WO2014087007A1 - Anticorps de neutralisation de plasmodium falciparum - Google Patents

Anticorps de neutralisation de plasmodium falciparum Download PDF

Info

Publication number
WO2014087007A1
WO2014087007A1 PCT/EP2013/075867 EP2013075867W WO2014087007A1 WO 2014087007 A1 WO2014087007 A1 WO 2014087007A1 EP 2013075867 W EP2013075867 W EP 2013075867W WO 2014087007 A1 WO2014087007 A1 WO 2014087007A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
antibody
antibodies
amino acid
binding molecule
Prior art date
Application number
PCT/EP2013/075867
Other languages
English (en)
Inventor
Hedda WARDEMANN
Original Assignee
MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V.
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 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. filed Critical MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V.
Publication of WO2014087007A1 publication Critical patent/WO2014087007A1/fr

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • 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 invention relates to binding molecules, in particular antibodies or antigen- binding fragments thereof, capable of binding to the merozoite surface protein 3 (MSP 3) of Plasmodium spec. Binding molecules, antibodies or antigen-binding fragments of these antibodies of the present invention are capable of inhibiting Plasmodium growth or infection without activating cells of the host's immune system.
  • the invention also provides for polypeptides comprising the heavy and/or light chains, isolated CDRs or the variable regions of the binding molecules, antibodies or antigen-binding fragments of these antibodies. Also antigen-binding fragments that bind to MSP 3 or fragments thereof are disclosed.
  • compositions like pharmaceutical or diagnostic compositions, comprising the binding molecules, antibodies or antigen-binding fragments of these antibodies disclosed herein or nucleic acid molecules encoding the same are also provided herein.
  • the invention relates to binding molecules, in particular antibodies or antigen- binding fragments thereof, that specifically bind to the merozoite surface protein 3 (MSP3) of Plasmodium spec, in particular Plasmodium falciparum.
  • MSP3 merozoite surface protein 3
  • the inventive binding molecules in particular antibodies or antigen-binding fragments thereof as provided herein, are capable of inhibiting Plasmodium spec, in particular Plasmodium falciparum, growth or infection in the absence of cells of a host's immune system in vitro.
  • Plasmodium falciparum Five species of Plasmodium can infect and be transmitted by humans. The vast majority of deaths are caused by P. falciparum while P. vivax, P. ovale, and P. malariae cause a generally milder form of malaria. Malaria after Plasmodium falciparum (Pf) infection is a leading course of morbidity and mortality, primarily in children in sub-Saharan Africa. The World Health Organization has estimated that in 2010, there were 216 million documented cases of malaria. That year, between 655,000 and 1 .2 million people died from the disease (roughly 2000-3000 per day) (Nayyar et al., (2012) The Lancet Infectious Diseases, 12(6), 488-96).
  • Plasmodium spec infection is established after injection of small numbers of sporozoites into the skin of vertebrates, such as for example humans in the case of Plasmodium falciparum, by female Anopheles mosquitoes. From the skin, sporozoites migrate quickly via the blood stream to the liver to infect hepatocytes for a single round of massive replication and differentiation into red blood cell invasive merozoites.
  • the pre-erythrocytic phase of the infection is asymptomatic and clinical malaria symptoms develop only during the following erythrocytic phase, which is characterized by repeated cycles of intra-erythrocytic replication, release from bursting red blood cells, and reinvasion of erythrocytes.
  • An antigen based vaccine against malaria comprising fusion proteins derived from Plasmodium falciparum glutamate-rich protein (GLURP) genetically coupled to other Plasmodium falciparum derived protein or homologues thereof has also been described in US201 1/0020387.
  • Said vaccine is proposed to also comprise the Merozoite surface protein 3 (MSP 3) from Plasmodium falciparum.
  • MSP 3 Merozoite surface protein 3
  • MSP 3 The merozoite surface protein 3 (MSP 3) is a non-integral surface-associated protein found in Plasmodium spec. MSP 3 was identified using the serum of semi- immune humans by reactivity of crude parasite lysate in immunoblots and through inhibition of Plasmodium falciparum growth in vitro in presence of blood monocytes in an antibody-dependent cellular inhibition assay (ADCI); see e.g. Oeuvray (1994), Mem Inst Oswaldo Cruz 89, 77-80. MSP 3 is a 48 kDa protein with an N-terminal polymorphic region containing three blocks of alanine-rich hepta repeats and a C- terminal region containing a conserved leucine zipper sequence. MSP 3 is associated with the surface of merozoite through non-covalent linkages with other surface proteins. The function of MSP-3 is unknown.
  • ADCI Antibody-Dependent Cellular Inhibition
  • the ADCI mechanism is known as a mechanism of defense where antibodies need to cooperate with monocytes in order to be effective against parasites, in particular Plasmodium.
  • these anti-MSP3 antibodies compulsorily need cells of the host's immune system, in particular monocytes, to induce Plasmodium growth inhibition.
  • Antibodies disclosed in WO03/016354 are thus dependent on the ADCI mechanism.
  • the technical problem underlying the present invention is the provision of improved medical and diagnostic means and methods for Plasmodium associated diseases, in particular malaria.
  • binding molecules in particular antibodies or antigen-binding fragments thereof, binding to MSP 3 of Plasmodium spec, in particular Plasmodium falciparum, are found that inhibit Plasmodium spec. infection/invasion independent from the Antibody-Dependent Cellular Inhibition (ADCI) mechanism and without activating cells of the host's immune system.
  • ADCI Antibody-Dependent Cellular Inhibition
  • binding molecules, antibodies or antigen-binding fragments of these antibodies provided herein are highly specific in their binding to MSP 3.
  • binding molecules, antibodies or antigen-binding fragments of these antibodies do not cross-react with other antigens, like GLURP, other MSP molecules or corresponding family members, molecules like human insulin, lipopolysaccharide (E. coli) or dsDNA (salmon sperm).
  • antigens like GLURP, other MSP molecules or corresponding family members, molecules like human insulin, lipopolysaccharide (E. coli) or dsDNA (salmon sperm).
  • the invention relates to binding molecules, in particular antibodies or antigen-binding fragments thereof, that specifically bind to the merozoite surface protein 3 (MSP3) of Plasmodium spec, in particular Plasmodium falciparum.
  • MSP3 merozoite surface protein 3
  • the inventive binding molecules, in particular antibodies or antigen-binding fragments thereof, as provided herein are capable of inhibiting Plasmodium spec, in particular Plasmodium falciparum, growth and/or infection and/or invasion in the absence of cells of a host's immune system in vitro.
  • the binding molecules, antibodies and/or antigen-binding fragments of these antibodies of the present invention show surprising advantageous effects through their binding to MSP 3.
  • Advantageous effects of the inventive binding molecules, antibodies and/or antigen-binding fragments of these antibodies comprise a surprisingly low IC 5 0 value for Plasmodium spec, growth, in particular for Plasmodium falciparum, more particularly for 3D7 cells, most particularly for 3D7 LUC cells, high percentage of growth inhibition in a growth inhibition assay of Plasmodium spec, in particular Plasmodium falciparum, more particularly 3D7 LUC , HB3, DD2, IT2 and 3D7 WT cells, high inhibition in an invasion inhibition assay of Plasmodium spec, in particular Plasmodium falciparum, and a mechanism of inhibition independent from the antibody-dependent cellular inhibition (ADCI) mechanism and without activating cells of the host's immune system.
  • ADCI antibody-dependent cellular inhibition
  • Growth inhibition and invasion inhibition assays in order to determine IC 5 0 values or invasion inhibition may be performed with synchronized late stage 3D7 Luc , 3D7 WT , DD2, IT2 or HB3 cultures at a starting parasitemia of 0.2% or 0.5%, respectively.
  • Cells may be cultured in duplicates in the presence of recombinant monoclonal antibodies or protein G purified serum IgG. Uninfected cultures, chemical inhibitors (chloroquine and artemisinin at 50 mM), whole serum (10%) and mGO53 (non-Pf reactive recombinant monoclonal antibody) may be used as internal controls.
  • 3D7 Luc growth may be measured following merozoite release and re-invasion after 24 h using luminescence readers. Growth of 3D7, HB3, IT2 and DD2 cultures may be assessed after 48h using SYBR green (Smilkstein et al. (2004) Antimicrob. Agents Chemotherapy 48, 1803-6).
  • the binding molecules, in particular antibodies or antigen-binding fragments thereof, of the present invention inhibit Plasmodium spec, growth and/or invasion, in particular Plasmodium falciparum, with surprisingly high efficiency in vitro and in in vitro assays without the presence of cells of a host's immune system.
  • the combined presence of (a) binding molecules, antibodies or antigen-binding fragments of these antibodies of the present invention and (b) cells of a host's immune system may lead to additional positive effects in vivo. Said additional effects may be synergistic effects that may lead to improved efficiency, in particular improved growth inhibition and/or invasion inhibition of Plasmodium spec, growth, in particular Plasmodium falciparum in vivo.
  • This has also consequences for the medical use of the binding molecules/antibodies or antigen-binding fragments of these antibodies of the present invention, i.e. their in vivo medical use, e.g. as (a) vaccine(s).
  • the invention relates to binding molecules, in particular antibodies or antigen-binding fragments thereof, that specifically bind to the merozoite surface protein 3 (MSP3) of Plasmodium spec, in particular Plasmodium falciparum.
  • MSP3 merozoite surface protein 3
  • the inventive binding molecules in particular antibodies or antigen-binding fragments thereof as provided herein, are capable of inhibiting Plasmodium spec, in particular Plasmodium falciparum, growth and/or infection and/or invasion in the absence of cells of a host's immune system in vitro.
  • binding molecules, antibodies and/or antigen-binding fragments of these antibodies of the present invention can function based on antigen/epitope binding alone.
  • "Function” in this regard is to be understood as inhibiting Plasmodium spec. growth, in particular Plasmodium falciparum, and preventing from Plasmodium associated diseases, in particular malaria.
  • the present invention provides for binding molecules/antibodies/antigen- binding fragments which are particularly useful in the prevention, treatment and/or diagnosis of Plasmodium falciparum infection, in particular of malaria.
  • the "antigen-binding fragment(s)" may be constituted from different herein disclosed CDRs originating from different clones of the herein defined and provided antibodies.
  • the invention further provides nucleic acids encoding the binding molecules, antibodies and/or antibody fragments of these antibodies according to the invention. Also provided are vectors, such as expression vectors, comprising said nucleic acids, and host cells comprising said nucleic acids or said vectors.
  • compositions comprising polypeptides, nucleic acids, vector or host cells of the present invention. Said compositions may be used for treating or preventing Plasmodium associated diseases, in particular malaria.
  • a kit for detecting a Plasmodium, in particular Plasmodium falciparum, infection comprising polypeptides and/or nucleic acids of the present invention is provided.
  • the kit of the present invention optionally further comprises (a) reaction buffer(s), storage solutions, wash solutions and/or remaining reagents or materials required for the conduction of the assays as described herein.
  • parts of the kit of the invention can be packaged individually in vials or bottles or in combination in containers or multicontainer units. These vials/bottles/containers or multicontainers may, in addition to the polypeptides and/or nucleic acids described and provided herein, comprise preservatives or buffers for storage.
  • the kit may contain instructions for use.
  • kit of the present could be employed in a variety of applications, e.g., as diagnostic kit, as research tool or as therapeutic tool. Additionally, the kit of the invention may contain further means for detection suitable for scientific, medical and/or diagnostic purposes.
  • the manufacture of the kit of the present invention follows preferably standard procedures which are known to the person skilled in the art.
  • the present invention thus relates to binding molecules that specifically bind to the merozoite surface protein 3 (MSP 3) of Plasmodium spec.
  • binding molecules that specifically bind to the merozoite surface protein 3 (MSP 3) of Plasmodium falciparum are provided.
  • Binding molecules of the present invention are in particular antibodies or antigen-binding fragments thereof.
  • binding molecule in accordance with the present invention relates to molecules capable of binding to an antigen, in particular MSP3 of Plasmodium spec.
  • Binding molecules within the meaning of the present invention may also be antibodies and/or antigen- binding fragments thereof that still retain their binding specificity.
  • antibody in accordance with the present invention, comprises polyclonal and monoclonal antibodies as well as derivatives or fragments thereof which still retain the binding specificity. Techniques for the production of antibodies are well known in the art and described, e.g. in Harlow and Lane "Antibodies, A Laboratory
  • antibody in accordance with the invention also includes embodiments such as chimeric, single chain and humanized antibodies, as well as antibody fragments, like, inter alia, Fab fragments, fusion proteins consisting of Eph receptors, ephrin or phosphatase extracellular domains and Fc.
  • Antibody fragments or derivatives further comprise F(ab')2, Fv fragments, scFvs, single domain V H or V-like domains, such as VhH or V-NAR-domains, as well as multimeric formats such as minibodies, diabodies, tribodies, tetrabodies or chemically conjugated Fab'-multimers; see, for example, Harlow and Lane (1988) and (1999), Altshuler (2010) Biochemistry
  • the (antibody) derivatives can be produced by peptidomimetics.
  • techniques described for the production of single chain antibodies see, inter alia, US Patent 4,946,778, can be adapted to produce single chain antibodies specific for polypeptide(s) and fusion proteins of this invention.
  • transgenic animals may be used to express humanized antibodies specific for polypeptides and fusion proteins of this invention.
  • the antibody of this invention is a monoclonal antibody.
  • any technique which provides antibodies produced by continuous cell line cultures, can be used. Examples for such techniques include the original hybridoma technique (Kohler and Milstein (1975) Nature 256, 495) as further developed by the art, the trioma technique, the human B-cell hybridoma technique (Kozbor (1983) Immunology Today 4, 72) and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al. (1985) Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., 77).
  • humanized antibodies also relate to humanized antibodies.
  • "Humanized" forms of non-human (e.g. murine or rabbit) antibodies are chimeric immunoglobulins, immunoglobulin chains or fragments thereof (such as Fv, Fab, Fab', F(ab')2 or other antigen-binding subsequences of antibodies) which contain minimal sequence derived from non-human immunoglobulin.
  • humanized antibodies are human immunoglobulins (recipient antibody) in which residues from a complementary determining region (CDR) of the recipient are replaced by residues from a CDR of a non-human species (donor antibody) such as mouse, rat or rabbit having the desired specificity, affinity and capacity.
  • CDR complementary determining region
  • humanized antibody may comprise residues, which are found neither in the recipient antibody nor in the imported CDR or framework sequences. These modifications are made to further refine and optimize antibody performance.
  • the humanized antibody will comprise substantially all of at least one, and typically two variable domains, in which all or substantially all of the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin consensus sequence.
  • the humanized antibody may also comprise at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin.
  • Fc immunoglobulin constant region
  • a popular method for humanization of antibodies involves CDR grafting, where a functional antigen-binding site from a non-human 'donor' antibody is grafted onto a human 'acceptor' antibody.
  • CDR grafting methods are known in the art and described, for example, in US 5,225,539, US 5,693,761 and US 6,407,213.
  • Another related method is the production of humanized antibodies from transgenic animals that are genetically engineered to contain one or more humanized immunoglobulin loci which are capable of undergoing gene rearrangement and gene conversion (see, for example, US 7,129,084). Further methods for designing and producing humanized antibodies are described in US 07/290,975, US 07/310,252 and US 2003/0229208 or by Queen PNAS (1989), 10029-10033.
  • antibody comprises antibody constructs, which may be expressed in cells, e.g. antibody constructs which may be transfected and/or transduced via, amongst others, viruses or plasmid vectors.
  • the antibody described in the context of the invention is capable to specifically bind/interact with an epitope of the mentioned polypeptide, preferably MSP 3 being as defined further herein.
  • the term "specifically binding/interacting with” as used in accordance with the present invention means that the antibody does not or essentially does not cross-react with an epitope of similar structure.
  • Cross-reactivity of a panel of antibodies under investigation may be tested, for example, by assessing binding of said panel of antibodies under conventional conditions to the epitope of interest as well as to a number of more or less (structurally and/or functionally) closely related epitopes. Only those antibodies that bind to the epitope of interest in its relevant context (e.g.
  • a conformational or discontinuous epitope is characterized for polypeptide antigens by the presence of two or more discrete amino acid residues which are separated in the primary sequence, but come together on the surface of the molecule when the polypeptide folds into the native protein/antigen (Sela (1969) Science 166, 1365; Laver (1990) Cell 61 , 553).
  • the two or more discrete amino acid residues contributing to the epitope are present on separate sections of one or more polypeptide chain(s). These residues come together on the surface of the molecule when the polypeptide chain(s) fold(s) into a three-dimensional structure to constitute the epitope.
  • a continuous or linear epitope consists of two or more discrete amino acid residues, which are present in a single linear segment of a polypeptide chain.
  • Antibody fragments can also be produced directly by recombinant host cells and the antibody phage libraries discussed above.
  • Fab'-SH fragments can be directly recovered from E. coli and chemically coupled to form F(ab')2 fragments (Carter et al (1992) Bio/Technology 10:163-167).
  • F(ab')2 fragments can be isolated directly from recombinant host cell culture.
  • the antibody of choice is a single chain Fv fragment (scFv). See WO 93/16185; U.S. Pat. No. 5,571 ,894; and U.S. Pat. No. 5,587,458.
  • the antibody fragment may also be a "linear antibody", e.g., as described in U.S. Pat. No. 5,641 ,870. Such linear antibody fragments may be monospecific or bispecific.
  • Bispecific antibodies with binding specificities for at least two different epitopes may bind to two different epitopes of the MSP 3 protein.
  • An anti-MSP 3 arm may be combined, for example, with an arm which binds to a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g. CD2 or CD3), or Fc receptors for IgG (FcyR), such as FcyRI (CD64), FcyRII (CD32) and FcyRIII (CD16) so as to focus cellular defense mechanisms to the MSP 3-expressing cell.
  • a triggering molecule on a leukocyte such as a T-cell receptor molecule (e.g. CD2 or CD3), or Fc receptors for IgG (FcyR), such as FcyRI (CD64), FcyRII (CD32) and FcyRIII (CD16)
  • Bispecific antibodies may also be used to localize cytotoxic agents to cells which express MSP 3 (WO96/16673; U.S. Pat. No. 5,837,234; WO98/02463; U.S. Pat. No. 5,821 ,337). Purification methods for bispecific antibodies have been disclosed (WO93/08829; Traunecker et al (1991 ) EMBO J. 10:3655-3659; WO 94/04690; Suresh et al (1986) Methods in Enzymology 121 :210; U.S. Pat. No. 5,731 ,168). Bispecific antibodies can be produced using leucine zippers (Kostelny et al (1992) J. Immunol. 148(5):1547-1553), and single- chain Fv (sFv) dimers (Gruber et al (1994) J. Immunol. 152:5368).
  • bispecific antibodies from antibody fragments
