean J Parasitol. 54(4):431-437 (English. https://doi.org/10.3347/kjp.2016.54.4.431)
  • Watier H, Verwaerde C, Landau I, Werner E, Fontaine J, Capron A, Auriault C (1992). T-cell dependent immunity and thrombocytopenia in rats infected with Plasmodium chabaudi. Infect Immune, 60:136-142
  • Werner E B E, William W R Taylor, Holder A A. (1998) A Plasmodium chabaudi protein contains a repetitive region with a predicted spectrin-like structure. Mol Biochem Parasitol. 94:185-196.

Claims (24)

1-23. (canceled)
24. A vaccine V comprising:
(A) at least one isolated polypeptide strand P comprising or consisting of at least nine consecutive amino acid moieties of SEQ ID NO: 2, of SEQ ID NO: 3 or of SEQ ID NOs: 2 and 3, or a polynucleotide strand encoding for said polypeptide strand P; and
(B) at least one pharmaceutically acceptable carrier or excipient.
25. The vaccine V of claim 24, wherein the isolated polypeptide strand P is obtained from heterologous expression.
26. The vaccine V of claim 24, wherein the isolated polypeptide strand P is obtained from heterologous expression in bacterial or eukaryotic cells.
27. The vaccine V of claim 24, wherein the at least one isolated polypeptide strand P comprises or consists of at least nine consecutive amino acid moieties of a sequence having at least 80% sequence homology to a peptide sequence selected from the group consisting of SEQ ID NO: 2, SEQ ID NO: 3, SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
28. The vaccine V of claim 24, wherein said vaccine V comprises an adjuvant supporting immunologic stimulation.
29. The vaccine V of claim 24, wherein said vaccine V comprises at least one adjuvant supporting immunologic stimulation selected from the group consisting of alum and an immunostimulatory peptide.
30. The vaccine V of claim 24, wherein the at least one isolated polypeptide strand P consists of or comprises a peptide strand having at least 80% sequence homology to sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression.
31. The vaccine V of claim 24, wherein the at least one isolated polypeptide strand P consists of or comprises a peptide strand having a sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression.
32. The vaccine V of claim 24, wherein the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising or consisting of a fraction of SEQ ID NO: 2 truncated by at least 100 amino acid moieties in length and comprising at least two peptide sequences selected from the group consisting of SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
33. The vaccine V of claim 24 wherein the at least one isolated polypeptide strand P is a truncated version of SEQ ID NO: 2 comprising or consisting of a fraction of SEQ ID NO: 2 truncated by at least 100 amino acid moieties in length and comprising all of the peptide sequences SEQ ID NO: 7, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 14, SEQ ID NO: 15, and SEQ ID NO: 16.
34. The vaccine V of claim 24, wherein said vaccine V comprises or consists of:
(A) at least one isolated polypeptide strand P comprising or consisting of a peptide strand having at least 80% sequence homology to a sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression; and
(B) at least one adjuvant supporting immunologic stimulation and optionally one or more further pharmaceutically acceptable carriers.
35. The vaccine V of claim 24, wherein said vaccine V comprises or consists of:
(A) at least one isolated polypeptide strand P comprising or consisting of a peptide strand of sequence SEQ ID NO: 2 or SEQ ID NO: 3 obtained from heterologous expression; and
(B) at least one adjuvant supporting immunologic stimulation selected from the group consisting of alum and an immunostimulatory peptide, and optionally one or more further pharmaceutically acceptable carriers.
36. The vaccine V of claim 24, wherein the polynucleotide strand encoding for said polypeptide strand P is double or single stranded DNA or double or single stranded RNA, or an analogue of double or single stranded DNA or double or single stranded RNA.
37. The vaccine V of claim 24, wherein the polynucleotide strand encoding for said polypeptide strand P is a plasmid.
38. A method for preventing malaria in a patient, wherein the patient is administered with a sufficient amount of a vaccine V of claim 24.
39. A method for preparing an antibody AB binding to Plasmodium falciparum or Plasmodium vivax comprising the following steps:
(i) providing:
(a) a vaccine V according to claim 24, and
(b) an organism O suitible for generating antibodies;
(ii) administering the organism O with the vaccine V;
(iii) waiting until the subjected organism of step (ii) shows an immune response against the antigens of the vaccine V;
(iv) obtaining antibody-generating cells C of the organism O of step (iii);
(v) optionally hybridizing the antibody-generating cells of step (iv) with myeloma cells obtaining immortalized antibody-generating cells C1;
(vi) optionally isolating the nucleotide encoding for the antibody AB of interest and transfer it to another antibody-generating cell type C2 suitible for expressing the antibody AB;
(vii) cultivating the antibody-generating cells C, C1 or C2 of any of steps (iv) to (vi) under conditions enabling the production of the antibody AB; and
(viii) isolating the antibody AB from step (vii).
40. An antibody or antibody fragment AB binding a polypeptide strand P SEQ ID NO: 2 or SEQ ID NO: 3 or a polynucleotide strand encoding for said polypeptide strand P with a dissociation constant of not more than 1000 nM.
41. An antibody or antibody fragment AB binding a polypeptide strand P SEQ ID NO: 2 or SEQ ID NO: 3 or a polynucleotide strand encoding for said polypeptide strand P with a dissociation constant of not more than 1000 nM, wherein said antibody or antibody fragment AB is obtained from a method of claim 39.
42. The antibody or antibody fragment AB of claim 40, wherein said antibody or antibody fragment AB is a monoclonal humanized antibody.
43. The antibody or antibody fragment AB of claim 40, wherein said antibody or antibody fragment AB bears a binding affinity to a polypeptide strand P of SEQ ID NO: 2 or SEQ ID NO: 3 of a dissociation constant of not more than 100 nM, not more than 50 nM, not more than 20 nM, not more than 10 nM, or not more than 5 nM.
44. A method for treating or preventing malaria in a patient, wherein the patient is administered with a sufficient amount of an antibody or antibody fragment AB of claim 40.
45. A method for diagnosing malaria in a patient, wherein the patient is administered with a sufficient amount of an antibody or antibody fragment AB of claim 40.
46. A method for staining Plasmodium falciparum or Plasmodium vivax in vitro, said method comprising the steps of:
(i) providing
(a) an optionally fixed sample S containing Plasmodium falciparum or Plasmodium vivax, and
(b) an optionally stained antibody or antibody fragment AB according to claim 40;
(ii) contacting the sample S with the optionally stained antibody or antibody fragment AB;
(iii) optionally contacting the treated sample of step (ii) with a second antibody or antibody fragment AB2 which is stained and selectively binds to the antibody or antibody fragment AB.
US16/615,490 2017-05-24 2018-05-24 Plasmodium falciparum and plasmodium vivax vaccine Pending US20200261561A9 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
DE102017004987.4 2017-05-24
DE102017004987 2017-05-24
PCT/EP2018/063703 WO2018215612A1 (en) 2017-05-24 2018-05-24 Plasmodium falciparum and plasmodium vivax vaccine