  • Techniques for generating bispecific antibodies from antibody fragments have also been described, such as using chemical linkage wherein intact antibodies are proteolytically cleaved to generate F(ab')2 fragments (Brennan et al (1985) Science 229:81 ).
  • Fab'-SH fragments can be recovered from E. coli and chemically coupled to form bispecific antibodies (Shalaby et al (1992) J. Exp. Med. 175:217-225.
  • the "diabody” technology provides an alternative method for making bispecific antibody fragments (Hollinger et al (1993) Proc. Natl. Acad. Sci. USA 90:6444-6448).
  • a "Fab fragment” generally is comprised of one light chain and the CH1 and variable regions of one heavy chain.
  • the heavy chain of a Fab molecule cannot form a disulfide bond with another heavy chain molecule.
  • An "Fc" region generally contains two heavy chain fragments comprising the CH2 and CH3 domains of an antibody.
  • the two heavy chain fragments are held together by two or more disulfide bonds and by hydrophobic interactions of the CH3 domains.
  • a "Fab' fragment” generally contains one light chain and a portion of one heavy chain that contains the V H domain and the C H domain and also the region between the CH and C H2 domains, such that an interchain disulfide bond can be formed between the two heavy chains of two Fab' fragments to form a F(ab') 2 molecule.
  • a “F(ab')2 fragment” generally contains two light chains and two heavy chains containing a portion of the constant region between the CH1 and CH2 domains, such that an interchain disulfide bond is formed between the two heavy chains.
  • a F(ab')2 fragment thus is composed of two Fab' fragments that are held together by a disulfide bond between the two heavy chains.
  • the "Fv region” generally comprises the variable regions from both the heavy and light chains, but lacks the constant regions.
  • Antibodies with more than two valencies are contemplated.
  • Multivalent, Octopus" antibodies with three or more antigen binding sites and two or more variable domains can be readily produced by recombinant expression of nucleic acid encoding the polypeptide chains of the antibody (US 2002/0004586; WO01/77342).
  • trispecific antibodies can be prepared (Tutt et al (1991 ) J. Immunol. 147:60.)
  • Binding molecules/antibodies/antigen-binding fragments provided herein are preferably in the lgG1 or lgG3 framework, more preferred human lgG1 or lgG3 framework. Binding molecules, antibodies or antigen-binding fragments of these antibodies in the lgG1 or lgG3, preferably human lgG1 or lgG3, framework, are particularly preferred for use in vaccine therapy.
  • the binding molecule, antibody or antigen-binding fragment of this antibody is conjugated to one or more therapeutic agents.
  • conjugated means that the antibody is bound to the therapeutic agent(s) via any type of bonding, and thus includes bonding via fusion proteins (in case the therapeutic agent is of peptidic nature) or any other type of coupling or linkage between the therapeutic agent and the binding molecule, antibody or antigen-binding fragment of this antibody.
  • Conjugated to a therapeutic agent is thus to be construed as including fused to, linked to or coupled to a therapeutic agent.
  • Therapeutic agent refers to any molecule (including small molecules, macromolecules, peptides, polypeptides, proteins, radioactive isotopes, etc) exerting a beneficial effect in the treatment of diseases in humans or other mammals.
  • therapeutic agents are suitable for the therapy of Plasmodium associated diseases, in particular malaria.
  • therapeutic agents also comprises toxins, in particular toxins used in malaria therapy.
  • therapeutic agents that can be conjugated to the binding molecules, antibodies or antigen-binding fragments of these antibodies of the invention targeting MSP 3 include, but are not limited to, antimalaria agents such as quinine, chloroquine, amodiaquine, pyrimethamine, proguanil, sulphonamides, mefloquine, atovaquone, primaquine, artemisinin, halofantrine, doxycycline, clindamycin, artesunat, artemether, artemisinin derivates, quinidine, lumefantrine or ferroquine.
  • antimalaria agents such as quinine, chloroquine, amodiaquine, pyrimethamine, proguanil, sulphonamides, mefloquine, atovaquone, primaquine, artemisinin, halofantrine, doxycycline, clindamycin, artesunat, artemether, artemisinin derivates, quinidine, lumefant
  • binding molecules, antibodies or antigen-binding fragements of the invention may also be conjugated to nanoparticles comprising human serum albumin (HSA) to optimize preparation and uptake of antibodies in cancer cells, as described, for example, by Steinhauser et al., Biomaterials 2006 Oct;27(28):4975-83.
  • HSA human serum albumin
  • the thereapeutic agent(s), such as toxin(s), are preferably suitable for the treatment of Plasmodium associated diseases, in particular malaria.
  • Such conjugates with binding molecules, antibodies or antigen-binding fragments of these antibodies, which binds to MSP 3 can readily be prepared for various types of binding molecules, antibodies or antigen-binding fragments of these antibodies, as disclosed herein, including chimeric antibodies, deimmunized antibodies, humanized antibodies, and fully humanized/fully human antibodies which can be generated recombinantly.
  • Techniques for conjugating agents, such as the therapeutic agents described above, to binding molecules, antibodies or antigen- binding fragments of these antibodies are well known (see, e.g., Arnon et al., "Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy," in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al.
  • Conjugates can be prepared using a variety of cleavable linkers such as for example disulfide-based linkers, hydrazone linkers or peptide linkers (Alley et al. (2010) Curr Opin Chem Biol 14(4):529-37; Webb (201 1 ) Nat.Biotech, 29(4):297-8) or the TAP linker technology from ImmunoGen.
  • cleavable linkers such as for example disulfide-based linkers, hydrazone linkers or peptide linkers (Alley et al. (2010) Curr Opin Chem Biol 14(4):529-37; Webb (201 1 ) Nat.Biotech, 29(4):297-8) or the TAP linker technology from ImmunoGen.
  • binding molecules, antibodies or antigen-binding fragments of these antibodies, of the invention can also be conjugated to or have a detectable label to molecules for diagnostic purposes.
  • a binding molecule, antibody or antigen- binding fragment of this antibody, which binds to MSP 3 can be conjugated to a detectable label ⁇ e.g., for imaging purposes) for diagnosing or detecting endometrial malaria.
  • Suitable detectable markers include, but are not limited to, a radioisotope, a nanoparticle, a fluorescent compound, a bioluminescent compound, chemiluminescent compound, a metal chelator or an enzyme.
  • the MSP 3 epitope to be bound to by the binding molecules, antibodies or antigen-binding fragments of these antibodies of this invention is chosen from one or more of the peptide sequences of MSP 3 as described herein. Said MSP 3 epitope is an extracellular epitope.
  • the binding molecule, antibody or antigen-binding fragment of this antibody may be a monoclonal antibody or a recombinant antibody, or a fragment thereof that binds to MSP 3 or a MSP 3 epitope.
  • the recombinant antibody may be a chimeric antibody, a humanized antibody, a recombinantly produced and engineered fully human/human antibody, or a fragment of such an antibody that binds to MSP 3 or a MSP 3 epitope.
  • the binding molecules, antibodies or antigen-binding fragments of these antibodies of this invention bind to the MSP 3 epitopes as provided herein, see e.g. SEQ ID NOs 244.
  • the binding molecule, antibody or antigen-binding fragment of this antibody which binds to MSP 3 may be conjugated to another agent.
  • the binding molecule, antibody or antigen-binding fragment of this antibody may be conjugated to a toxin, a therapeutic agent, or a detectable label.
  • the binding molecule/antibody/antigen-binding fragment blocks an active site or a binding site on MSP 3 or inhibits the activity of MSP 3 or a MSP 3 fragment or epitope.
  • the binding molecule, antibody or antigen-binding fragment of this antibody may be a therapeutic antibody or a diagnostic antibody.
  • the binding molecule, antibody or antigen-binding fragment of this antibody is a therapeutic antibody.
  • the binding molecule, antibody or antigen-binding fragment of this antibody is a diagnostic antibody.
  • the binding molecules, antibodies or antigen-binding fragments of these antibodies of the invention are expected to bind to various isoforms of MSP 3.
  • Isoforms of MSP 3 include but are not limited to homologues proteins to the amino acid sequence as provided by SEQ ID NO:242 or MSP 3 proteins comprising amino acid sequences which are over their full length at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to SEQ ID NO:242.
  • the binding molecules, antibodies or antigen-binding fragments of these antibodies specifically bind to MSP 3 as shown by SEQ ID NO:242 or fragments thereof or to amino acid sequences that are at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to SEQ ID NO:242 or to amino acid stretches that are at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to SEQ ID NO:244 or fragments thereof, like the fragment defined in SEQ ID NO:246. It is more preferred that the binding molecules/antibodies/antigen-binding fragments provided herein bind to an epitope comprising the amino acid sequence as shown by SEQ ID NO:244. It is even more preferred that the binding molecules, antibodies or antigen-binding fragments of these antibodies bind to an epitope comprising the amino acid sequence as shown by SEQ ID NO:246.
  • Antibody M071 Xi0199 as provided herein is an inventive example of a binding molecule binding to an epitope comprising the amino acid sequence as shown by SEQ ID NO:246.
  • epitope refers to a sequence of at least about 3 to 5, preferably about 5 to 10 or 15, and not more than about 180 amino acids (or any integer there between), which define a sequence that by itself or as part of a larger sequence, binds to an antibody generated in response to such sequence. Said sequence may also be or may be part of a conformational epitope as defined further below. There is no critical upper limit to the length of the fragment, which may comprise nearly the full-length of the protein sequence, or even a fusion protein comprising two or more epitopes from the MSP 3 polyprotein. An epitope for use in the invention is not limited to a polypeptide having the exact sequence of the portion of the parent protein from which it is derived.
  • epitopes encompasses sequences identical to the native sequence, as well as modifications to the native sequence, such as deletions, additions and substitutions (generally conservative in nature).
  • Regions of a given polypeptide that include an epitope can be identified using any number of epitope mapping techniques, well known in the art. See, e.g., Epitope Mapping Protocols in Methods in Molecular Biology, Vol. 66 (Glenn E. Morris, Ed., 1996) Humana Press, Totowa, New Jersey.
  • linear epitopes may be determined by e.g., concurrently synthesizing large numbers of peptides on solid supports, the peptides corresponding to portions of the protein molecule, and reacting the peptides with antibodies while the peptides are still attached to the supports. Such techniques are known in the art and described in, e.g., U.S. Patent No.
  • conformational epitopes are readily identified by determining spatial conformation of amino acids such as by, e.g., x-ray crystallography and 2-dimensional nuclear magnetic resonance. See, e.g., Epitope Mapping Protocols, supra.
  • Antigenic regions of proteins can also be identified using standard antigenicity and hydropathy plots, such as those calculated using, e.g., the Omiga version 1 .0 software program available from the Oxford Molecular Group. This computer program employs the Hopp/Woods method, Hopp et al., Proc. Natl. Acad Sci USA (1981 ) 78:3824-3828 for determining antigenicity profiles, and the Kyte-Doolittle technique, Kyte et al., J.
  • the term “conformational epitope” refers to a portion of a full-length protein, or an analog or mutein thereof, having structural features native to the amino acid sequence encoding the epitope within the full-length natural protein. Native structural features include, but are not limited to, three and four dimensional structure.
  • the length of the epitope defining sequence can be subject to wide variations as these epitopes are believed to be formed by the three-dimensional shape of the antigen (e.g., folding).
  • amino acids defining the epitope can be relatively few in number, but widely dispersed along the length of the molecule, being brought into correct epitope conformation via folding.
  • the portions of the antigen between the residues defining the epitope may not be critical to the conformational structure of the epitope. For example, deletion or substitution of these intervening sequences may not affect the conformational epitope provided sequences critical to epitope conformation are maintained.
  • Conformational epitopes present on the MSP 3 protein are readily identified using methods discussed above, like e.g. x-ray crystallography and 2-dimensional nuclear magnetic resonance. Moreover, the presence or absence of a conformational epitope in a given polypeptide can be readily determined through screening the antigen of interest with an antibody (polyclonal serum or monoclonal to the conformational epitope) and comparing its reactivity to that of a denatured version of the antigen which retains only linear epitopes (if any). In such screening using polyclonal antibodies, it may be advantageous to absorb the polyclonal serum first with the denatured antigen and see if it retains antibodies to the antigen of interest.
  • a conformational epitope may be produced recombinantly and may be expressed in a cell from which it is extractable under conditions which preserve its desired structural features, e.g. without denaturation of the epitope. Such recombinantly produced epitopes may be useful in the preparation of vaccines against
  • Plasmodium spec Accordingly, the present invention also provides for vaccines comprising a Plasmodium spec, epitope as disclosed herein, in particular SEQ ID NO: 1
  • epitopes of MSP 3 as disclosed herein and homologues epitopes, i.e. epitopes that are at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to SEQ ID NO: 244 or that comprise amino acids/fragments of SEQ ID NO: 244 that are at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to such fragments.
  • epitopes as disclosed herein, in particular SEQ ID NOs: 242 and 244, and homologues thereof which are at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to these epitopes can be inter alia used in active immunization. Accordingly, these epitopes can be used most preferably against Plasmodium infections for the prevention and/or treatment of Plasmodium associated diseases, in particular malaria.
  • passive immunization/vaccination with recombinantly procuded antibodies or the recombinantly produced antibodies provided herein is envisaged in a medical setting, i.e. these antibodies can also be used as pharmaceuticals and vaccines.
  • Cells for recombinantly producing such epitopes include bacteria, yeast, insect, and mammalian cells. Expression and isolation of recombinant conformational epitopes is described in e.g., International Publication Nos. WO96/04301 , WO94/01778, WO95/33053, WO92/08734. Alternatively, it is possible to express the antigens and further renature the protein after recovery. It is also understood that chemical synthesis may also provide conformational antigen mimitopes that cross-react with the "native" antigen's conformational epitope.
  • the invention also provides for binding molecules, wherein the binding molecule comprises at least one CDR3 of a V H -region as encoded by a nucleic acid molecule selected from the group consisting of SEQ ID NOs: 5, 17, 29, 41 , 53, 65, 77, 89, 101 , 1 13, 125, 137, 149, 161 and 173 or at least one CDR3 amino acid sequence of an V H -region selected from the group consisting of SEQ ID NOs: 6, 18, 30, 42, 54, 66, 78, 90, 102, 1 14, 126, 138, 150, 162 and 174.
  • a nucleic acid molecule selected from the group consisting of SEQ ID NOs: 5, 17, 29, 41 , 53, 65, 77, 89, 101 , 1 13, 125, 137, 149, 161 and 173 or at least one CDR3 amino acid sequence of an V H -region selected from the group consisting of SEQ ID NOs: 6, 18, 30, 42, 54, 66, 78,
  • binding molecules wherein the binding molecule comprises
  • V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 14, 26, 38, 50, 62, 74, 86, 98, 1 10, 122, 134, 146, 158 and 170
  • said CDR2 having an amino acid sequence selected from the group consisting of SEQ ID NOs: 4, 16, 28, 40, 52, 64, 76, 88, 100, 1 12, 124, 136, 148, 160 and 172
  • said CDR3 having an amino acid sequence selected from the group consisting of SEQ ID NOs: 6, 18, 30, 42, 54, 66, 78, 90, 102, 1 14, 126, 138, 150, 162 and 174;
  • V L which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having an amino acid sequence selected from the group consisting of SEQ ID NOs: 8, 20, 32, 44, 56, 68, 80, 92, 104, 1 16, 128, 140, 152, 164 and 176, said CDR2 having an amino acid sequence selected from the group consisting of SEQ ID NOs: 10, 22, 34, 46, 58, 70, 82, 94, 106, 1 18, 130, 142, 154, 166 and 178, and said CDR3 having an amino acid sequence selected from the group consisting of SEQ ID NOs: 12, 24, 36, 48, 60, 72, 84, 96, 108, 120, 132, 144, 156, 168 and 180.
  • these binding molecules, antibodies, and/or antigen-binding fragments of these antibodies comprise the variable regions and/or CDRs as provided herein.
  • the invention also provides for binding molecules, antibodies and/or antigen-binding fragments of these antibodies that comprise CDRs and/or variable regions that are at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical in their protein sequence (amino acid identity) to one or more of the CDRs and variable regions as provided herein.
  • the present invention also provides for binding molecules, antibodies and antigen-binding fragments of these antibodies that bind to/recognize the same epitope as any of the antibodies as disclosed herein.
  • binding molecules, antibodies or antigen-binding fragments of these antibodies of the invention as disclosed below exhibit remarkable functional properties which make them particularly suitable for use in the therapy or diagnosis of Plasmodium spec, in particular Plasmodium falciparum, associated disorders, including malaria.
  • said binding molecules, antibodies and/or antigen-binding fragments of these antibodies show very high affinity for MSP 3.
  • an anti-MSP 3-antibody comprising: a V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3; said CDR1 having an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 14, 26, 38, 50, 62, 74, 86, 98, 1 10, 122, 134, 146, 158, and 170, or said CDR1 having an amino acid sequence with at least 80% overall sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 2, 14, 26, 38, 50, 62, 74, 86, 98, 1 10, 122, 134, 146, 158, and 170;
  • said CDR2 having an amino acid sequence selected from the group consisting of SEQ ID NOs: 4, 16, 28, 40, 52, 64, 76, 88, 100, 1 12, 124, 136, 148, 160, and 172, or said CDR2 having an amino acid sequence with at least 80% overall sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 4, 16, 28, 40, 52, 64, 76, 88, 100, 1 12, 124, 136, 148, 160, and 172; and
  • said CDR3 having an amino acid sequence selected from the group consisting of SEQ ID NOs: 6, 18, 30, 42, 54, 66, 78, 90, 102, 1 14, 126, 138, 150, 162, and 174, or said CDR3 having an amino acid sequence with at least 80% overall sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 6, 18, 30, 42, 54, 66, 78, 90, 102, 1 14, 126, 138, 150, 162, and 174; and
  • V L which comprises the hypervariable regions CDR1 , CDR2 and CDR3; said CDR1 having an amino acid sequence selected from the group consisting of SEQ ID NOs: 8, 20, 32, 44, 56, 68, 80, 92, 104, 1 16, 128, 140, 152, 164, and 176, or said CDR1 having an amino acid sequence with at least 80% overall sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 8, 20, 32, 44, 56, 68, 80, 92, 104, 1 16, 128, 140, 152, 164, and 176;
  • said CDR2 having an amino acid sequence selected from the group consisting of SEQ ID NOs: 10, 22, 34, 46, 58, 70, 82, 94, 106, 1 18, 130, 142, 154, 166, and 178, or said CDR2 having an amino acid sequence with at least 80% overall sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 10, 22, 34, 46, 58, 70, 82, 94, 106, 1 18, 130, 142, 154, 166, and 178; and
  • said CDR3 having an amino acid sequence selected from the group consisting of SEQ ID NO: 12, 24, 36, 48, 60, 72, 84, 96, 108, 120, 132, 144, 156, 168, and 180, or said CDR3 having an amino acid sequence with at least 80% overall sequence identity to an amino acid sequence selected from the group consisting of SEQ ID NOs: 12, 24, 36, 48, 60, 72, 84, 96, 108, 120, 132, 144, 156, 168, and 180.
  • the invention also provides for polypeptides comprising the heavy and/or light chain, isolated CDRs, the variable regions thereof, or antigen-binding fragments thereof that bind to MSP 3 or fragments thereof.
  • CDR Complementarity Determining Region
  • V variable domains of an antibody.
  • Each variable domain (the heavy chain V H and light chain V L ) of an antibody comprises three complementarity determining regions sometimes called hypervariable regions, flanked by four relatively conserved framework regions or "FRs".
  • FRs relatively conserved framework regions
  • the present invention provides binding molecules, antibodies and/or antigen-binding fragments of these antibodies comprising a light chain variable domain comprising any one of the amino acid sequences of SEQ ID NOs: 184, 188, 192, 196, 204, 208, 212, 216, 220, 224, 228, 232 or 236 and/or a heavy chain variable domain comprising any one of the amino acid sequences of SEQ ID NOs: 182, 186, 190, 194, 202, 206, 210, 214, 218, 222, 226, 230 or 234.
  • the invention also provides binding molecules, antibodies and/or antigen-binding fragments of these antibodies, that have a light chain variable domain comprising any one of the amino acid sequences of SEQ ID NOs: 184, 188, 192, 196, 204, 208, 212, 216, 220, 224, 228, 232 or 236 and a heavy chain variable domain comprising any one of the amino acid sequences of SEQ ID NOs: 182, 186, 190, 194, 202, 206, 210, 214, 218, 222, 226, 230 or 234.
  • binding molecules, antibodies and/or antigen-binding fragments of these antibodies of the present invention comprise a light chain variable domain comprising the amino acid sequence of SEQ ID NO: 184 and/or a heavy chain variable domain comprising the amino acid sequence of SEQ ID NO: 182.
  • This binding molecule is herein illustrated as M071 Xi0199.
  • the present invention also provides for binding molecules, antibodies and/or antigen-binding fragments of these antibodies and heavy and/or light chain variable domains thereof, comprising the CDRs of the heavy chain variable domain and/or light chain variable domain set forth in any one of SEQ ID NOs: 182, 186, 190, 194, 202, 206, 210, 214, 218, 222, 226, 230 or 234 and SEQ ID NOs: 184, 188, 192, 196, 204, 208, 212, 216, 220, 224, 228, 232 or 236, respectively, and/or comprising CDRs derived from said variable domains. It is preferred that the binding molecules, antibodies and/or antigen-binding fragments of these antibodies of the invention comprise the CDRs of the heavy chain
  • the CDR sequences of the heavy and/or light chains provided by the present invention can be determined using standard methods well known in the art.
  • the CDR sequences in antibody variable domains can be determined using methods as described, e.g., in Kabat, E.A., et al. (1991 ) Sequences of
  • V domain delineation system For example, setting the above parameter "V domain delineation system” to "IMGT" results in CDR sequences of the heavy and light chains that comprise a core region of the CDR sequences provided herein and that may comprise additions/deletions from/to the CDR sequences as provided herein.
  • the heavy chains of antibodies provided herein may comprise one, two, or preferably all three of the CDRs of the heavy chain variable domain of any one of
  • CDRs are determined according to any of the methods known in the art.
  • the heavy chain of antibodies provided herein may alternatively or additionally comprise one, two, or, preferably, three CDRs which comprise the respective CDR(s) of the heavy chain variable domain, wherein said CDR comprises any one of SEQ ID NOs: 2, 4, 6, 14, 16, 18, 26, 28, 30, 38, 40,
  • CDRs derived from the respective CDR(s) of the heavy chain variable domain having any one of SEQ ID NOs: 182, 186, 190, 194, 202, 206, 210, 214, 218, 222,
  • a CDR derived from a respective other CDR of a given sequence e.g., derived from a CDR of any one of SEQ ID NOs: 2, 4, 6, 14, 16, 18, 26, 28, 30,
  • 126, 134, 136, 138, 146, 148, 150, 158, 160 or 162 may comprise one or more, preferably two or more, more preferably three or more, even more preferably four or more and most preferably five or more amino acids of the amino acid sequences as set forth in the SEQ ID NOs provided herein and as summarized in Table 1 .
  • a CDR sequence as provided herein comprises one of the amino acid sequences as set forth in SEQ ID NOs: 2, 4, 6, 14, 16, 18, 26, 28, 30, 38, 40, 42, 62, 64, 66, 74, 76, 78, 86, 88, 90, 98, 100, 102, 1 10, 1 12, 1 14, 122, 124, 126, 134, 136, 138, 146, 148, 150, 158, 160 or 162 and as summarized in Table 1 . It is furthermore preferred that all three CDR sequences of a heavy chain of a binding molecule, antibody or antigen-binding fragment of this antibody of the invention are comprised in the CDR sequences as provided herein.
  • a binding molecule, antibody or antigen-binding fragment of this antibody of the invention comprises a heavy chain variable domain comprising CDRs comprising SEQ ID NOs: 2, 4 and 6 or comprising CDRs having at least 80% overall sequence identity to a sequence as shown in SEQ ID NOs: 2, 4 and 6.
  • the antibody light chains provided herein may comprise one, two, or preferably all three of the CDRs of the light chain variable domain of any one of
  • the light chain of antibodies provided herein may alternatively or additionally comprise one, two, or, preferably, three CDRs which comprise the respective CDR(s) of the light chain variable domain, wherein said CDR comprises any one of SEQ ID NOs: 8, 10, 12, 20, 22, 24, 32, 34, 36, 44, 46, 48, 68, 70, 72, 80,
  • a CDR derived from a respective other CDR of a given sequence e.g., derived from a CDR of any one of SEQ ID NOs: 8, 10, 12, 20, 22, 24, 32, 34, 36, 44, 46, 48, 68, 70, 72,
  • 152, 154, 156, 164, 166 or 168 may comprise one or more, preferably two or more, more preferably three or more, even more preferably four or more and most preferably five or more amino acids of the amino acid sequences as set forth in the
  • a CDR sequence as provided herein comprises one of the amino acid sequences as set forth in SEQ ID NOs: 8, 10, 12, 20, 22, 24, 32, 34, 36, 44, 46, 48,
  • a binding molecule, antibody or antigen-binding fragment of this antibody of the invention comprises a heavy chain variable domain comprising CDRs comprising SEQ ID NOs: 8, 10 and 12 or comprising CDRs having at least 80% overall sequence identity to a sequence as shown in SEQ ID NOs: 8, 10 and 12.
  • binding molecules/antibodies/antigen-binding fragments of the present invention preferably comprise six CDRs. It is more preferred that a binding molecule, antibody and/or antigen-binding fragment of this antibody of the invention, comprises one, two, three, four, five, or, preferably, six CDR sequences of a pair of heavy and light chains as provided herein. Pairs of heavy and light chains are preferably those of the antibodies as provided herein.
  • one, two, three, four, five or, preferably, six of the CDR sequences are comprised in the light and/or heavy chain sequences provided herein and as set forth in SEQ ID NOs: 182, 184, 186, 188, 190, 192, 194, 196, 202, 204, 206, 208, 210, 212, 214, 216, 218, 220, 222, 224, 226, 228, 230, 232, 234 and 236.
  • binding molecules, antibodies and/or antigen-binding fragments of these antibodies of the present invention more preferably comprise CDR sequences as shown in SEQ ID NOs: 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38, 40, 42, 44, 46, 48, 62, 64, 66, 68, 70, 72, 74, 76, 78, 80, 82, 84, 86, 88, 90, 92, 94, 96, 98, 100, 102, 104, 106, 108, 1 10, 1 12, 1 14, 1 16, 1 18, 120, 122, 124, 126, 128, 130, 132, 134, 136, 138, 140, 142, 144, 146, 148, 150, 152, 154, 156, 158, 160, 162, 164, 166 or 168.
  • binding molecules, antibodies and/or antigen-binding fragments of these antibodies of the invention comprise CDR sequences as shown in SEQ ID NOs: 2, 4, 6, 8, 10 and 12 or comprise CDRs having at least 80% overall sequence identity to a sequence as shown in SEQ ID NOs: 2, 4, 6, 8, 10 and 12.
  • the binding molecule, antibody and/or antigen-binding fragment of this antibody comprises a variable V H -region as encoded by a nucleic acid molecule as shown in a SEQ ID NO selected from the group consisting of SEQ ID NOs: 181 , 185, 189, 193, 197, 201 , 205, 209, 213, 217, 221 , 225, 229, 233 and 237 or a variable V H -region as shown in a SEQ ID NO selected from the group consisting of SEQ ID NOs: 182, 186, 190, 194, 198, 202, 206, 210, 214, 218, 222,
  • the binding molecule, antibody and/or antigen-binding fragment of this antibody comprises a variable V H -region as encoded by a nucleic acid molecule as shown in a SEQ ID NO: 181 or a variable V H -region as shown in a SEQ ID NO: 182.
  • the binding molecule, antibody and/or antigen-binding fragment of this antibody comprises a V L -region as encoded by a nucleic acid molecule as shown in a SEQ ID NO selected from the group consisting of SEQ ID NOs: 183, 187, 191 , 195, 199, 203, 207, 21 1 , 215, 219, 223,
  • the binding molecule, antibody and/or antigen-binding fragment of this antibody comprises a variable V L - region as encoded by a nucleic acid molecule as shown in a SEQ ID NO: 183 or a variable V L -region as shown in a SEQ ID NO: 184.
  • the binding molecules, antibodies and/or antigen-binding fragments of these antibodies of the present invention or their corresponding immunoglobulin chain(s) can therefore be further modified by at least one conservative amino acid substitution(s) in any one of the amino acid sequences provided herein.
  • at least one conservative amino acid substitution(s) in any one of the amino acid sequences provided herein it is preferred with increasing preference that less than 10, less than 9, less than 8, less than 7, less than 6, less than 5, less than 4, less than 3, less than 2 amino acids are, or 1 amino acid is conservatively substituted in any one of the amino acid sequences provided herein.
  • conservative substitution is widely used in the state of the art and specifies the replacement of an amino acid in a polypeptide by an amino acid with similar characteristics. Similar characteristics are for example size, hydrophobicity, or charge. As it is well known, amino acids are classified as being positively charged, negatively charged, having an uncharged side chain or a hyrophobic side chain. Examples for a conservative substitution are Leu to lie, Arg for Lys, Phe to Trp, Asp to Glu, Ser to Thr, or vice versa. In general, the overall functioning of a amino acid sequence in particular amino acid sequence of CDR is likely not to be essentially affected by conservative substitution and may even be improved.
  • Monoclonal antibodies can be prepared, for example, by the well-established techniques as originally described in Kohler and Milstein, Nature 256 (1975), 495, and Galfre, Meth. Enzymol. 73 (1981 ), 3, which in this case comprise the fusion of mouse myeloma cells to spleen cells derived from immunized mammals with modifications developed by the art.
  • binding molecules, antibodies or antigen-binding fragments of these antibodies of the invention can be coupled to a labelling group, a toxin, or an anti-malaria drug as defined herein above by methods well known in the art.
  • Such coupling may be conducted chemically after expression of the antibody or antigen to the site of attachment or the coupling product may be engineered into the antibody or antigen of the invention at the DNA level.
  • the DNAs are then expressed in a suitable host system as described herein below, and the expressed proteins are collected and renatured, if necessary. Coupling may be achieved via a linker known in the state of the art.
  • linkers that release the toxin, or an anti-malaria drug under acidic or reducing conditions or upon exposure to specific proteases may be employed with this technology.
  • binding molecules, antibodies or antigen-binding fragments of these antibodies inhibit Plasmodium spec, growth, in particular Plasmodium falciparum, more particularly 3D7 cells, most particularly 3D7 LUC cells, with an IC 50 value of at least 30 nM, 20 nM, 15 nM, 10 nM, 5 nM, 4 nM, 3 nM, 2 nM or lower.
  • IC 5 o values within the scope of the present invention are determined using methods well known in the art, in particular using a growth inhibition assay using 3D7 LUC , HB3, DD2, IT2 or 3D7 WT cells, in particular using cells expressing marker genes, more particularly expressing luciferase.
  • IC 50 values of antibodies of the present invention are provided in Figure 12.
  • Plasmodium growth inhibition or invasion inhibition about three assays are employed routinely in the art. Particularly useful assays have been described in the art, e.g., in Methods in Malaria Research, fifth Edition, MR4/ATCC, 2008.
  • Plasmodium spec in particular Plasmodium falciparum, may be cultured according to methods known in the art and as described in, e.g., Maier et al. (2013) Methods in Molecular Biology 923:3-15.
  • the herein described Plasmodium growth inhibition assay (PGIA) and Plasmodium invasion inhibition assay (PIIA) use standard means and methods that are well known in the art.
  • Hepes-buffered media in particular Hepes- buffered RPMI-1640 medium. Most preferably, 25 mM Hepes-buffered RPMI-1640 is used.
  • the used medium may further comprise additives.
  • the used medium may further comprise sodium bicarbonate and/or gentamicin and/or hypoxanthine at various concentrations. It is preferred to use about 0.25% sodium bicarbonate and/or about 0.01 mg/ml gentamicin and/or about 0.05 g/l hypoxanthine.
  • Media may be kept at various temperatures and under various air conditions.
  • Plasmodium spec, in particular Plasmodium falciparum, cultures are kept at 37°C.
  • an artificial atmosphere is preferred. Particularly preferred are high concentrations of nitrogen, e.g., 80% or more nitrogen, preferably 90% nitrogen. Other gases may also be controlled when culturing Plasmodium. Preferred conditions contain equal amounts of oxygen and carbon dioxide. Particularly preferred conditions contain 5% oxygen and 5% carbon dioxide.
  • the above culturing methods may be employed using various strains of Plasmodium spec, in particular Plasmodium falciparum. It is preferred to use 3D7 Luc , 3D7, DD2 or HB3 cells. Further additives may be added to the culturing medium depending on the used strain. In particular, when using 3D7 cells, pyrimethamine is added to the culture.
  • molecules e.g., binding molecules, antibodies or antigen- binding fragments of these antibodies, to be tested to affect Plasmodium growth
  • molecules e.g., binding molecules, antibodies or antigen-binding fragments of these antibodies
  • molecules may be added to the starting culture and/or during Plasmodium growth. It is preferred to add molecules, e.g., binding molecules, antibodies and/or antigen- binding fragments of these antibodies at a certain starting parasitemia. It is particularly preferred to add molecules, e.g., binding molecules, antibodies and/or antigen-binding fragments of these antibodies at a starting parasitemia of 0.2% or 0.5%.
  • parasitemia in this regard is to be understood to be the quantitative content of parasites.
  • parasitemia is to be understood as the percentage of infected red blood cells in relation to the total number of red blood cells. Parasitemia may be assessed using standard means and methods well known in the art, e.g. by using methods described in Methods in Malaria Research, fifth Edition, MR4/ATCC, 2008.
  • Molecules e.g., binding molecules, antibodies and/or antigen-binding fragments of these antibodies may be added to Plasmodium cells at varying concentrations.
  • binding molecules, antibodies and/or antigen-binding fragments of these antibodies may be added at a concentration of 6,4 g/ml and/or 100 pg/ml, see, e.g., figure 1 .
  • IC 5 0 values may be determined using standard methods known in the art.
  • IC 5 0 values may be determined using a variety of concentrations of binding molecules, antibodies and/or antigen-binding fragments of these antibodies of the invention and assessing the inhibition of each concentration. Inhibition may then be plotted against said concentrations in order to determine IC 5 0 values.
  • Plasmodium growth/inhibition and/or invasion inhibition may be assessed using various means and methods well known in the art. In particular, Plasmodium growth/inhibition and/or invasion inhibition may be assessed using SYBR green as described in, e.g., Methods in Malaria Research, fifth Edition, MR4/ATCC, 2008. In particular, Plasmodium growth/inhibition and/or invasion inhibition may be assessed after 48h. Furthermore, Plasmodium growth/inhibition and/or invasion inhibition may be assessed using luminescence readers, e.g., when using 3D7 Luc cells. When using 3D7 cells, it is particularly preferred to assess growth/inhibition and/or invasion inhibition following merozoite release and re-invasion after 24h.
  • the assay as described by Lucumi et al. (2010) Antimicrobial agents and chemotherapy 54(9):3597-604 may be used.
  • Said assay may also be used for other Plasmodium falciparum, in particular 3D7, DD2 or HB3 cells, when adapting the detection method accordingly.
  • the detection method may be changed from luminescence detection to the above SYBR green detection method.
  • PGIA and PIAA conditions are as follows:
  • PKIA Growth inhibition assays
  • PIAA Plasmodium invasion inhibition
  • IC 5 0 values may be performed with synchronized late stage 3D7 Luc , 3D7 WT , DD2, IT2 or HB3 cultures at a starting parasitemia of 0.2% or 0.5%, respectively.
  • Cells may be cultured in duplicates in the presence of recombinant monoclonal antibodies or protein G purified serum IgG. Uninfected cultures, chemical inhibitors (chloroquine and artemisinin at 50 mM), whole serum (10%) and mGO53 (non-Pf reactive recombinant monoclonal antibody) may be used as internal controls.
  • 3D7 Luc growth may be measured following merozoite release and re-invasion after 24 h using luminescence readers. Growth of 3D7, HB3, IT2 and DD2 cultures may be assessed after 48h using SYBR green (Smilkstein et al. (2004) Antimicrob. Agents Chemotherapy 48, 1803-6).
  • Plasmodium growth inhibition (PGIA) and/or Plasmodium invasion inhibition (PIAA) assays may be performed as follows. Parasites were cultured under standard conditions. In brief, Plasmodium falciparum clone 3D7, DD2, HB3, and IT2 were cultivated at 5% hematocrit in RPMI 1640 supplemented with 0.05 g/l hypoxanthine (Sigma), 25 mM HEPES, 0.5% Albumax II (Life Technologies), 0.25% sodium bicarbonate and 0.01 mg/ml gentamicin (Sigma). All cultures were kept at 37 °C under 90% nitrogen, 5% oxygen, and 5% carbon dioxide.
  • PKIA Plasmodium growth inhibition
  • PIAA Plasmodium invasion inhibition
  • 3D7 Luc cultures were supplemented with 40 ng/ml pyrimethamine to ensure transgene expression.
  • Neutralization assays were performed with synchronized late stage 3D7 Luc or 3D7, DD2, and HB3 cultures at a starting parasitemia of 0.2% or 0.5%, respectively.
  • Parasites were cultured in duplicates in the presence of recombinant monoclonal antibodies or protein G purified serum IgG. Uninfected cultures, chemical inhibitors (chloroquine and artemisinin at 50 mM), whole serum (10%) and mGO53 (non-Pf reactive recombinant monoclonal antibody) were used as internal controls.
  • 3D7 Luc growth was measured following merozoite release and re-invasion after 24 h by quantifying luciferase activity using Bright-Glo Luciferase substrate (Promega). Arbitrary luminescence units were measured on a VICTOR light reader (Perkin Elmer, 1420 Luminescence Counter). Growth of 3D7, HB3, IT4 and DD2 cultures was assessed after 48h using SYBR green (Smilkstein et al., 2004). Fluorescence was measured on a Fluoroskan Ascent reader (Thermo Labsystems).
  • the binding molecules, antibodies and/or antigen-binding fragments of these antibodies of the present invention are capable of inhibiting growth of a broad range of Plasmodium falciparum strains, in particular 3D7 Luc , 3D7 WT , DD2, IT2 and HB3.
  • antibodies M071 Xi0199, M071 Xi2204 and M036Xi0326 inhibit Plasmodium falciparum strains 3D7 Luc , 3D7 WT , DD2, IT2 and HB3 with a high inhibition rate according to values given in Figure 12.
  • These antibodies have particular high inhibition rates. It is preferred that inhibition rate of Plasmodium falciparum strains is above 50%, more preferred above 60% and most preferred above 80%.
  • antibody M071 Xi0199 shows surprisingly good inhibition of Plasmodium falciparum strains, in particular 3D7 Luc , 3D7 WT , DD2, IT2 and HB3, with values above 50%.
  • M071 Xi0199 has particularly high inhibition rates and inhibits growth of Pf-3D7 LUC cells to about 88%, of Pf-HB3 cells to about
  • Inhibition rates may have a variance of 15 to 20% due to
  • M071 Xi0199 shows no cross-reactivity with other antigens, like GLURP, human insulin, lipopolysaccharide (E. coli) or dsDNA (salmon sperm) and an IC 50 value for Pf-
  • binding molecule, antibody and/or antigen- binding fragment of this antibody preferably comprises six CDRs encoded by nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 1 , 3, 5, 7, 9 and 1 1 .
  • Said six CDRs preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 2, 4, 6, 8, 10 and 12, respectively. More preferably, said binding molecule, antibody and/or antigen- binding fragment of this antibody comprises V H and V L nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 181 and 183, respectively.
  • V H and V L chains preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 182 and 184, respectively. It is particularly preferred that said antibody is M071 Xi0199.
  • M071 Xi2204 has particularly high inhibition rates and inhibits growth of Pf-3D7 LUC cells to about 67%, of Pf-HB3 cells to about
  • Inhibition rates may have a variance of 15 to 20% due to
  • M071 Xi2204 shows no cross-reactivity with other antigens, like GLURP, human insulin, lipopolysaccharide (E. coli) or dsDNA (salmon sperm) and an IC 5 o value for Pf-
  • binding molecule, antibody and/or antigen-binding fragment of this antibody preferably comprises six CDRs encoded by nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NO: 1
  • Said six CDRs preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 14, 16,
  • said binding molecule, antibody and/or antigen-binding fragment of this antibody comprises V H and V L nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 185 and 187, respectively.
  • Said V H and V L chains preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 186 and 188, respectively. It is particularly preferred that said antibody is M071 Xi2204.
  • M071 Xi0237 has particularly high inhibition rates and inhibits growth of Pf-3D7 LUC cells to about 75% as compared to untreated Plasmodium falciparum in vitro cultures as well as Plasmodium falciparum in vitro cultures of the respective Plasmodium treated with a non- Plasmodium falciparum reactive control antibody. Inhibition rates may have a variance of 15 to 20% due to Plasmodium biology, in particular due to schizogony. In addition, M071 Xi0237 shows no cross-reactivity with other antigens, like GLURP, human insulin, lipopolysaccharide (E.
  • GLURP human insulin
  • lipopolysaccharide E.
  • binding molecule, antibody and/or antigen-binding fragment of this antibody preferably comprises six CDRs encoded by nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 25, 27, 29, 31 , 33 and 35.
  • Said six CDRs preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 26, 28, 30, 32, 34 and 36, respectively. More preferably, said binding molecule, antibody and/or antigen-binding fragment of this antibody comprises V H and VL nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 189 and 191 , respectively.
  • V H and V L chains preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 190 and 192, respectively. It is particularly preferred that said antibody is M071 Xi0237.
  • M071 Xi2127 has particularly high inhibition rates and inhibits growth of P -3D7 cells to about 79% as compared to untreated Plasmodium falciparum in vitro cultures as well as Plasmodium falciparum in vitro cultures of the respective Plasmodium treated with a non- Plasmodium falciparum reactive control antibody. Inhibition rates may have a variance of 15 to 20% due to Plasmodium biology, in particular due to schizogony. In addition, M071 Xi2127 shows no cross-reactivity with other antigens, like GLURP, human insulin, lipopolysaccharide (E.
  • GLURP human insulin
  • lipopolysaccharide E.
  • binding molecule, antibody and/or antigen-binding fragment of this antibody preferably comprises six CDRs encoded by nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 37, 39, 41 , 43, 45 and 47.
  • Said six CDRs preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 38, 40, 42, 44, 46 and 48, respectively. More preferably, said binding molecule, antibody and/or antigen-binding fragment of this antibody comprises V H and VL nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 193 and 195, respectively.
  • V H and V L chains preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 194 and 196, respectively. It is particularly preferred that said antibody is M071 Xi2127.
  • M071 Xi2057 has particularly high inhibition rates and inhibits growth of Pf-3D7 LUC cells to about 89% as compared to untreated
  • Plasmodium falciparum in vitro cultures as well as Plasmodium falciparum in vitro cultures of the respective Plasmodium treated with a non-
  • Inhibition rates may have a variance of 15 to 20% due to Plasmodium biology, in particular due to schizogony.
  • M071 Xi2057 shows no cross-reactivity with other antigens, like GLURP, human insulin, lipopolysaccharide (E. coli) or dsDNA (salmon sperm) and an IC 5 o value for Pf-3D7 LUC cells of about 20 nM.
  • the herein provided binding molecule, antibody and/or antigen-binding fragment of this antibody preferably comprises six CDRs encoded by nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 61 , 63, 65, 67, 69 and 71 .
  • Said six CDRs preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs 62, 64, 66, 68, 70 and 72, respectively. More preferably, said binding molecule, antibody and/or antigen-binding fragment of this antibody comprises V H and V L nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 201 and 203, respectively.
  • V H and V L chains preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs 202 and 204, respectively. It is particularly preferred that said antibody is M071 Xi2057.
  • M070Xi3010 has particularly high inhibition rates and inhibits growth of Pf-3D7 LUC cells to about 89% as compared to untreated
  • Plasmodium falciparum in vitro cultures as well as Plasmodium falciparum in vitro cultures of the respective Plasmodium treated with a non-
  • Inhibition rates may have a variance of 15 to 20% due to Plasmodium biology, in particular due to schizogony.
  • M070Xi3010 shows no cross-reactivity with other antigens, like GLURP, human insulin, lipopolysaccharide (E. coli) or dsDNA (salmon sperm) and an IC 5 o value for Pf-3D7 LUC cells of about 8.5 nM.
  • the herein provided binding molecule, antibody and/or antigen-binding fragment of this antibody preferably comprises six
  • Said six CDRs preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or
  • said binding molecule, antibody and/or antigen-binding fragment of this antibody comprises V H and V L nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98,
  • V H and V L chains preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 206 and 208, respectively. It is particularly preferred that said antibody is M070Xi3010.
  • M071 Xi0227 has particularly high inhibition rates and inhibits growth of Pf-3D7 LUC cells to about 92% as compared to untreated Plasmodium falciparum in vitro cultures as well as Plasmodium falciparum in vitro cultures of the respective Plasmodium treated with a non- Plasmodium falciparum reactive control antibody. Inhibition rates may have a variance of 15 to 20% due to Plasmodium biology, in particular due to schizogony. In addition, M071 Xi0227 shows no cross-reactivity with other antigens, like GLURP, human insulin, lipopolysaccharide (E.
  • GLURP human insulin
  • lipopolysaccharide E.
  • binding molecule, antibody and/or antigen-binding fragment of this antibody preferably comprises six CDRs encoded by nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 85, 87, 89, 91 , 93 and 95.
  • Said six CDRs preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 86, 88, 90, 92, 94 and 96, respectively. More preferably, said binding molecule, antibody and/or antigen-binding fragment of this antibody comprises V H and VL nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 209 and 21 1 , respectively.
  • V H and V L chains preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 210 and 212, respectively. It is particularly preferred that said antibody is M071 Xi0227.
  • M071 Xi0081 has particularly high inhibition rates and inhibits growth of Pf-3D7 LUC cells to about 92% as compared to untreated
  • Plasmodium falciparum in vitro cultures as well as Plasmodium falciparum in vitro cultures of the respective Plasmodium treated with a non- Plasmodium falciparum reactive control antibody. Inhibition rates may have a variance of 15 to 20% due to Plasmodium biology, in particular due to schizogony.
  • M071 Xi0081 shows no cross-reactivity with other antigens, like GLURP, human insulin, lipopolysaccharide (E. coli) or dsDNA (salmon sperm) and an IC 5 o value for Pf-3D7 LUC cells of about 9 nM.
  • the herein provided binding molecule, antibody and/or antigen-binding fragment of this antibody preferably comprises six CDRs encoded by nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 97, 99, 101 , 103, 105 and 107.
  • Said six CDRs preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 98, 100, 102, 104, 106 and 108, respectively.
  • said binding molecule, antibody and/or antigen-binding fragment of this antibody comprises V H and V L nucleic acid sequences having at least 75, 85, 90, preferably
  • V H and V L chains preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95,
  • said antibody is M071 Xi0081 .
  • M071 Xi0124 has particularly high inhibition rates and inhibits growth of Pf-3D7 LUC cells to about 94% as compared to untreated
  • Plasmodium falciparum in vitro cultures as well as Plasmodium falciparum in vitro cultures of the respective Plasmodium treated with a non-
  • Inhibition rates may have a variance of 15 to 20% due to Plasmodium biology, in particular due to schizogony.
  • M071 Xi0124 shows no cross-reactivity with other antigens, like GLURP, human insulin, lipopolysaccharide (E. coli) or dsDNA (salmon sperm) and an IC 5 o value for Pf-3D7 LUC cells of about 10 nM.
  • the herein provided binding molecule, antibody and/or antigen-binding fragment of this antibody preferably comprises six
  • Said six CDRs preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 1 10, 1 12, 1 14, 1 16, 1 18 and 120, respectively. More preferably, said binding molecule, antibody and/or antigen-binding fragment of this antibody comprises V H and V L nucleic acid sequences having at least 75, 85, 90, preferably
  • V H and V L chains preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95,
  • said antibody is M071 Xi0124.
  • M036Xi0326 has particularly high inhibition rates and inhibits growth of Pf-3D7 LUC cells to about 93%, of Pf-HB3 cells to about 87%, of Pf-DD2 cells to about 60%, of Pf-YT2 cells to about 61 % and of Pf-3D7 WT cells to about 75% as compared to untreated Plasmodium falciparum in vitro cultures as well as Plasmodium falciparum in vitro cultures of the respective Plasmodium treated with a non-Plasmodium falciparum reactive control antibody. Inhibition rates may have a variance of 15 to 20% due to Plasmodium biology, in particular due to schizogony.
  • M036Xi0326 shows an IC 5 o value for Pf-3D7 LUC cells of about 1 1 nM.
  • the herein provided binding molecule, antibody and/or antigen-binding fragment of this antibody preferably comprises six CDRs encoded by nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 121 , 123, 125, 127, 129 and 131 .
  • Said six CDRs preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 122, 124, 126, 128, 130 and 132, respectively. More preferably, said binding molecule, antibody and/or antigen- binding fragment of this antibody comprises V H and V L nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 221 and 223, respectively.
  • V H and V L chains preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 222 and 224, respectively. It is particularly preferred that said antibody is M036Xi0326.
  • M070Xi3195 has particularly high inhibition rates and inhibits growth of Pf-3D7 LUC cells to about 93% as compared to untreated Plasmodium falciparum in vitro cultures as well as Plasmodium falciparum in vitro cultures of the respective Plasmodium treated with a non- Plasmodium falciparum reactive control antibody. Inhibition rates may have a variance of 15 to 20% due to Plasmodium biology, in particular due to schizogony. In addition, M070Xi3195 shows an IC 5 o value for Pf-3D7 LUC cells of about 7 nM.
  • the herein provided binding molecule, antibody and/or antigen-binding fragment of this antibody preferably comprises six CDRs encoded by nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 133, 135, 137, 139, 141 and 143.
  • Said six CDRs preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 134, 136, 138, 140, 142 and 144, respectively.
  • said binding molecule, antibody and/or antigen- binding fragment of this antibody comprises V H and V L nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 225 and 227, respectively.
  • Said V H and V L chains preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 226 and 228, respectively. It is particularly preferred that said antibody is M070Xi3195.
  • M070Xi3062 has particularly high inhibition rates and inhibits growth of Pf-3D7 LUC cells to 87% as compared to untreated
  • Plasmodium falciparum in vitro cultures as well as Plasmodium falciparum in vitro cultures of the respective Plasmodium treated with a non-
  • Inhibition rates may have a variance of 15 to 20% due to Plasmodium biology, in particular due to schizogony.
  • M070Xi3062 shows an IC 5 o value for Pf-3D7 LUC cells of about 2 nM.
  • the herein provided binding molecule, antibody and/or antigen-binding fragment of this antibody preferably comprises six CDRs encoded by nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 145, 147, 149, 151 , 153 and 155.
  • Said six CDRs preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 146, 148, 150, 152, 154 and 156, respectively. More preferably, said binding molecule, antibody and/or antigen- binding fragment of this antibody comprises V H and V L nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 229 and 231 , respectively.
  • V H and V L chains preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 230 and 232, respectively. It is particularly preferred that said antibody is M070Xi3062.
  • M071 Xi2217 has particularly high inhibition rates and inhibits growth of Pf-3D7 LUC cells to about 92%, of Pf-HB3 cells to about 75%, of Pf-DD2 cells to about 32%, of Pf-YT2 cells to about 79% and of Pf-3D7 WT cells to about 21 % as compared to untreated Plasmodium falciparum in vitro cultures as well as Plasmodium falciparum in vitro cultures of the respective Plasmodium treated with an non-Plasmodium falciparum reactive control antibody. Inhibition rates may have a variance of 15 to 20% due to Plasmodium biology, in particular due to schizogony.
  • M071 Xi2217 shows an IC 50 value for Pf-3D7 LUC cells of about 5 nM.
  • the herein provided binding molecule, antibody and/or antigen-binding fragment of this antibody preferably comprises six CDRs encoded by nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 157, 159, 161 , 163, 165 and 167.
  • Said six CDRs preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 158, 160, 162, 164, 166 and 168, respectively. More preferably, said binding molecule, antibody and/or antigen- binding fragment of this antibody comprises V H and V L nucleic acid sequences having at least 75, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with nucleic acid sequences as set forth in SEQ ID NOs: 233 and 235, respectively.
  • V H and V L chains preferably comprise amino acid sequences having at least 80, 85, 90, preferably 95, 96, 97, 98, 99 or 100% overall sequence identity with amino acid sequences as set forth in SEQ ID NOs: 234 and 236, respectively. It is particularly preferred that said antibody is M071 Xi2217.
  • M071 Xi0092 has particularly high inhibition rates and inhibits growth of Pf-3D7 LUC cells to about 84% as compared to untreated Plasmodium falciparum in vitro cultures as well as Plasmodium falciparum in vitro cultures of the respective Plasmodium treated with a non- Plasmodium falciparum reactive control antibody. Inhibition rates may have a variance of 15 to 20% due to Plasmodium biology, in particular due to schizogony. In addition, M071 Xi0092 shows no cross-reactivity with other antigens, like GLURP, human insulin, lipopolysaccharide (E. coli) or dsDNA (salmon sperm) and an IC 5 o value for Pf-3D7 LUC cells of about 8 nM.
  • other antigens like GLURP, human insulin, lipopolysaccharide (E. coli) or dsDNA (salmon sperm) and an IC 5 o value for Pf-3
  • M036Xi0003 has particularly high inhibition rates and inhibits growth of Pf-3D7 LUC cells to about 46%, of Pf-HB3 cells to about 75%, of Pf-DD2 cells to about 82%, of Pf-YT2 cells to about 75% and of Pf-3D7 WT cells to about 69% as compared to untreated Plasmodium falciparum in vitro cultures as well as Plasmodium falciparum in vitro cultures of the respective Plasmodium treated with an non-Plasmodium falciparum reactive control antibody. Inhibition rates may have a variance of 15 to 20% due to Plasmodium biology, in particular due to schizogony.
  • M036Xi0003 shows no cross-reactivity with other antigens, like GLURP, human insulin, lipopolysaccharide (E. coli) or dsDNA (salmon sperm) and an IC 5 o value for Pf- 3D7 LUC cells of about 7.5 nM.
  • the binding molecule, antibody and/or antigen-binding fragment of this antibody of the present invention is M071 Xi0199, M071 Xi2204, M071 Xi0237, M071 Xi2127, M071Xi2057, M070Xi3010, M070Xi3062, M071 Xi2217, M071 Xi0227, M071 Xi0081 , M071Xi0124, M036Xi0326 or M070Xi3195.
  • the binding molecule, antibody and/or antigen-binding fragment of this antibody of the present invention is M071 Xi0199, M071 Xi2204, M071 Xi0237, M071 Xi2127, M071Xi2057, M070Xi3062, M071 Xi2217, M071 Xi0081 , M071 Xi0124, M036Xi0326 or M070Xi3195.
  • the binding molecule, antibody and/or antigen-binding fragment of this antibody of the present invention is M071 Xi0199, M071 Xi2204, M071 Xi2057, M070Xi3062, M071 Xi2217 or M071 Xi0124.
  • the binding molecule, antibody and/or antigen-binding fragment of this antibody of the present invention is M071 Xi0199.
  • the invention further provides nucleic acids encoding the binding molecules, antibodies or antibody fragments of these antibodies according to the invention. Also provided are nucleic acid molecules encoding the CDRs and/or the variable regions as provided herein.
  • the invention also relates to and provides vectors, such as expression vectors, comprising said nucleic acids, and host cells comprising said nucleic acids or said vectors.
  • the nucleic acid molecule of the invention encoding the antibody of the invention may be, e.g. DNA, cDNA, RNA or synthetically produced DNA or RNA or recombinantly produced chimeric nucleic acid molecule comprising any of those nucleic acid molecules either alone or in combination.
  • the nucleic acid molecule may also be genomic DNA corresponding to the entire gene or a substantial portion thereof or to fragments and derivatives thereof.
  • nucleotide sequence may correspond to the naturally occurring nucleotide sequence or may contain single or multiple nucleotide substitutions, deletions or additions required that the nucleic acid of the invention comprises nucleic acids provided herein, or such sequences wherein at least one amino acid is conservatively substituted or comprises the nucleic acids as defined by any nucleotide sequence provided herein.
  • the nucleic acid molecule is a cDNA molecule.
  • An embodiment of the invention also relates to a vector comprising the nucleic acid molecule in an expressible form.
  • the vector of the invention may be, for example, a phage, plasmid, viral or retroviral vector.
  • Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host/cells.
  • the nucleic acid molecule may be inserted into several commercially available vectors.
  • Non-limiting examples include prokaryotic plasmid vectors, such as the pUC-series, pBluescript (Stratagene), the pET-series of expression vectors (Novagen) or pCRTOPO (Invitrogen) and vectors compatible with an expression in mammalian cells like pREP (Invitrogen), pcDNA3 (Invitrogen), pCEP4 (Invitrogen), pMCI neo (Stratagene), pXT1 (Stratagene), pSG5 (Stratagene), EBO-pSV2neo, pBPV-1 , pdBPVMMTneo, pRSVgpt, pRSVneo, pSV2-dhfr, plZD35, pLXIN, pSIR (Clontech), pIRES-EGFP (Clontech), p
  • Plant expression vectors comprise pGEM-T (Promega), pCAMBIA 1391 (Cambia), GATEWAY (Invitrogen), pGreen and pGreenll (PGREEN).
  • Examples for plasmid vectors suitable for Pichia pastoris comprise e.g. the plasmids pAO815, pPIC9K and pPIC3.5K (all Intvitrogen).
  • vectors can contain one or more origin of replication (ori) and inheritance systems for cloning or expression, one or more markers for selection in the host, e. g., antibiotic resistance, and one or more expression cassettes.
  • origins of replication include, for example, the Col E1 , the SV40 viral and the M 13 origins of replication.
  • the coding sequences inserted in the vector can e.g. be synthesized by standard methods, or isolated from natural sources. Ligation of the coding sequences to transcriptional regulatory elements and/or to other amino acid encoding sequences can be carried out using established methods.
  • Transcriptional regulatory elements parts of an expression cassette
  • These elements comprise regulatory sequences ensuring the initiation of transcription (e. g., translation initiation codon, promoters, such as naturally-associated or heterologous promoters and/or insulators), internal ribosomal entry sites (IRES) (Owens, Proc. Natl. Acad. Sci.
  • polynucleotide of the invention is operatively linked to such expression control sequences allowing expression in prokaryotes or eukaryotic cells.
  • the vector may further comprise nucleotide sequences encoding secretion signals as further regulatory elements. Such sequences are well known to the person skilled in the art.
  • leader sequences capable of directing the expressed polypeptide to a cellular compartment may be added to the coding sequence of the polynucleotide of the invention. Such leader sequences are well known in the art.
  • the vector comprises a selectable marker.
  • selectable markers include neomycin, ampicillin, and hygromycine, kanamycin resistance and the like.
  • An expression vector according to this invention is capable of directing the replication, and the expression, of the polynucleotide and encoded antibody of this invention.
  • Suitable expression vectors which comprise the described regulatory elements are known in the art.
  • V H and V L region of the antibody of the invention may be encoded by different expression vectors.
  • nucleic acid molecules as described herein above may be designed for direct introduction or for introduction via liposomes, phage vectors or viral vectors (e.g. adenoviral, retroviral) into a host.
  • viral vectors e.g. adenoviral, retroviral
  • baculoviral systems or systems based on Vaccinia Virus or Semliki Forest Virus can be used as eukaryotic expression systems for the nucleic acid molecules of the invention.
  • a typical mammalian expression vector contains the promoter element, which mediates the initiation of transcription of mRNA, the protein coding sequence, and signals required for the termination of transcription and polyadenylation of the transcript. Moreover, elements such as origin of replication, drug resistance gene, regulators (as part of an inducible promoter) may also be included.
  • the lac promoter is a typical inducible promoter, useful for prokaryotic cells, which can be induced using the lactose analogue isopropyl ⁇ -thiogalactopyranosid ("IPTG").
  • IPTG lactose analogue isopropyl ⁇ -thiogalactopyranosid
  • the polynucleotide of interest may be ligated between e.g.
  • PelB leader signal which directs the recombinant protein in the periplasm and the gene III in a phagemid called pHEN4 (described in Ghahroudi et al, 1997, FEBS Letters 414:521 -526). Additional elements might include enhancers, Kozak sequences and intervening sequences flanked by donor and acceptor sites for RNA splicing. Highly efficient transcription can be achieved with the early and late promoters from SV40, the long terminal repeats (LTRs) from retroviruses, e.g., RSV, HTLVI, HIVI, and the early promoter of the cytomegalovirus (CMV).
  • LTRs long terminal repeats
  • CMV cytomegalovirus
  • cellular elements can also be used (e.g., the human actin promoter).
  • the recombinant polypeptide can be expressed in stable cell lines that contain the gene construct integrated into a chromosome.
  • the co-transfection with a selectable marker such as dhfr, gpt, neomycin, hygromycin allows the identification and isolation of the transfected cells.
  • the transfected nucleic acid can also be amplified to express large amounts of the encoded polypeptide.
  • the expression vectors will preferably include at least one selectable marker.
  • Such markers include dihydrofolate reductase, G418 or neomycin resistance for eukaryotic cell culture and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria.
  • appropriate hosts include, but are not limited to, bacterial cells, such as E. coli, Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast cells; insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS, HEK 293 and Bowes melanoma cells; and plant cells. Appropriate culture mediums and conditions for the above-described host cells are known in the art.
  • Another embodiment of the invention relates to a non-human host cell comprising the vector of the invention.
  • Said host cell may be a prokaryotic or eukaryotic cell.
  • the polynucleotide or vector of the invention which is present in the host cell may either be integrated into the genome of the host cell or it may be maintained extrachromosomally.
  • the nucleic acid molecule of the invention can be used for "gene targeting” and/or “gene replacement", for restoring a mutant gene or for creating a mutant gene via homologous recombination; see for example Mouellic, Proc. Natl. Acad. Sci. USA, 87 (1990), 4712-4716; Joyner, Gene Targeting, A Practical Approach, Oxford University Press.
  • Suitable prokaryotic host cells comprise e.g. bacteria of the species Escherichia, Bacillus, Streptomyces and Salmonella typhimurium.
  • Suitable eukaryotic host cells are e.g. fungal cells, inter alia, yeasts such as Saccharomyces cerevisiae or Pichia pastoris or insect cells such as Drosophila S2 and Spodoptera Sf9 cells and plant cells as well as mammalian cells. Appropriate culture media and conditions for the above-described host cells are known in the art.
  • Mammalian host cells that could be used include, human Hela, HEK293, H9 and Jurkat cells, mouse NIH3T3 and C127 cells, Cos 1 , Cos 7 and CV1 , quail QC1 -3 cells, mouse L cells, mouse C2C12 cells, BHK (baby hamster kidney cells) and Chinese hamster ovary (CHO) cells.
  • hosts such as primary mammalian cells such as mouse embryonic fibroblasts (MEF).
  • the recombinant antibody can be expressed in stable cell lines that contain the gene construct integrated into a chromosome.
  • said cell is a primary cell or primary cell line.
  • Primary cells are cells which are directly obtained from an organism.
  • the invention also relates transgenic non-human animals comprising one or more nucleic acid molecules of the invention that may be used to produce the antibody of the invention.
  • Antibodies can be produced in and recovered from tissue or body fluids, such as milk, blood or urine, of goats, cows, horses, pigs, rats, mice, rabbits, hamsters or other mammals. See, e. g., U. S. Patent Nos. 5,827,690, 5,756,687, 5,750,172, and 5,741 ,957.
  • the invention relates to a method for producing a binding molecule, antibody and/or antigen-binding fragment of this antibody of the invention comprising (a) culturing the host of the invention under conditions that allow synthesis of said antibody; and (b) recovering said antibody from said culture.
  • the transformed host cells can be grown in fermentors and cultured according to techniques known in the art to achieve optimal cell growth.
  • the whole binding molecules, antibodies or antigen-binding fragments of these antibodies of the present invention can be purified according to standard procedures of the art, including ammonium sulfate precipitation, affinity columns, column chromatography, gel electrophoresis and the like; see, Scopes, "Protein Purification", Springer-Verlag, N.Y. (1982).
  • the binding molecule, antibody and/or antigen-binding fragment of this antibody of the invention can then be isolated from the growth medium, cellular lysates, or cellular membrane fractions.
  • the isolation and purification of the, e.g., microbially expressed binding molecules, antibodies and/or antigen-binding fragments of these antibodies of the invention may be by any conventional means such as, for example, preparative chromatographic separations and immunological separations such as those involving the use of monoclonal or polyclonal antibodies directed, e.g., against the constant region of the antibody of the invention.
  • diagnostic and therapeutic methods using the binding molecules, antibodies, antigen-binding fragments of these antibodies or nucleotides of the present invention are provided.
  • binding molecules antibodies, antigen-binding fragments of these antibodies, nucleic acids, vectors or host cells may be used in vaccine therapy, as medicaments or in diagnosis of Plasmodium associated diseases, in particular malaria.
  • the invention relates to a pharmaceutical composition and a diagnostic composition comprising the binding molecules, antibodies and/or antigen-binding fragments of these antibodies of the invention, the nucleic acid molecule of the invention, the vector of the invention and/or the host cell of the invention.
  • composition as employed herein defines a composition comprising at least one antibody, nucleic acid molecule, vector, and/or host of the invention which are also referred in the following collectively as compound.
  • the diagnostic composition of the invention is useful in the detection of a Plasmodium infection of different cells, tissues or another suitable sample, comprising contacting a sample with a binding molecule, antibody and/or antigen- binding fragment of this antibody of the invention and detecting the presence of Plasmodium spec, preferably Plasmodium falciparum. Accordingly, the diagnostic composition of the invention may be used for assessing the onset or the disease status as defined herein below.
  • the infection status of malaria disease can be targeted with the binding molecule, antibody and/or antigen-binding fragment of this antibody of the invention.
  • the cells which have bound the binding molecule, antibody and/or antigen-binding fragment of this antibody of the invention might thus be attacked by immune system functions such as the complement system or by cell- mediated cytotoxicity.
  • the binding molecule, antibody and/or antigen-binding fragment of this antibody of the invention is coupled to a labelling group.
  • Such binding molecules, antibodies and/or antigen- binding fragments of these antibodies are particularly suitable for diagnostic applications.
  • the diagnostic composition of the invention can be administered as sole active agent or can be administered in combination with other agents.
  • the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising the binding molecule, antibody and/or antigen-binding fragment of this antibody of the invention, the nucleic acid molecule of the invention, the vector of the invention and/or the host cell of the invention.
  • the pharmaceutical composition is preferably administered to vertebrates. Most preferred it is administered to humans.
  • the pharmaceutical compositions described herein can be administered to the subject at a suitable dose.
  • the pharmaceutical composition for use in accordance with the present invention can be formulated in conventional manner according to methods found in the art, using one or more physiological carriers or excipient, see, for example Ansel et al., "Pharmaceutical
  • the pharmaceutical composition may, accordingly, be administered orally, parenterally, such as subcutaneously, intravenously, intramuscularly, intraperitoneally, intrathecally, transdermally, transmucosally, subdurally, locally or topically via iontopheresis, sublingually, by inhalation spray, aerosol or rectally and the like in dosage unit formulations optionally comprising conventional pharmaceutically acceptable excipients.
  • the pharmaceutical composition of the invention can take the form of, for example, tablets or capsules prepared by conventional means with pharmaceutical acceptable excipients such as binding agents (e.g., pregelatinised maize starch, polyvinylpyrrolidone, hydroxypropyl methylcellulose), fillers (e.g., lactose, microcrystalline cellulose, calcium hydrogen phosphate), lubricants (e.g., magnesium stearate, talc, silica), disintegrants (e.g., potato starch, sodium starch glycolate), or wetting agents (e.g., sodium lauryl sulphate).
  • binding agents e.g., pregelatinised maize starch, polyvinylpyrrolidone, hydroxypropyl methylcellulose
  • fillers e.g., lactose, microcrystalline cellulose, calcium hydrogen phosphate
  • lubricants e.g., magnesium stearate, talc, silica
  • disintegrants e.g., potato star
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium ion, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • the composition if desired, can also contain minor amounts emulsifying agents, or pH buffering agents. These compositions can be in the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences” by E.W. Martin. Such compositions will contain a therapeutically effective amount of the aforementioned compounds, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration.
  • Liquid preparations for oral administration can be in the form of, for example, solutions, syrups, or suspensions, or can be presented as a dry product for constitution with water or other suitable vehicle before use.
  • Such liquid preparation can be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol, syrup, cellulose derivatives, hydrogenated edible fats), emulsifying agents (e.g., lecithin, acacia), non-aqueous vehicles (e.g., almond oil, oily esters, ethyl alcohol, fractionated vegetable oils), preservatives (e.g., methyl or propyl-p-hydroxycarbonates, sorbic acids).
  • the preparations can also contain buffer salts, flavouring, colouring and sweetening agents as deemed appropriate.
  • Preparations for oral administration can be suitably formulated to give controlled release of the pharmaceutical composition of the invention.
  • the pharmaceutical composition of the invention is conveniently delivered in the form of an aerosol spray presentation from a pressurised pack or a nebulizer, with the use of a suitable propellant (e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas).
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit can be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, for example, gelatine, for use in an inhaler or insufflator can be formulated containing a powder mix of the pharmaceutical composition of the invention and a suitable powder base such as lactose or starch.
  • the pharmaceutical composition of the invention may be formulated for parenteral administration by injection, for example, by bolus injection or continuous infusion.
  • Site of injections include intravenous, intraperitoneal or subcutaneous.
  • Formulations for injection can be presented in units dosage form (e.g., in phial, in multi-dose container, sealed ampoules or vials, as an aqueous solution or as a lyophilized formulation for reconstitution), and with an added preservative.
  • the pharmaceutical composition of the invention can take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and can contain formulatory agents such as suspending, stabilizing, or dispersing agents.
  • the agent can be in powder form for constitution with a suitable vehicle (e.g., sterile pyrogen-free water) before use.
  • a suitable vehicle e.g., sterile pyrogen-free water
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition can also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilised powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients can be mixed prior to administration.
  • Transdermal compositions are typically formulated as a topical ointment or cream containing the active ingredient(s), generally in an amount ranging from about 0.01 to about 20% by weight, preferably from about 0.1 to about 20% by weight, preferably from about 0.1 to about 10% by weight, and more preferably from about 0.5 to about 15% by weight.
  • the active ingredients When formulated as a ointment, the active ingredients will typically be combined with either a paraffinic or a water- miscible ointment base. Alternatively, the active ingredients may be formulated in a cream with, for example an oil-in-water cream base.
  • transdermal formulations are well-known in the art and generally include additional ingredients to enhance the dermal penetration of stability of the active ingredients or the formulation. All such known transdermal formulations and ingredients are included within the scope of this invention.
  • the compounds of this invention can also be administered by a transdermal device. Accordingly, transdermal administration can be accomplished using a patch either of the reservoir or porous membrane type, or of a solid matrix variety.
  • composition of the invention can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • the pharmaceutical composition of the invention can also, if desired, be presented in a pack, or dispenser device which can contain one or more unit dosage forms containing the said agent.
  • the pack can for example comprise metal or plastic foil, such as blister pack.
  • the pack or dispenser device can be accompanied with instruction for administration.
  • composition of the invention can be administered as sole active agent or can be administered in combination with other agents, preferably ones that are known in the art to be suitable for treatment of the disease in question.
  • the pharmaceutical composition is formulated generally by mixing it at the desired degree of purity, in a unit dosage injectable form (solution, suspension, or emulsion), with a pharmaceutically acceptable carrier, i.e., one that is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation.
  • a pharmaceutically acceptable carrier i.e., one that is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation.
  • the formulations are prepared by contacting the components of the pharmaceutical composition uniformly and intimately with liquid carriers or finely divided solid carriers or both. Then, if necessary, the product is shaped into the desired formulation.
  • carrier vehicles include water, saline,
  • Non aqueous vehicles such as fixed oils and ethyl oleate are also useful herein, as well as liposomes.
  • the carrier suitably contains minor amounts of additives such as substances that enhance isotonicity and chemical stability.
  • Such materials are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, succinate, acetic acid, and other organic acids or their salts; antioxidants such as ascorbic acid; low molecular weight (less than about ten residues) polypeptides, e.g., polyarginine or tripeptides; proteins, such as serum albumin, gelatin, hydrophilic polymers such as polyvinylpyrrolidone; amino acids, such as glycine, glutamic acid, aspartic acid, or arginine; monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; counterions such as sodium; and/or nonionic surfactants such as polysorbates, poloxamers, or PEG.
  • buffers such as phosphate, citrate, succinate, ace
  • the components of the pharmaceutical composition to be used for therapeutic administration must be sterile. Sterility is readily accomplished by filtration through sterile filtration membranes (e.g., 0.2 micron membranes).
  • Therapeutic components of the pharmaceutical composition generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
  • Dosage regimens are adjusted to provide the optimum desired response (e.g., a therapeutic or a prophylactical response).
  • a single bolus e.g. an intravenously administered bolus
  • several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation.
  • parenteral e.g. intravenously administered
  • compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • inventive binding molecules inventive antibodies or inventive antigen-binding fragments of these antibodies as described herein, the dosage may range from about 0.0001 to 100 mg/kg, and more usually 5 to 50 mg/kg, of the host body weight.
  • dosages can be 5 mg/kg body weight, 10 mg/kg body weight, 20 mg/kg body weight, 30 mg/kg body weight, 40 mg/kg body weight or 50 mg/kg body weight.
  • An exemplary, non-binding and non-limiting treatment regime entails administration once per week, once every two weeks, once every three weeks, once every four weeks, once a month, once every 3 months or once every three to 6 months.
  • Exemplary dosage regimens for an anti-MSP 3 binding molecules, antibodies or antigen-binding fragments of these antibodies of this disclosure include 5 mg/kg body weight or 10 mg/kg body weight via intravenous administration, with the antibody being given using one of the following dosing schedules: (i) every ten days for two dosages, then every ten days if parasitemia is recurring.
  • two or more therapeutic binding molecules, antibodies and/or antigen-binding fragments of these antibodies with different binding specificities are administered simultaneously, in which case the dosage of each binding molecule, antibody and/or antigen-binding fragment of this antibody administered falls within the ranges indicated.
  • the binding molecule, antibody and/or antigen-binding fragment of this antibody is usually administered on multiple occasions. Intervals between single dosages can be, for example, weekly, monthly, every three months or yearly. Intervals can also be irregular as indicated by measuring blood levels of antibody to the target antigen in the patient.
  • the binding molecule, antibody and/or antigen-binding fragment of this antibody of the present invention can be administered as a sustained release formulation, in which case less frequent administration is required.
  • Dosage and frequency vary depending on the half-life of the antibody in the patient. In general, human antibodies show the longest half life, followed by humanized antibodies, chimeric antibodies, and nonhuman antibodies.
  • the dosage and frequency of administration can vary depending on whether the treatment is prophylactic or therapeutic. In prophylactic applications, a relatively low dosage is administered at relatively infrequent intervals over a long period of time. Some patients continue to receive treatment for the rest of their lives.
  • a relatively high dosage at relatively short intervals is sometimes required until progression of the disease is reduced or terminated, and preferably until the patient shows partial or complete amelioration of symptoms of disease. Thereafter, the patient can be administered a prophylactic regime.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of the present disclosure may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level will depend upon a variety of pharmacokinetic factors including the activity of the particular compositions of the present disclosure employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compositions employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a "therapeutically effective dosage" of an anti-MSP 3 binding molecule, antibody and/or antigen-binding fragment of this antibody of this invention preferably results in a decrease in severity of disease symptoms, an increase in frequency and duration of disease symptom-free periods, or a prevention of impairment or disability due to the disease affliction.
  • a "therapeutically effective dosage” preferably inhibits Plasmodium spec, growth by at least about 20%, more preferably by at least about 40%, even more preferably by at least about 60%, and still more preferably by at least about 80% relative to untreated subjects (or cells in cell based studies).
  • the ability of a compound to inhibit Plasmodium spec, growth can be evaluated in an in vitro model system predictive of efficacy.
  • One of ordinary skill in the art would be able to determine such amounts based on such factors as the subject's size, the severity of the subject's symptoms, and the particular composition or route of administration selected.
  • compositions of the present invention comprising binding molecules, antibodies and/or antigen-binding fragments of these antibodies of the invention are particularly useful in the treatment of infected patients, in particular patients infected by Plasmodium spec, in particular Plasmodium falciparum. Accordingly, compositions of the invention may be administered after infection occurred. Administering compositions of the invention after infection occurred may lead to reduced parasitemia and reduced symptoms, in particular symptoms associated with malaria, such as headache, fever, shivering, joint pain, vomiting, hemolytic anemia, jaundice, hemoglobin in the urine, retinal damage, convulsions, paroxysm, abnormal posturing, nystagmus, conjugate gaze palsy, opisthotonus, seizures, and/or coma.
  • the binding molecules, antibodies and/or antigen-binding fragments of these antibodies described and provided herein are useful to treat acute malaria as well to target late stages (e.g. asexual blood stages) of Plasmodium spec.
  • the inventive molecules are also likely useful for targeting early skin, skin-to-liver and/or liver stages of Plasmodium spec.
  • the inventive molecules may be used in the acute phase of Plasmodium spec, infection where active vaccination approaches would be bound to fail, since standard active vaccination needs time to activate the host's immune system to provide protection and during that time the parasite can still infect the host.
  • the inventive binding molecules, antibodies and/or antigen- binding fragments of these antibodies can be applied as they act quickly.
  • the binding molecules, antibodies and/or antigen-binding fragments of these antibodies described and provided herein may be used to target skin, liver as well as asexual blood phases of Plasmodium spec, (in particular of Plasmodium falciparum).
  • the herein provided binding molecules, antibodies and/or antigen-binding fragments of these antibodies may be used to ameliorate the symptoms of acute malaria, for example to ameliorate the fever attacks of malaria.
  • cyclic administration of the inventive molecules may be used to specifically ameliorate and/or abolish the fever attacks associated with malaria infection.
  • the means and methods provided herein may be used prophylactically in order to prevent the occurrence and/or manifestation of malaria.
  • the inventive binding molecules, antibodies and/or antigen-binding fragments of these antibodies may be administered prophylactically to persons travelling to endemic areas (such as sub-Saharan Africa).
  • binding molecules, antibodies and/or antigen-binding fragments of these antibodies described and provided herein are particularly useful in the prophylaxis as well as treatment of acute Plasmodium spec, infection since the main target of the inventive molecules is the merozoite stage of Plasmodium spec.
  • the herein provided binding molecules, antibodies and/or antigen-binding fragments of these antibodies may be used in co- therapy with at least one other active agent.
  • pharmaceutical compositions known to be effective in the treatment of Plasmodium spec. associated diseases, in particular malaria may be used in co-therapy together with the herein provided binding molecules, antibodies and/or antigen-binding fragments of these antibodies.
  • Examples of conventional anti-malaria medicaments include artemisinin, dihydroartemisinin, chloroquine, hydroxychloroquine, amodiaquine, lumefantrine, mefloquine, sulfadoxine/pyrimethamine, piperaquine, quinine, clindamycin as well as medicaments against fever (such as ibuprofen or acetylsalicylic acid). These medicaments may be used in co-therapy approaches with the herein defined inventive molecules.
  • the co-therapy may be a simultaneous, sequential or separate administration.
  • the two or more active components may be administered in one single administration form (e.g. in one single intravenous solution bag, or in one single tablet) or may be administered in separate dosages.
  • the co-administration of the binding molecules, antibodies and/or antigen-binding fragments of these antibodies as provided herein and the other anti-malaria medicament may lead to synergistic effects resulting in reduced symptoms and/or faster treatment of malaria as compared to the administration of the inventive binding molecules, antibodies or antigen-binding fragments of these antibodies or the other anti-malaria medicament(s) alone.
  • Symptoms associated with malaria include headache, fever, shivering, joint pain, vomiting, hemolytic anemia, jaundice, hemoglobin in the urine, retinal damage, convulsions, paroxysm, abnormal posturing, nystagmus, conjugate gaze palsy, opisthotonus, seizures, and/or coma.
  • the term "synergistic effect" is commonly known in the art and used herein to describe a situation where the combined effect of two or more active agents is greater than the sum of the individual active agents. In other words, two or more active agents can interact in a way that the presence of one active agent enhances or magnifies the effect(s) of the second. In particular, synergistic effects may lead to more effective treatment of symptoms associated with malaria, e.g. of the above described symptoms.
  • Treatment of Plasmodium spec, infection, in particular of Plasmodium falciparum infection may include both, treatment of blood stages as well as clearance of liver forms. Accordingly, as mentioned above, it is envisaged in accordance with the present invention that the herein provided binding molecules, antibodies and/or antigen-binding fragments of these antibodies are used to target blood and/or liver stages of Plasmodium spec, in particular of Plasmodium falciparum.
  • a composition of the present invention can be administered via one or more routes of administration using one or more of a variety of methods known in the art.
  • routes and/or mode of administration will vary depending upon the desired results.
  • Preferred routes of administration for binding molecules, antibodies or antigen-binding fragments of these antibodies of this invention include intravenous, intramuscular, intradermal, intraperitoneal, subcutaneous, spinal or other parenteral routes of administration, for example by injection or infusion.
  • parenteral administration means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural and intrasternal injection and infusion.
  • binding molecule, antibody and/or antigen-binding fragment of this antibody of the present invention is administered intravenously.
  • Binding molecules, antibodies and/or antigen-binding fragments of these antibodies as disclosed in this invention may also be used to monitor Plasmodium vaccination/infection status.
  • efficacy of vaccination e.g. a vaccination with MSP 3 or in a vaccination with antigens of MSP 3 as disclosed herein for use in vaccination of a subject/individual/human patient in need of such vaccination
  • Monitoring vaccination/infection status may, inter alia, comprise determining the antibody titer after vaccination (with e.g. MSP 3/MSP 3 antigens like the epitopes provided herein).
  • the binding molecules, antibodies or antigen-binding fragments of these antibodies as disclosed in this invention may also be used in control assays, e.g. as control titer to be used to monitor efficacy of vaccination.
  • the binding molecule, antibody and/or antigen- binding fragment of this antibody, the nucleic acid molecule, the vector or the host cell of the invention are particularly useful in treating or inhibiting the particular diseases mentioned herein.
  • Binding molecules, antibodies and/or antigen-binding fragments of these antibodies comprising the CDR and/or V H and/or V L sequences given below are particularly useful for use in the methods provided herein or as component of pharmaceutical and/or diagnostic compositions provided herein.
  • a binding molecule, antibody and/or antigen-binding fragment of this antibody comprising a V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 2, said CDR2 having the amino acid sequence SEQ ID NO: 4, and said CDR3 having the amino acid sequence SEQ ID NO: 6; and a V L which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 8, said CDR2 having the amino acid sequence SEQ ID NO: 10, and said CDR3 having the amino acid sequence SEQ ID NO: 12.
  • a binding molecule, antibody and/or antigen-binding fragment of this antibody comprising a V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 14, said CDR2 having the amino acid sequence SEQ ID NO: 16, and said CDR3 having the amino acid sequence SEQ ID NO: 18; and a V L which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 20, said CDR2 having the amino acid sequence SEQ ID NO: 22, and said CDR3 having the amino acid sequence SEQ ID NO: 24.
  • a binding molecule, antibody and/or antigen-binding fragment of this antibody comprising a V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 26, said CDR2 having the amino acid sequence SEQ ID NO: 28, and said CDR3 having the amino acid sequence SEQ ID NO: 30; and a V L which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 32, said CDR2 having the amino acid sequence SEQ ID NO: 34, and said CDR3 having the amino acid sequence SEQ ID NO: 36.
  • a binding molecule, antibody and/or antigen-binding fragment of this antibody comprising a V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 38, said CDR2 having the amino acid sequence SEQ ID NO: 40, and said CDR3 having the amino acid sequence SEQ ID NO: 42; and a V L which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 44, said CDR2 having the amino acid sequence SEQ ID NO: 46, and said CDR3 having the amino acid sequence SEQ ID NO: 48.
  • a binding molecule, antibody and/or antigen-binding fragment of this antibody comprising a V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 50, said CDR2 having the amino acid sequence SEQ ID NO: 52, and said CDR3 having the amino acid sequence SEQ ID NO: 54; and a V L which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 56, said CDR2 having the amino acid sequence SEQ ID NO: 58, and said CDR3 having the amino acid sequence SEQ ID NO: 60.
  • a binding molecule, antibody and/or antigen-binding fragment of this antibody comprising a V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 62, said CDR2 having the amino acid sequence SEQ ID NO: 64, and said CDR3 having the amino acid sequence SEQ ID NO: 66; and a V L which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 68, said CDR2 having the amino acid sequence SEQ ID NO: 70, and said CDR3 having the amino acid sequence SEQ ID NO: 72.
  • a binding molecule, antibody and/or antigen-binding fragment of this antibody comprising a V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 74, said CDR2 having the amino acid sequence SEQ ID NO: 76, and said CDR3 having the amino acid sequence SEQ ID NO: 78; and a V L which comprises the hypervariable regions CDR1 ', CDR2' and CDR3', said CDR1 ' having the amino acid sequence SEQ ID NO: 80, said CDR2' having the amino acid sequence SEQ ID NO: 82, and said CDR3' having the amino acid sequence SEQ ID NO: 84.
  • a binding molecule, antibody and/or antigen-binding fragment of this antibody comprising a V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 86, said CDR2 having the amino acid sequence SEQ ID NO: 88, and said CDR3 having the amino acid sequence SEQ ID NO: 90; and a V L which comprises the hypervariable regions CDR1 ', CDR2' and CDR3', said CDR1 ' having the amino acid sequence SEQ ID NO: 92, said CDR2' having the amino acid sequence SEQ ID NO: 94, and said CDR3' having the amino acid sequence SEQ ID NO: 96.
  • a binding molecule, antibody and/or antigen-binding fragment of this antibody comprising a V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 98, said CDR2 having the amino acid sequence SEQ ID NO: 100, and said CDR3 having the amino acid sequence SEQ ID NO: 102; and a V L which comprises the hypervariable regions CDR1 ', CDR2' and CDR3', said CDR1 ' having the amino acid sequence SEQ ID NO: 104, said CDR2' having the amino acid sequence SEQ ID NO: 106, and said CDR3' having the amino acid sequence SEQ ID NO: 108.
  • a binding molecule, antibody and/or antigen-binding fragment of this antibody comprising a V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 1 10, said CDR2 having the amino acid sequence SEQ ID NO: 1 12, and said CDR3 having the amino acid sequence SEQ ID NO: 1 14; and a V L which comprises the hypervariable regions CDR1 ', CDR2' and CDR3', said CDR1 ' having the amino acid sequence SEQ ID NO: 1 16, said CDR2' having the amino acid sequence SEQ ID NO: 1 18, and said CDR3' having the amino acid sequence SEQ ID NO: 120.
  • a binding molecule, antibody and/or antigen-binding fragment of this antibody comprising a V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 122, said CDR2 having the amino acid sequence SEQ ID NO: 124, and said CDR3 having the amino acid sequence SEQ ID NO: 126; and a V L which comprises the hypervariable regions CDR1 ', CDR2' and CDR3', said CDR1 ' having the amino acid sequence SEQ ID NO: 128, said CDR2' having the amino acid sequence SEQ ID NO: 130, and said CDR3' having the amino acid sequence SEQ ID NO: 132.
  • a binding molecule, antibody and/or antigen-binding fragment of this antibody comprising a V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 134, said CDR2 having the amino acid sequence SEQ ID NO: 136, and said CDR3 having the amino acid sequence SEQ ID NO: 138; and a V L which comprises the hypervariable regions CDR1 ', CDR2' and CDR3', said CDR1 ' having the amino acid sequence SEQ ID NO: 140, said CDR2' having the amino acid sequence SEQ ID NO: 142, and said CDR3' having the amino acid sequence SEQ ID NO: 144.
  • binding molecule, antibody and/or antigen-binding fragment of this antibody comprising a V H which comprises the hypervariable regions CDR1 ,
  • said CDR1 having the amino acid sequence SEQ ID NO: 146, said CDR2 having the amino acid sequence SEQ ID NO: 148, and said CDR3 having the amino acid sequence SEQ ID NO: 150; and a V L which comprises the hypervariable regions CDR1 ', CDR2' and CDR3', said CDR1 ' having the amino acid sequence SEQ ID NO: 152, said CDR2' having the amino acid sequence SEQ ID NO: 154, and said CDR3' having the amino acid sequence SEQ ID NO: 156.
  • a binding molecule, antibody and/or antigen-binding fragment of this antibody comprising a V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 158, said CDR2 having the amino acid sequence SEQ ID NO: 160, and said CDR3 having the amino acid sequence SEQ ID NO: 162; and a V L which comprises the hypervariable regions CDR1 ', CDR2' and CDR3', said CDR1 ' having the amino acid sequence SEQ ID NO: 164, said CDR2' having the amino acid sequence SEQ ID NO: 166, and said CDR3' having the amino acid sequence SEQ ID NO: 168.
  • a binding molecule, antibody and/or antigen-binding fragment of this antibody comprising a V H which comprises the hypervariable regions CDR1 , CDR2 and CDR3, said CDR1 having the amino acid sequence SEQ ID NO: 170, said CDR2 having the amino acid sequence SEQ ID NO: 172, and said CDR3 having the amino acid sequence SEQ ID NO: 174; and a V L which comprises the hypervariable regions CDR1 ', CDR2' and CDR3', said CDR1 ' having the amino acid sequence SEQ ID NO: 176, said CDR2' having the amino acid sequence SEQ ID NO: 178, and said CDR3' having the amino acid sequence SEQ ID NO: 180.
  • binding molecules particularly useful for use in the methods of the present invention or as component of the pharmaceutical and/or diagnostic composition of the present invention, which comprise the following V H and V L regions as determined by the amino acid sequences given below.
  • the invention thus provides for an antibody comprising the V H region determined by the amino sequence of SEQ ID NO. 182 and the V L region determined by the amino sequence of SEQ ID NO: 184.
  • an antibody comprising the V H region determined by the amino sequence of SEQ ID NO: 186 and the V L region determined by the amino sequence of SEQ ID NO: 188.
  • an antibody comprising the V H region determined by the amino sequence of SEQ ID NO: 190 and the V L region determined by the amino sequence of SEQ ID NO: 192.
  • an antibody comprising the V H region determined by the amino sequence of SEQ ID NO: 194 and the V L region determined by the amino sequence of SEQ ID NO: 196.
  • an antibody comprising the V H region determined by the amino sequence of SEQ ID NO: 198 and the V L region determined by the amino sequence of SEQ ID NO: 200.
  • an antibody comprising the V H region determined by the amino sequence of SEQ ID NO: 202 and the V L region determined by the amino sequence of SEQ ID NO: 204.
  • an antibody comprising the V H region determined by the amino sequence of SEQ ID NO: 206 and the V L region determined by the amino sequence of SEQ ID NO: 208.
  • an antibody comprising the V H region determined by the amino sequence of SEQ ID NO: 210 and the V L region determined by the amino sequence of SEQ ID NO: 212.
  • an antibody comprising the V H region determined by the amino sequence of SEQ ID NO 214 and the V L region determined by the amino sequence of SEQ ID NO: 216.
  • an antibody comprising the V H region determined by the amino sequence of SEQ ID NO: 218 and the V L region determined by the amino sequence of SEQ ID NO: 220. Also provided herein is an antibody comprising the V H region determined by the amino sequence of SEQ ID NO: 222 and the V L region determined by the amino sequence of SEQ ID NO: 224.
  • an antibody comprising the V H region determined by the amino sequence of SEQ ID NO: 226 and the V L region determined by the amino sequence of SEQ ID NO: 228.
  • an antibody comprising the V H region determined by the amino sequence of SEQ ID NO: 230 and the V L region determined by the amino sequence of SEQ ID NO: 232.
  • an antibody comprising the V H region determined by the amino sequence of SEQ ID NO: 234 and the V L region determined by the amino sequence of SEQ ID NO: 236.
  • an antibody comprising the V H region determined by the amino sequence of SEQ ID NO: 238 and the V L region determined by the amino sequence of SEQ ID NO: 240.
  • Antibodies of the present invention are also shown in the following tables 1 and 2.
  • each of these antibodies can bind to MSP 3
  • their CDRs can be combined to create other anti-MSP 3 antibodies or fragments of these antibodies of the invention.
  • MSP 3 binding of said antibodies generated by combination of the above described CDRs can be tested using the binding assays described in the Examples.
  • therapeutic antibodies of the invention comprise one or more of the CDR sequences above or a sequence at least 75% identical, preferably at least 80% identical thereto.
  • the therapeutic antibody of the invention comprises one or more of the CDR sequences above or a sequence at least 90% identical thereto (e.g. 90%, 95%, 96%, 97%, 98%, 99% or more).
  • the invention thus provides an isolated binding molecule, antibody and/or antigen- binding fragment of this antibody that specifically binds MSP 3 wherein said binding molecule, antibody and/or antigen-binding fragment of this antibody has one or more CDRs, preferably 2 or more CDRs, more preferably 3 or more CDRs, and still more preferably 6 CDRs, each CDR corresponding to a CDR chosen from the CDRs of M071 Xi0199, M071Xi2204, M071Xi0237, M071Xi2127, M071 Xi0092, M071 Xi2057, M070Xi3010, M071Xi0227, M071 Xi0081 , M071 Xi0124, M036Xi0326, M070Xi3195, M070Xi3062, M071 Xi2217 or M036Xi0003 as defined herein, or a CDR sequence having 75% or more (e.g. 80%,
  • the invention also provides a nucleic acid encoding one or more of the above listed CDR sequences or a CDR sequence at least 75% identical, preferably at least 80% identical thereto.
  • the nucleic acid encodes one or more of the above listed CDR sequences or a CDR sequence at least 90% identical thereto.
  • the invention also involves one or more of the CDR sequences above or a CDR sequence at least 75% identical, preferably at least 80% identical, more preferably at least 90% identical thereto wherein said CDR sequences is in the context of an antibody framework.
  • the antibody framework is a human antibody framework.
  • FIG. 1 Memory B cell sorting.
  • A Relative 3D7 Luc inhibition activity of purified serum IgG from study participants at 100 g/ml in comparison to polyclonal serum IgG preparations from non-immune controls (0% inhibition) and 50 mM chloroquine (100% inhibition, dashed gray line). Dots represent individual samples. Selected donors for antibody cloning are indicated.
  • B Gating strategy for the flow cytometric isolation of CM and AtM. Representative plots from MP070 are shown.
  • C Frequency of circulating CM and AtM.
  • D IgG GMZ2 reactivity in serum as measured by ELISA. Dotted line shows a Pf-na ' fve control serum.
  • E Flow cytometric analysis of GMZ2-reactive CM and AtM in peripheral blood from MP036, MP070 and MP071 compared to a malaria na ' fve donor
  • F Frequency of circulating GMZ2-reactive CM and AtM in all donors as determined in (E).
  • Figure 2 Antibody repertoire of anti-GMZ2 CM and AtM.
  • IGHV and IGHJ A
  • IGLVK and IGLJK B
  • IGLVL and IGLJL C
  • Numbers of sequences analyzed are indicated. P-values were calculated using Fisher's exact test.
  • D Absolute number of V segment somatic hypermutations. Geometric means with s.d. and the absolute numbers of sequences analyzed are indicated. P-values were determined using Student's t-test.
  • E Shaded pie areas indicate clonally expanded GMZ2-reactive CM and AtM. Absolute numbers of sequences analyzed are indicated.
  • Dashed line indicates the threshold OD 0 5nm for positive reactivity.
  • Light grey line at the bottom represents the negative control antibody mGO53 (Wardemann (2003) Science;
  • N indicates the numbers of tested antibodies.
  • B Antibody reactivity (light grey) with Pf schizonts (dark grey) in iRBCs as measured by indirect immunofluorescence.
  • C-D ELISA reactivity of GMZ2-reactive antibodies
  • A with MSP3
  • C and GLURP
  • E Bars summarize the mean frequency of MSP3, GLURP and MSP3+GLURP reactive antibodies with sem in all three donors.
  • FIG. 4 CM and AtM antibody polyreactivity.
  • A ELISA reactivity with dsDNA, LPS and insulin of CM and AtM antibodies from MP071 .
  • B Frequency of polyreactive CM and AtM antibodies from MP036, MP070, and MP071 .
  • (C) Symbols indicate antibody polyreactivity as the mean area under curve (AUC) for reactivity with dsDNA, insulin, and LPS. Horizontal lines indicate mean levels of antibody polyreactivity and sem in all three donors. P-values were calculated using ANOVA test.
  • FIG. 5 Pf merozoite neutralizing activity of CM and AtM antibodies.
  • A 3D7 Luc transgenic parasite growth in the presence of individual antibodies (black lines; MP070) or the indicated controls.
  • B Relative 3D7 Luc inhibition activity of GMZ2-reactive antibodies at 6.4 g/ml. Horizontal lines indicate mean inhibition activity.
  • C 3D7 Luc inhibition activity of individual MSP3, GLURP and polyreactive CM and AtM antibodies as in (B).
  • D Relative Pf 3D7, HB3 (Honduras), DD2 (Thailand) and IT4 (Brazil) inhibition activity compared to chloroquine (100%). Parasites were detected using SYBR green.
  • AtM express surface markers associated with B cell activation and show signs of recent cell divisions.
  • Figure 7 Study participants blood count. Blood count of three male donors from the Lambarene area, Gabon.
  • Figure 9 Overview of antibodies isolated from donor MP036. Ig gene and antibody reactivity information of antibodies isolated from donor MP036.
  • Figure 10 Overview of antibodies isolated from donor MP070. Ig gene and antibody reactivity information of antibodies isolated from donor MP070.
  • Figure 11 Overview of antibodies isolated from donor MP071. Ig gene and antibody reactivity information of antibodies isolated from donor MP071 .
  • FIG. 12 Table giving percentage of Plasmodium growth inhibition Antibodies M071 Xi0199, M071Xi2204, M071Xi0237, M071Xi2127, M071 Xi0092, M071 Xi2057, M070Xi3010, M071Xi0227, M071Xi0081 , M071 Xi0124, M036Xi0326, M070Xi3195, and M036Xi0003 have been analyzed using methods described above in order to assess their ability to inhibit growth of different Plasmodium strains.
  • the present invention is additionally described by way of the following illustrative non- limiting examples that provide a better understanding of the present invention and of its many advantages.
  • a panel of recombinant monoclonal antibodies from single GMZ2-binding primary human memory B cells from immune adults from a Pf endemic area was created (Lambarene, Gabon).
  • 201 recombinant monoclonal antibodies from classical and atypical memory B cells were screened for reactivity with the two protein subunits MSP3 2 i 0-380 3nd GLURP27-500 that are contained in the GMZ2 fusion protein and are expressed in Pf blood stages.
  • MSP3-reactive antibodies were identified. The majority of MSP3-reactive antibodies showed cross-reactivity with GLURP and/or polyreactivity with structurally diverse self- and non-self antigens (insulin, dsDNA, LPS) as tested by ELISA. Surprisingly, antibodies of the present invention showed specificity with MSP3 and lacked cross- /polyreactivity with GLURP or other self- and non-self antigens in the ELISA. At the same time, antibodies showed inhibition of diverse Pf strains in in vitro assays. Further, antibodies of the present invention, in particular M071 Xi0199, showed high affinity with low off-rate and specificity for MSP3 as reflected by surface plasmon resonance measurements. Specifically, antibodies, in particular M071 Xi0199, of the present invention recognized a conserved epitope in the range of aa 264 and 276 as determined by ELISA using overlapping peptides.
  • M071 Xi0199 carried IgH and IgL somatic mutations and used the IGHV3-74/IGHJ4 rearrangement in association with an IGKV3-20/IGKJ2 gene.
  • IGHV3-74 was used by only 5 out of 201 cloned antibodies, and by only one out of 93 anti-MSP3 antibodies.
  • Peripheral blood mononuclear cells were purified using Percoll gradient density centrifugation. Flow cytometric analyses were carried out on an LSR II instrument (BD Bioscience). IgG memory B cells were isolated using an ARIA II flow cytometric cell sorter (BD Biosciences). Pf GLURP, MSP3 and GMZ2 antigens were produced and purified as previously described (Theisen et al. (2004) Vaccine 22:1 188-98). GMZ2 was chemically coupled to Alexa-647 according to the manufacturer's instructions (Life Technologies).
  • mouse antibodies were used: anti-human CD19-PE-Cy7, anti-human lgG-APC-H7, anti-human CD27-FITC, anti- human CD38-V450, anti-human CD86-FITC, anti-human CD138-FITC (all from Beckton Dickinson), anti-human CD21 -PE, anti-human CD21 -APC, anti-human CD24-biotin, anti-human CD27-biotin, anti-human CD84-biotin, anti-human HLA-DR-APC (all from Ebioscience), anti-human Ki-67-PE (Biolegend), and anti-human FcRL4-biotin antibody (obtained from G.H.
  • Single-cell sorting and antibody cloning was performed as previously described with the following modifications: cells were sorted into 384-well plates and volumes of RT-PCR reactions were scaled by a factor of 0.25 (Scheid et al. (201 1 ) Science 333, 1633-7; Tiller et al. (2008) Journal Immunology Methods 329, 1 12-24). In brief, single cells were isolated using flow cytometric cell sorting. cDNA was generated in the original 384-well sort plates. Ig gene transcripts were amplified by nested RT-PCR, sequenced and cloned into lgG1 heavy and Igk or Igl light chain expression vectors, respectively.
  • HEK293T cells were co-transfected with Ig heavy and matching Ig light chain plasmids.
  • Recombinant monoclonal antibodies were purified from culture supernatant under sterile conditions using protein G beads.
  • Antibodies were eluted from protein G beads using 0.1 M glycine (pH3) and neutralized in 10 x PBS for use in ELISA and neutralization assays. IgG concentrations were determined by ELISA.
  • ELISAs antigens were coated at 10 g/ml (double-stranded DNA, LPS) or 5 g/ml
  • IgG-HRP antibodies were detected using one-step ABTS substrate (Roche). Area- under-curve (AUC) values were calculated using Graphpad PRISM. Antibodies were considered polyreactive when they recognized at least two out of the three structurally different antigens: double-stranded DNA, insulin and LPS. Internal high (ED38) and low (JB40) positive and negative (mGO53) control antibodies were used in all assays to set thresholds for polyreactivity (Meffre et al. (2004) Journal experimental medicine 199, 145-50; Tiller et al. (2008) Journal Immunology Methods 329, 1 12-24; Wardemann et al. (2003) Science 301 , 1374-7).
  • Plasmodium falciparum clone 3D7, DD2, HB3, and IT2 were cultivated at 5% hematocrit in RPMI 1640 supplemented with 0.05 g/l hypoxanthine (Sigma), 25 mM HEPES, 0.5% Albumax II (Life Technologies), 0.25% sodium bicarbonate and 0.01 mg/ml gentamicin (Sigma). All cultures were kept at 37 °C under 90% nitrogen, 5% oxygen, and 5% carbon dioxide. 3D7 Luc cultures were supplemented with 40 ng/ml pyrimethamine to ensure transgene expression.
  • Neutralization/Inhibition assays were performed with synchronized late stage 3D7 Luc or 3D7, DD2, and HB3 cultures at a starting parasitemia of 0.2% or 0.5%, respectively. Parasites were cultured in duplicates in the presence of recombinant monoclonal antibodies or protein G purified serum IgG. Uninfected cultures, chemical inhibitors (chloroquine and artemisinin at 50 mM), whole serum (10%) and mGO53 (non-Pf reactive recombinant monoclonal antibody) were used as internal controls.
  • 3D7 Luc growth was measured following merozoite release and re-invasion after 24 h by quantifying luciferase activity using Bright-Glo Luciferase substrate (Promega). Arbitrary luminescence units were measured on a VICTOR light reader (Perkin Elmer, 1420 Luminescence Counter). Growth of 3D7, HB3, IT2 and DD2 cultures was assessed after 48h using SYBR green (Smilkstein et al. (2004) Antimicrob. Agents Chemotherapy 48, 1803-6). Fluorescence was measured on a Fluoroskan Ascent reader (Thermo Labsystems).
  • Late stage percoll-sorbitol purified Pf 3D7 iRBCs were allowed to settle on poly-L lysine coated glass slides.
  • Cells were fixed with 4% paraformaldehyde/0.0075% glutaraldehyde before permeabilization with 0.2% Triton X-100. Blocking with BSA (3%) was performed before incubation with individual human recombinant monoclonal antibodies (10 ug/ml). Bound antibodies were detected using Cy3-conjugated mouse anti-human IgG-Fc (Jackson Immuno Research). Hoechst33342 was used to stain parasite DNA (Life Technologies). Images were acquired on a Leica DMR microscope with HCX PL FLUOTAR 100x/1 .3 objective using a Nikon DXM 1200F digital camera. Nikon ACT-1 Vers. 2.7 was used as acquisition software.
  • CM, AtM, naive B cells (CD19+lgM+lgD+) and plasma blasts (CD19+CD27+CD38+ lgG+) were bulk isolated by flow cytometric cell sorting.
  • Secretory and membrane IgG transcipts were amplified by RT-PCR using a IGHV-gene forward primer mix and 5- TCCTCGCGCGACCCCGAGAGC-3 (secretory) or 5-
  • GTCCACAGCCCGTCCAGCTCCC-3 (membrane) as reverse primer, respectively.
  • Amplification was performed using HotStar Taq (Qiagen) at 94°C-15 min, 50 x (94 °C- 30s, 68 °C-45s, 72 °C-30s), 72 °C-10 min for secretory and 94 °C - 15 min, 50 x (94 °C- 30s, 65 °C-45s, 72 °C 2min), 72 °C -10 min for membrane IgG transcripts.
  • PCR products were separated on 2% agarose gels.
  • MSP3 (PF10_0345) and GLURP (PF10_0344) SNP analysis was performed using next- generation sequencing data of 222 Pf isolates supplied by the MalariaGen Genomic Epidemiology Network ((Manske et al. (2012) Nature 487, 375-9, Data Release 1 .0).
  • CM and AtM cells differed in their surface phenotype but were derived from the same population, the likelihood of not observing any shared clusters within an individual donor can be approximated as (1 - a / c) A b, where c is the calculated overall number of clusters, a the number of unique clusters sampled from the first population and b the number of unique samplings from the second population.
  • Antibodies of the present invention possess diagnostic and therapeutic value due to its antigen-specificity, high affinity with low k 0ff -rate, and broad Pf inhibitory activity.
  • antibodies may be used as standard in tests that measure IgG serum antibody levels against Pf blood stages. These antibodies could replace pooled serum preparations that are currently used to assess antibody levels, e.g. in response to immunization with MSP3 or after natural infection.
  • the antibody may also be used to detect Pf blood stage parasites for example by flow cytometry or indirect immunofluorescence of erythrocytes from infected human donors.
  • the high degree of MSP3 gene homology in wild Pf isolated suggests that the claimed antibodies are suited to detect Pf strains from geographic areas around the world.
  • claimed antibodies could be of therapeutic use in passive immunization strategies to treat acutely infected patients with live-threatening malaria symptoms, especially in cases of infection with drug-resistant Pf strains.
  • the antibodies may be used as prophylactic treatment in high-risk groups, e.g. travellers.
  • the target epitope identified by the present antibodies may be exploited as subunit vaccine candidate.
  • the high degree of gene homology in the target epitope of MSP3 (Manske et al. (2012) Nature 487, 375-9) and the broad antibody Pf inhibitory activity makes the target epitope an ideal candidate for the development of a subunit vaccine.
  • Assays used to test effectiveness of the antibodies provided herein are well known in the art.
  • the inventors determined Plasmodium growth inhibition of a panel of antibodies according to an assay as described by Lucumi et al. (2010) Antimicrobial Agents Chemotherapy 54, 3597-604 and in the following.
  • Growing Plasmodium falciparum cultures were synchronized by sorbitol lysis one day before incubation with human recombinant monoclonal antibodies by pelleting asynchronous cultures by centrifugation at 2000 g and resuspension in 5% sorbitol/ddH 2 O.
  • Cultures were incubated for 15 min at 37°C before spinning at 900 g for 10 min to sediment intact ring containing and uninfected RBCs. Cultures were washed twice with RPMI and ring containing fraction was resuspended in medium at 5% hematocrit, gas (80% N 2 , 15% CO2, 5% O2) and returned to 37°C incubator for 24h. Parasitemia was determined by Giemsa-stain and the culture was diluted to a starting parasitemia of 0.2%.
  • Control preparations comprising antibody-free solution, uninfected RBCs at same hematokrit, IgG fraction from malaria-na ' fve individuals, chloroquine (50 mM final concentration) and purified IgGs from immune individuals were prepared.
  • dilution series containing antibody-solution and incomplete Plasmodium culture medium (0.5% Albumax, 0.25% sodium bicarbonate, 0.01 mg/ml gentamicin, 0.05 g/l hypoxanthine, 40 mg/ml pyromethanin in 1 I RPMI) were prepared, to which Pf-infected RBC-solution was added. Solutions were incubated in a gas chamber at 37°C for 24h.
  • Antibody M071Xi0199 directly inhibits Pf-merozoite growth with an IC 5 o of about 5 nM by binding to the Pf-MSP3 epitope EEEAEETEEEELE (amino acid 264-276, Genbank accession number L07944) with an K D of 4.44 x 10 "8 M.
  • M071Xi0199-VH-CDR2-aa RMSRDGSTVSYADSVKG (SEQ ID NO: 4)
  • VDJ M071Xi0199-VH-complete-nucleotide sequence
  • GTCACCGTCTCCTCA (SEQ ID NO: 181 )
  • M071Xi0199-VH-CDR1 -nucleotide AGGTACTGGATGCAC (SEQ ID NO: 1 )
  • M071Xi0199-VH-CDR3-nucleotide C AAATTTTCCTG G G CTAC (SEQ ID NO: 5)
  • M071Xi0199-VL-CDR2-nucleotide GCTGCATCCGACAGGGCCACT (SEQ ID NO: 9)
  • M071Xi0199-VL-CDR3-nucleotide CTCCAATATGGAAGCTCACCTCACAGT (SEQ ID NO: 1 1 )
  • Antibody M071 Xi2204 directly neutralizes Pf-merozoite replication with an IC 5 o of 1 1 .93 nM by binding to an epitope within amino acids 183 - 354 of P -MSP3 (Genbank accession number L07944).
  • M071Xi2204-VH-CDR2-aa YIRNKAYGGTTEYAASMRG (SEQ ID NO: 16)
  • M071Xi2204-VH-CDR3-aa EGTAMVHTFDI (SEQ ID NO: 18)
  • M071Xi2204-VH-CDR1 -nucleotide GATTATGCTATGAGC
  • M071Xi2204-VL-CDR2-aa DVSNRPS (SEQ ID NO: 22)
  • M071Xi2204-VL-CDR3-aa SSYTSSSSLV (SEQ ID NO: 24)
  • M071Xi2204-VL-CDR2-nucleotide GATGTCAGTAATCGGCCCTCA (SEQ ID NO: 21 )
  • M071Xi2204-VL-CDR3-nucleotide AGCTCATATACAAGCAGCAGCTCTCTCGTC (SEQ ID NO: 23)
  • Antibody M071Xi0237 directly neutralizes Pf-merozoite replication with an IC 5 o of 10.10 nM by binding to an epitope within amino acids 183 - 354 of P -MSP3 (Genbank accession number L07944).
  • M071Xi0237-VH-CDR2-aa SISSSAGTIYYADSVKG (SEQ ID NO: 28)
  • M071Xi0237-VH-CDR1 -nucleotide GACTACTACATGAAC (SEQ ID NO: 25)
  • M071Xi0237-VL-CDR2-nucleotide GGTGCATCCACCAGGGCCGCT (SEQ ID NO:
  • M071Xi0237-VL-CDR3-nucleotide CAGCAGTATAATACCTGGCCGACG (SEQ ID NO: 35)
  • Antibody M071Xi2127 directly neutralizes Pf-merozoite replication with an IC 5 o of 7.53 nM by binding to an epitope within amino acids 183 - 354 of P -MSP3 (Genbank accession number L07944).
  • GGAACCACGGTCACCGTCTCCTCAG (SEQ ID NO: 193) M071Xi2127-VH-CDR1 -nucleotide: ACCTATGGTATCAGC (SEQ ID NO: 37)
  • M071Xi2127-VH-CDR3-nucleotide GGCAGGGTGGCCGCGGGTTTTGACTAC (SEQ ID NO: 41 )
  • M071Xi2127-VL-CDR2-nucleotide AGTAATAATCAGCGGCCCTCA (SEQ ID NO: 45)
  • Antibody M071 Xi0092 directly neutralizes Pf-merozoite replication with an IC 5 o of 7.89 nM by binding to an epitope within amino acids 183 - 354 of P -MSP3 (Genbank accession number L07944).
  • M071Xi0092-VH-CDR1 -nucleotide AGTTCTGACTACTACTGGGCC (SEQ ID NO. 49)
  • M071Xi0092-VH-CDR2 -nucleotide AGTTCTGACTACTACTGGGCC (SEQ ID NO. 49)
  • CAAATTTATTATAGAGGGACCACCTACTA CAACCCGTCCCTCAAGAGT (SEQ ID NO: 51 )
  • M071Xi0092-VL-CDR1 -aa RSSQSLEHSDENTYLS (SEQ ID NO: 56)
  • M071Xi0092-VL-CDR3-aa MQAKQFPWT (SEQ ID NO: 60)
  • M071Xi0092-VL-CDR1 -nucleotide AGGTCTAGTCAAAGCCTCGAACACAGTGA
  • M071Xi0092-VL-CDR2-nucleotide AAGATCTCTCACCGGTTCTCT (SEQ ID NO: 57)
  • M071Xi0092-VL-CDR3-nucleotide ATGCAAGCAAAACAATTTCCGTGGACG (SEQ ID NO: 59) 6.) Antibody ⁇ 071 ⁇ 2057
  • Antibody M071Xi2057 directly neutralizes Pf-merozoite replication with an IC 5 0 of 20.34 nM by binding to an epitope within amino acids 183 - 354 of P -MSP3 (Genbank accession number L07944).
  • M071Xi2057-VH-CDR1 -nucleotide GAATATGGCTTGAAC (SEQ ID NO: 61 )
  • M071Xi2057-VL-CDR2-nucleotide GGGGCCTCCACCCGGGAATCC (SEQ ID NO: 69)
  • M071Xi2057-VL-CDR3-nucleotide CAACAATATTATAGTAATCCGATCACG (SEQ ID NO: 71 )
  • Antibody M070Xi3010 directly neutralizes Pf-merozoite replication with an IC 5 o of 8.44 nM by binding to an epitope within amino acids 183 - 354 of P -MSP3 (Genbank accession number L07944).
  • VDJ M070Xi3010-VH-complete-nucleotide sequence
  • M070Xi3010-VH-CDR1 -nucleotide AATTACTTTATGCAC (SEQ ID NO: 73)
  • EIVLTQSPDTLSLSPGERATLSCRANQSVTTDYLAWYKVKFGQAPRLLIYHTSRRAIGIP DRFSGSGSGTDFTLTISRLEPDDFVLYYCQQYENSPGTFGQGTKVEIK (SEQ ID NO: 208) M070Xi3010-VL-CDR1 -aa: RANQSVTTDYLA (SEQ ID NO: 80)
  • M070Xi3010-VL-CDR2-nucleotide CATACATCCAG AAG G GCCATT (SEQ ID NO: 81 )
  • M070Xi3010-VL-CDR3-nucleotide CAGCAATATGAAAACTCACCAGGAACG (SEQ ID NO: 83)
  • Antibody M071 Xi0227 neutralizes Pf-merozoite replication with an IC 5 o of 1 1 .75 nM by binding to an epitope within amino acids 183 - 354 of P -MSP3 (Genbank accession number L07944).
  • CTGGTCACCGTCTCCTCAG SEQ ID NO. 209
  • M071Xi0227-VH-CDR1 -nucleotide AATTATTGGATCGCC (SEQ ID NO: 85)
  • CTCATCTATCCACGGGACTCTGATACCA GATACAGCCCGTCCTTCCAAGGC SEQ ID NO: 87
  • M071Xi0227-VH-CDR3-nucleotide GGACAATTTAGATTCGCCCCC (SEQ ID NO: 89)
  • M071Xi0227-VL complete aa sequence (VJ):
  • M071Xi0227-VL-CDR2-nucleotide GATGCATCCAATTTGCAGACA (SEQ ID NO: 93)
  • M071Xi0227-VL-CDR3-nucleotide CAACAGTCTCATATTGTCCCGGTTACT (SEQ ID NO. 95)
  • Antibody M071 Xi0081 directly neutralizes Pf-merozoite replication with an IC 5 o of 8.93 nM by binding to an epitope within amino acids 183 - 354 of P -MSP3 (Genbank accession number L07944).
  • M071Xi0081-VH-CDR3-aa IGGSREGFQH (SEQ ID NO: 102)
  • M071Xi0081 -VH-CDR1 -nucleotide AGTCACTACTGGAGC (SEQ ID NO: 97) ⁇ 071 ⁇ 0081 -VH-CDR2 -nucleotide:
  • M071Xi0081 -VL-CDR2-nucleotide GATGATTCCGACCGGCCCTCA (SEQ ID NO:
  • Antibody M071 Xi0124 directly neutralizes Pf-merozoite replication with an IC 5 o of 10.02 nM by binding to an epitope within amino acids 183 - 354 of P -MSP3 (Genbank accession number L07944).
  • M071Xi0124-VH-CDR2-aa GTNWNGGRTAYADFVLG (SEQ ID NO: 1 12)
  • M071Xi0124-VH-CDR3-aa VPLSNGWHLSYFDY (SEQ ID NO: 1 14) M071Xi0124-VH-complete-nucleotide sequence (VDJ)
  • M071Xi0124-VH-CDR1 -nucleotide GATTATGGCATGAGC (SEQ ID NO: 109)
  • M071Xi0124-VL-CDR1 -aa RTSQSLASINLA (SEQ ID NO: 1 16)
  • M071Xi0124-VL-CDR2-nucleotide GATACTTCCACCAGGGCCACT (SEQ ID NO:
  • M071Xi0124-VL-CDR3-nucleotide CAGCAGTATGGTACCTTACCTCGCACT (SEQ ID NO: 1 19) 11.) Antibody ⁇ 036 ⁇ 0326
  • Antibody M036Xi0326 directly neutralizes Pf-merozoite replication with an IC 5 0 of 1 1 .14 nM by binding to an epitope within amino acids 183 - 354 of P -MSP3 (Genbank accession number L07944).
  • M036Xi0326-VH-CDR3-aa ELRRHYYDLRAFDI (SEQ ID NO: 126)
  • M036Xi0326-VH-CDR1 -nucleotide TACTCTGCCATGAAC (SEQ ID NO: 121 )
  • M036Xi0326-VL-CDR2-nucleotide ACTAATAATCAGCGGTCCTCA (SEQ ID NO:
  • Antibody M070Xi3195 directly neutralizes Pf-merozoite replication with an IC 5 o of 7.04 nM by binding to an epitope within amino acids 183 - 354 of P -MSP3 (Genbank accession number L07944).
  • M070Xi3195-VH-CDR1 -nucleotide AGTTACTACGGGACC (SEQ ID NO: 133)
  • ATCAAAC (SEQ ID NO: 227)
  • M070Xi3195-VL-CDR2-nucleotide TGGGCTTCTACCCGGGAGTCC (SEQ ID NO: 141 )
  • M070Xi3195-VL-CDR3-nucleotide CAACAATATTATACTACTTCCTACGCT (SEQ ID NO: 143)
  • Antibody M070Xi3062 directly neutralizes Pf-merozoite replication with an IC 5 o of 2.26 nM by binding to an epitope within amino acids 183 - 354 of P -MSP3 (Genbank accession number L07944).
  • M070Xi3062-VH-CDR1 -nucleotide GGTTACTACTGGAGC (SEQ ID NO: 145)
  • M070Xi3062-VL-CDR3-aa AAWDDSLSVWV (SEQ ID NO: 156)
  • M070Xi3062-VL-CDR2-nucleotide AGGAATAATCAGCGGCCCTCA (SEQ ID NO: 153)
  • Antibody M071 Xi2217 directly neutralizes Pf-merozoite replication with an IC 5 o of 5.01 nM by binding to an epitope within amino acids 183 - 354 of P -MSP3 (Genbank accession number L07944).
  • M071Xi2217-VH-CDR2-aa RIIPFLCMSNSAQRFQD (SEQ ID NO: 160)
  • M071Xi2217-VH-CDR1 -nucleotide AGCTATACAATCAAC (SEQ ID NO: 157)
  • GTCCTAG (SEQ ID NO: 235) ⁇ 071 ⁇ 2217-VL-CDR1 -nucleotide:
  • M071Xi2217-VL-CDR2-nucleotide GAGGATGACCAAAGACCCTCT (SEQ ID NO:
  • M071Xi2217-VL-CDR3-nucleotide CAGTCTTATGATGCCAGCAATCATCTTTGGGTT
  • Antibody M036Xi0003 directly neutralizes Pf-merozoite replication with an IC 5 o of 7.36 nM by binding to an epitope within amino acids 183 - 354 of P -MSP3 (Genbank accession number L07944).
  • M036Xi0003-VH-CDR2-aa YICGHGDHIYYADSVKG (SEQ ID NO: 172)
  • AGCTCACCTGCACTCTGAGCAGTGGGCACAGCAGCTACGCCATCGCATGGCATCA GCAGCAGCCAGAGAAGGGCCCTCGGTACTTGATGAAGCTTAACAGTGATGGCAGC
  • M036Xi0003-VL-CDR3-nucleotide CAG ACCTG G G GCACTG GCATTGTG GTG
  • Pf-MSP3 nucleic acid sequence (Genbank L07944) atgaaaagttttataaatattactctttcattattttttgttacatttatatatttatataaataatgttgctagtaaaga
  • Antibody epitope 1 nucleotide sequence (MSP3 183 354 )
  • Antibody epitope 1 amino acid sequence (MSP3 183_354 )
  • Antibody epitope 2 nucleotide sequence

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Veterinary Medicine (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biochemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Immunology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)

Abstract

L'invention concerne des molécules de liaison, en particulier des anticorps ou des fragments de liaison à un antigène de ceux-ci, qui se lient spécifiquement à la protéine 3 de surface mérozoïte (MSP3) de Plasmodium spec, en particulier Plasmodium falciparum. Ces molécules sont capables d'inhiber la croissance de ou l'infection par Plasmodium spec, en particulier Plasmodium falciparum, sans activer des cellules du système immunitaire de l'hôte in vitro. Ainsi, les molécules de liaison, en particulier les anticorps ou des fragments de liaison à un antigène de ceux-ci tels que fournis ici, sont capables d'inhiber la croissance de ou l'infection par Plasmodium spec, en particulier Plasmodium falciparum, en l'absence de cellules du système immunitaire d'un hôte in vitro. L'invention concerne également des polypeptides comprenant les chaînes lourdes et/ou légères, des CDR isolés ou les régions variables de ces molécules de liaison, anticorps ou fragments de liaison à un antigène de ces anticorps. L'invention concerne également l'utilisation de ces molécules de liaison, anticorps ou fragments de liaison à un antigène de ces anticorps, et des nucléotides correspondants en médecine humaine et vétérinaire, par exemple dans le traitement et le diagnostic de maladies associées à Plasmodium, en particulier la malaria. En outre, la présente invention concerne également des compositions, comme des compositions pharmaceutiques ou de diagnostic, comprenant les molécules de liaison, anticorps ou fragments de liaison à un antigène de ces anticorps divulgués ici ou des molécules d'acide nucléique codant pour ceux-ci.
PCT/EP2013/075867 2012-12-06 2013-12-06 Anticorps de neutralisation de plasmodium falciparum WO2014087007A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP12195971 2012-12-06
EP12195971.2 2012-12-06

Publications (1)

Publication Number Publication Date
WO2014087007A1 true WO2014087007A1 (fr) 2014-06-12

Family

ID=47278710

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2013/075867 WO2014087007A1 (fr) 2012-12-06 2013-12-06 Anticorps de neutralisation de plasmodium falciparum

Country Status (1)

Country Link
WO (1) WO2014087007A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018218068A1 (fr) * 2017-05-24 2018-11-29 Development Center For Biotechnology Anticorps humanisés anti-globo h et leurs utilisations dans des traitements du cancer
WO2018232349A1 (fr) * 2017-06-15 2018-12-20 Development Center For Biotechnology Conjugués anticorps-médicament contenant des anticorps anti-globo h et leurs utilisations

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003016354A2 (fr) * 2001-08-16 2003-02-27 Institut Pasteur Anticorps de recombinaison anti-plasmodium falciparum
US20030161840A1 (en) * 1992-10-19 2003-08-28 Institut Pasteur Plasmodium falciparum antigens inducing protective antibodies

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030161840A1 (en) * 1992-10-19 2003-08-28 Institut Pasteur Plasmodium falciparum antigens inducing protective antibodies
WO2003016354A2 (fr) * 2001-08-16 2003-02-27 Institut Pasteur Anticorps de recombinaison anti-plasmodium falciparum

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
C. OEUVRAY ET AL.: "Merozoite surface protein-3: A malaria protein inducing antibodies that promote Plasmodium falciparum killing by cooperation with blood monocytes.", BLOOD, vol. 84, no. 5, 1 September 1994 (1994-09-01), USA, pages 1594 - 1602, XP009008820 *
H. BOUHAROUN-TAYOUN ET AL.: "Plasmodium falciparum: production of human antibodies specific for the MSP-3 protein in the Hu-SPL-SCID mouse.", EXPERIMENTAL PARASITOLOGY, vol. 108, no. 1-2, 1 September 2004 (2004-09-01), USA, pages 47 - 52, XP004607417 *
S. SINGH ET AL.: "Identification of a conserved region of Plasmodium falciparum MSP3 targeted by biologically active antibodies to improve vaccine design.", JOURNAL OF INFECTIOUS DISEASES, vol. 190, no. 5, 1 September 2004 (2004-09-01), USA, pages 1010 - 1018, XP002538114 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018218068A1 (fr) * 2017-05-24 2018-11-29 Development Center For Biotechnology Anticorps humanisés anti-globo h et leurs utilisations dans des traitements du cancer
WO2018232349A1 (fr) * 2017-06-15 2018-12-20 Development Center For Biotechnology Conjugués anticorps-médicament contenant des anticorps anti-globo h et leurs utilisations

Similar Documents

Publication Publication Date Title
JP7317272B2 (ja) Tigit抗体、その抗原結合断片及びその医療用途 本願は、2019年9月29日に出願された出願番号cn201710908565.3に基づいたものであり、その優先権を主張する。その開示は、その全体が参照により本明細書に組み込まれる。
JP7295916B2 (ja) Psmaおよびcd3に対する二重特異性を有するt細胞エンゲージ抗体コンストラクト
US11692031B2 (en) Antibody constructs for CLDN18.2 and CD3
RU2725811C1 (ru) Антитела против 4-1bb человека и их применение
AU2022203450A1 (en) Bispecific Antibody Constructs For CDH3 And CD3
US11760794B2 (en) Neutralizing antibodies to plasmodium falciparum circumsporozoite protein and their use
EP3712170A1 (fr) Anticorps cd96, fragment de liaison à l'antigène de celui-ci et utilisation pharmaceutique associée
TW201702264A (zh) 結合cd38之抗體治療劑
JP2011032292A (ja) インターフェロンα受容体(IFNAR−1)に対するヒト化抗体
AU2018214223A1 (en) Low pH pharmaceutical composition comprising T cell engaging antibody constructs
WO2018153366A1 (fr) Anticorps anti-tim-3, fragment de liaison à l'antigène de celui-ci, et utilisations médicales de celui-ci
EP3962523A2 (fr) Anticorps neutralisants dirigés contre la protéine circumsporozoïte de plasmodium falciparum et leur utilisation
US20210277139A1 (en) Ifn-gamma-inducible regulatory t cell convertible anti-cancer (irtca) antibody and uses thereof
CA3094981A1 (fr) Anticorps anti-pfrh5 et leurs fragments de liaison a l'antigene
CN116472286A (zh) 结合tnfr2的抗体及其用途
US11834497B2 (en) Glucose transporter 4 antibodies, methods of making the same, and uses thereof
WO2014087007A1 (fr) Anticorps de neutralisation de plasmodium falciparum
CN110606892B (zh) 一种高亲和力高生物活性的lag-3抗体及其应用
AU2004283891B2 (en) MSP-3-like family of genes
WO2021257665A1 (fr) Anticorps monoclonal humain ciblant un site conservé sur la protéine circumsporozoïte de plasmodium falciparum
JP2018508206A (ja) L型電位開口型チャネルに対する抗体および関連方法
EP3283518B1 (fr) Anticorps anti-plasmodium pour parasite
RU2822550C2 (ru) Антитело против клаудина 18.2 и его применение
WO2024064826A1 (fr) Anticorps neutralisants dirigés contre la protéine plasmodium falciparum circumsporozoïte et leur utilisation
JP2022538438A (ja) 百日咳毒素結合タンパク質

Legal Events

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

Ref document number: 13818684

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13818684

Country of ref document: EP

Kind code of ref document: A1