Publications (2)

Publication Number Publication Date
US20200113988A1 US20200113988A1 (en) 2020-04-16
US20200261561A9 true US20200261561A9 (en) 2020-08-20

Family

ID=62530195

Family Applications (1)

Application Number Title Priority Date Filing Date
US16/615,490 Pending US20200261561A9 (en) 2017-05-24 2018-05-24 Plasmodium falciparum and plasmodium vivax vaccine

Country Status (4)

Country Link
US (1) US20200261561A9 (en)
EP (1) EP3630170A1 (en)
CN (1) CN110958886A (en)
WO (1) WO2018215612A1 (en)

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU1818200A (en) * 1998-11-05 2000-05-22 Daniel Carucci Chromosome 2 sequence of the human malaria parasite (plasmodium falciparum) and proteins of said chromosome useful in anti-malarial vaccines and diagnostic reagents
EP2116261A1 (en) 2008-05-07 2009-11-11 Institut Pasteur Sub-region of a plasmodium protein with improved vaccine potential and medical uses thereof

Also Published As

Publication number Publication date
WO2018215612A1 (en) 2018-11-29
EP3630170A1 (en) 2020-04-08
CN110958886A (en) 2020-04-03
US20200113988A1 (en) 2020-04-16

Similar Documents

Publication Publication Date Title
ES2699753T3 (en) Peptide mimics of claudine 18.2 and its uses
US11046745B2 (en) Peptide mimotopes of the CD3 T-cell co-receptor epsilon chain and uses thereof
RU2558301C2 (en) Polypeptides for bonding with "receptor of advanced glycation endproducts", their compositions and methods, which they take part in
US20070003562A1 (en) GLURP-MSP3 fusion protein, immunogenic compositions and malarial vaccines containing it
JP2022028646A (en) Immune-modulating compounds
JP2004532005A (en) Chimera molecule containing a module capable of inducing specific cells and a module controlling the apoptosis-inducing function of the permeable transition pore complex (PTPC)
Wickramarachchi et al. Identification and characterization of a novel Plasmodium falciparum merozoite apical protein involved in erythrocyte binding and invasion
EP3612562A2 (en) Plasmodium sporozoite npdp peptides as vaccine and target novel malaria vaccines and antibodies binding to
US20220227853A1 (en) Neutralizing antibodies to plasmodium falciparum circumsporozoite protein and their use
Dutta et al. Strain-transcending neutralization of malaria parasite by antibodies against Plasmodium falciparum enolase
JP7186940B2 (en) Antibody variants that cross the blood-brain barrier and uses thereof
JP2014527084A (en) Hyr1-derived composition and method of treatment using the same
JP2018527936A (en) Nepovirus coat protein fusion polypeptide and use thereof
US9120869B1 (en) Synthetic antigen based on the ligand domain of the plasmodium vivax duffy binding protein
US20200261561A9 (en) Plasmodium falciparum and plasmodium vivax vaccine
AU2005207998A1 (en) Biotherapeutics, diagnostics and research reagents
US20230365633A1 (en) Griffithsin for rhabdoviridae infections
EP4240407A1 (en) Antigen binding domain with reduced clipping rate
Alba et al. 3D structure and immunogenicity of Plasmodium falciparum sporozoite induced associated protein peptides as components of fully-protective anti-malarial vaccine
KR20230027701A (en) Antibody or antigen binding fragment thereof specifically binding to Koi herpesvirus and Uses thereof
KR20230140889A (en) p62 protein fragment derived from African swine fever virus as recombinant antigen, and uses thereof
US20180221468A1 (en) Haemophilus influenzae immunogen
KR20230137070A (en) p30 protein fragment derived from African swine fever virus as recombinant antigen, and uses thereof
KR20130104816A (en) The polyclonal antibody raised against the heterocapsa triquetra alpha-tubulin for detecting dinoflagellate in ballast water
Siddiqui et al. RETRACTED

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCV Information on status: appeal procedure

Free format text: NOTICE OF APPEAL FILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION