WO2017163049A1 - Anticorps anti-paludéens qui se lient à la protéine circumsporozoïte - Google Patents

Anticorps anti-paludéens qui se lient à la protéine circumsporozoïte Download PDF

Info

Publication number
WO2017163049A1
WO2017163049A1 PCT/GB2017/050786 GB2017050786W WO2017163049A1 WO 2017163049 A1 WO2017163049 A1 WO 2017163049A1 GB 2017050786 W GB2017050786 W GB 2017050786W WO 2017163049 A1 WO2017163049 A1 WO 2017163049A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
domain
mammal
plasmodium
amino acid
Prior art date
Application number
PCT/GB2017/050786
Other languages
English (en)
Inventor
Qi LIANG
Sean Matthew CARROLL
Daniel Eric EMERLING
Allan Bradley
Original Assignee
Kymab Limited
Atreca, Inc.
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 Kymab Limited, Atreca, Inc. filed Critical Kymab Limited
Publication of WO2017163049A1 publication Critical patent/WO2017163049A1/fr
Priority to US15/977,762 priority Critical patent/US11066464B2/en

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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • A61P31/06Antibacterial agents for tuberculosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • 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
    • C07K2317/30Immunoglobulins specific features characterized by aspects of specificity or valency
    • C07K2317/34Identification of a linear epitope shorter than 20 amino acid residues or of a conformational epitope defined by amino acid residues
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • 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

  • This invention relates to treating or preventing malaria, and to antibodies conferring protection against infection by malarial parasites such as Plasmodium falciparum by insect vector transmission.
  • the invention also relates to treating, preventing or diagnosing
  • Plasmodium infection in a mammal Plasmodium infection in a mammal.
  • the causative agent of malaria is a protozoal parasite, which is transmitted by mosquitos.
  • Plasmodium parasites such as P. falciparum
  • the infective stage is the sporozoite, which is transmitted from the mosquito to the vertebrate host, e.g., human. Preventing infection will involve preventing sporozoite transmission from the insect vector to humans. Decreasing infection by P. falciparum will depend on blocking transmission, or inhibiting the pre-erythrocytic phase of infection.
  • Targeting the asexual phases involved in the erythrocytic cycle (and disease) may only mimic naturally acquired immunity - that is, immunity to disease rather than immunity to infection.
  • Circumsporozoite protein is an antigen that is highly expressed on the P. falciparum sporozoite. CSP is displayed on the surface of the sporozoite and is involved in key parasite functions. Human antibodies against P. falciparum sporozoites have been described. Del Giudice et al synthesised a peptide of about 40 repeats of NANP (Asn-Ala- Asn-Pro) from CSP, (NANP) 4 o- This was recognised by monoclonal antibodies produced against P.
  • 3 individuals with the highest anti NANP antibody titres were challenged by experimental mosquito mediated infection with Plasmodium falciparum sporozoites.
  • 2 of the vaccines showed delayed parasitaemia and one showed no parasitaemia (Herington et al, Nature. 328: 257-259 1987), indicating that antibodies to NANP could be protective to infection.
  • a first generation vaccine (RTS,S) has been developed using portions of CSP, including part of the NANP repeats.
  • CSP-based vaccines have consistently shown 30-50% efficacy in prevention of erythrocytic-stage infection. This level of efficacy is not sufficient for eradication and new pre-erythrocytic treatments will need superior efficacy.
  • anti-malarial products include mefloquinine, doxycycline and
  • Atovaquone/proguanil which are used for chemoprophylaxis by travellers and the military.
  • Artemisinin and its derivatives also have anti-malarial action, and artemisinin combination therapies are a standard treatment worldwide for malaria.
  • sulphadoxine-pyrimethanime SP was initially almost 100% effective in curing malaria when introduced in 1977, but within five years was curing only 10% of cases due to drug resistance.
  • the once-popular chloroquine has lost its effectiveness in almost every part of the world.
  • 95% of African children treated for malaria were given chloroquine, even though the drug only cured half of malaria cases in many countries.
  • the invention provides novel anti-parasitic therapeutic and diagnostic agents, including prophylactic agents, especially antibodies.
  • the antibodies may be employed for diagnosing, treating, or preventing malaria or Plasmodium infection in humans and other mammals, or for reducing the risk of malaria or Plasmodium infection in humans and other mammals. Methods of diagnosis and treatment are described herein, including methods of administration to a mammal.
  • the antibodies may be used to confer protection against infection by malarial parasites such as Plasmodium falciparum by insect vector transmission.
  • Desirable features of the antibodies include inhibiting the pre-erythrocytic stage (e.g., sporozoite) of Plasmodium infection, reducing progression of Plasmodium infection in a mammal, reducing one or more symptoms of malaria, and reducing transmission or risk of transmission of Plasmodium to and/or from a mammal.
  • Inhibition of transmission of malarial Plasmodium parasites between mammals (especially humans) is of particular value in lessening spread of disease, limiting malaria and controlling reservoirs of infectious malarial parasites in populations. This invention offers particular advantages for use in countries and regions where malaria is prevalent, and for travellers, military personnel and health workers visiting or operating in such countries or regions.
  • This invention also relates to the use of antibody sequences as correlates of protection following vaccination to protect from malaria infection, which has utility in vaccine development programs, including pre-clinical and clinical trials, and in determining the level of protection achieved in individuals vaccinated against Plasmodium infection.
  • This invention also relates to the use of such antibodies to diagnose malaria infection, and diagnostic methods and kits are described herein.
  • Pharmaceutical compositions comprising the antibodies are also provided. Exemplary embodiments of the invention are set out in the appended claims.
  • Exemplary antibodies include antibodies 666, 667, 668 and 669, the sequences of which are set out herein. Antibodies having close structural similarity with these antibodies, are also described. These are antibodies 666-1 , 666-2, 666-3, 666-4 (antibody 666 lineage), 667-1 , 667-2, 667-3 (antibody 667 lineage), 668-1 (antibody 668 lineage) 669-1 , 669-2, 669- 3, 669-4, 669-5 and 669-6 (antibody 669 lineage), and antibodies having the sets of CDRs shown in Tables 5 to 7.
  • Antibody 667 and antibody 668 are believed to bind the NANP repeat region of the CSP polypeptide sequence. These antibodies have been characterised both in vitro and in vivo and found to inhibit pathologically relevant sporozoite functions and to protect against sporozoite infection as described in the appended Examples. Antibody 666 and antibody 669 are related in sequence to both antibody 667 and antibody 668, indicating they may share useful functional properties in common and that they may also bind the NANP region of CSP. Targeting an epitope within this region may confer particular advantages for inhibiting sporozoite infection. Antibodies that bind the NANP repeat region, and their use in therapeutic and prophylactic methods described herein, thus represent an aspect of the present invention. The invention also relates to antibodies that neutralise CSP.
  • An antibody according to the invention may be one that competes for binding to CSP of Plasmodium falciparum with an antibody (e.g., human lgG1 , or an scFv) comprising the heavy and light chain complementarity determining regions (CDRs) of antibody 666, antibody 667, antibody 668 or antibody 669.
  • an antibody e.g., human lgG1 , or an scFv
  • CDRs complementarity determining regions
  • An antibody according to the present invention may comprise one or more CDRs of any of antibody 666, 667, 668 or 669 (e.g., all 6 CDRs of any such antibody, or a set of HCDRs and/or LCDRs) or variants thereof as described herein.
  • An antibody according to the present invention may comprise one or more CDRs of an antibody of the "antibody 666 lineage", an antibody of the "antibody 667 lineage", an antibody of the “antibody 668 lineage” or an antibody of the "antibody 669 lineage” (e.g., all 6 CDRs of any such antibody, or a set of HCDRs and/or LCDRs) or variants thereof as described herein.
  • the "antibody 666 lineage” comprises antibody 666, antibody 666-1 , antibody 666-2, antibody 666-3 and antibody 666-4.
  • the "antibody 667 lineage” comprises antibody 667, antibody 667-1 , antibody 667-2 and antibody 667-3.
  • the "antibody 668 lineage" comprises antibody 668 and antibody 668-1 .
  • the "antibody 669 lineage” comprises antibody 669, antibody 669-1 , antibody 669-2, antibody 669-3, antibody 669-4, antibody 669-5 and antibody 669-6.
  • An antibody according to the present invention may comprise one or more CDRs as shown in Tables 5 to 7.
  • the antibody may comprise a set of 6 CDRs shown in Table 5 or Table 6, it may comprise a VH domain having a set of HCDRs as indicated in any of Tables 5 to 7, and/or it may comprise a VL domain having a set of LCDRs as indicated in Table 5 or Table 6.
  • the antibody may comprise an antibody VH domain comprising CDRs HCDR1 , HCDR2 and HCDR3 and an antibody VL domain comprising CDRs LCDR1 , LCDR2 and LCDR3, wherein the HCDR3 is an HCDR3 of an antibody selected from antibody 666, 667, 668 or 669 or comprises that HCDR3 with 1 , 2, 3, 4 or 5 amino acid alterations.
  • the HCDR2 may be the HCDR2 of the selected antibody or it may comprise that HCDR2 with 1 , 2, 3, 4 or 5 amino acid alterations.
  • the HCDR1 may be the HCDR1 of the selected antibody or it may comprise that HCDR1 with 1 , 2, 3, 4 or 5 amino acid alterations.
  • the antibody may comprise an antibody VL domain comprising CDRs HCDR1 , HCDR2 and HCDR3 and an antibody VL domain comprising CDRs LCDR1 , LCDR2 and LCDR3, wherein the LCDR3 is an LCDR3 of an antibody selected from antibody 666, 667,
  • the LCDR2 may be the LCDR2 of the selected antibody or it may comprise that LCDR2 with 1 , 2, 3, 4 or 5 amino acid alterations.
  • the LCDR1 may be the LCDR1 of the selected antibody or it may comprise that LCDR1 with 1 , 2, 3, 4 or 5 amino acid alterations.
  • An antibody may comprise:
  • an antibody VH domain comprising complementarity determining regions HCDR1 , HCDR2 and HCDR3, and
  • an antibody VL domain comprising complementarity determining regions LCDR1 , LCDR2 and LCDR3,
  • heavy chain complementarity determining regions are those of antibody
  • the light chain complementarity determining regions are those of antibody 666, antibody 667, antibody 668 or antibody 669, or comprise the antibody 666, 667, 668 or 669 light chain complementarity determining regions with 1 , 2, 3, 4 or 5 amino acid alterations.
  • the antibody may comprise an antibody VH domain comprising CDRs HCDR1 , HCDR2 and HCDR3 and an antibody VL domain comprising CDRs LCDR1 , LCDR2 and LCDR3, wherein
  • the HCDR1 , HCDR2 and HCDR3 are each independently selected from an antibody of the antibody 666 lineage, and/or wherein
  • the LCDR1 , LCDR2 and LCDR3 are each independently selected from an antibody of the antibody 666 lineage.
  • the HCDR1 may be the HCDR1 of any of antibodies 666, 666-1 , 666-2, 666-3 or 666-4
  • the HCDR2 may be the HCDR2 of any of antibodies 666, 666-1 , 666-2, 666-3 or 666-4
  • the HCDR3 may be the HCDR3 of any of antibodies 666, 666-1 , 666-2, 666-3 or 666- 4.
  • the LCDR1 may be the LCDR1 of any of antibodies 666, 666-1 , 666-2, 666-3 or 666-4
  • the LCDR2 may be the LCDR2 of any of antibodies 666, 666-1 , 666-2, 666-3 or 666- 4
  • the LCDR3 may be the LCDR3 of any of antibodies 666, 666-1 , 666-2, 666-3 or 666- 4.
  • the antibody comprises a set of HCDRs (HCDR1 , HCDR2 and HCDR3) from antibody 666-1 , antibody 666-2, antibody 666-3 or antibody 666-4.
  • the antibody optionally comprises a set of LCDRs (LCDR1 , LCDR2 and LCDR3) from the same antibody.
  • An antibody may comprise an antibody VH domain comprising CDRs HCDR1 , HCDR2 and HCDR3 and an antibody VL domain comprising CDRs LCDR1 , LCDR2 and LCDR3, wherein
  • the HCDR1 , HCDR2 and HCDR3 are each independently selected from an antibody of the antibody 667 lineage, and/or wherein
  • the LCDR1 , LCDR2 and LCDR3 are each independently selected from an antibody of the antibody 667 lineage.
  • the antibody comprises a set of HCDRs from antibody 667-1 , antibody 667-2 or antibody 667-3.
  • the antibody optionally comprises a set of LCDRs from the same antibody.
  • An antibody may comprise an antibody VH domain comprising CDRs HCDR1 , HCDR2 and HCDR3 and an antibody VL domain comprising CDRs LCDR1 , LCDR2 and LCDR3, wherein
  • the HCDR1 , HCDR2 and HCDR3 are each independently selected from an antibody of the antibody 668 lineage, and/or wherein
  • the LCDR1 , LCDR2 and LCDR3 are each independently selected from an antibody of the antibody 668 lineage.
  • the antibody comprises a set of HCDRs from antibody 668-1 .
  • the antibody optionally also comprises a set of LCDRs from antibody 668-1 .
  • An antibody may comprise an antibody VH domain comprising CDRs HCDR1 , HCDR2 and HCDR3 and an antibody VL domain comprising CDRs LCDR1 , LCDR2 and LCDR3, wherein
  • the HCDR1 , HCDR2 and HCDR3 are each independently selected from an antibody of the antibody 669 lineage, and/or wherein
  • the LCDR1 , LCDR2 and LCDR3 are each independently selected from an antibody of the antibody 669 lineage.
  • the antibody comprises a set of HCDRs from antibody 669-1 , 669-2, 669-
  • the antibody optionally comprises a set of LCDRs from the same antibody.
  • Example antibody CDR sequences for antibodies of the invention are as follows: HCDR1 G(Y/F)TFT(N/D)YAMH SEQ ID NO: 134
  • CSYAGSSAWV SEQ ID NO: 20
  • SSYAGSSTWV SEQ ID NO: 30
  • Residues shown in brackets are alternatives at a given position.
  • position 2 of a sequence Z(X/Y)Z has either residue X or residue Y, so that Z(X/Y)Z designates the sequences ZXZ and ZYZ.
  • the antibody has a HCDR3 sequence
  • the antibody has an LCDR3 sequence CSYAGSSAWV or SSYAGSSTWV, an LCDR1 sequence TGTS(N/S)DVG(I/S)YN (H/YV)S, and an LCDR2 sequence
  • Antibodies of the invention may comprise VH and/or VL domain framework regions corresponding to human germline gene segment sequences.
  • it may comprise one or more framework regions of antibody 666, antibody 667, antibody 668 or antibody 669.
  • the framework region or framework regions may be a FR1 , FR2, FR3 and/or FR4.
  • An antibody of the invention may comprise an antibody VH domain which
  • (i) is derived from recombination of a human heavy chain V gene segment, a human heavy chain D gene segment and a human heavy chain J gene segment, wherein
  • V segment is IGHV1 -3;
  • the D gene segment is IGHD3-10, IGHD3-9, IGHD4-1 1 or IGHD2-2; and/or the J gene segment is IGHJ6, or
  • (ii) comprises framework regions FR1 , FR2, FR3 and FR4, wherein
  • FR1 aligns with human germline V gene segment IGHV1 -3 with up to 5 amino acid alterations, e.g., 1 , 2, 3, 4 or 5 amino acid alterations,
  • FR2 aligns with human germline V gene segment IGHV1 -3 with up to 5 amino acid alterations, e.g., 1 , 2, 3, 4 or 5 amino acid alterations,
  • FR3 aligns with human germline V gene segment IGHV1 -3 with up to 5 amino acid alterations, e.g., 1 , 2, 3, 4 or 5 amino acid alterations, and/or
  • FR4 aligns with human germline J gene segment IGHJ6 with up to 5 amino acid alterations, e.g., 1 , 2, 3, 4 or 5 amino acid alterations.
  • a FR1 , FR2, FR3 or FR4 segment may align perfectly with the germline gene segment, i.e., with no amino acid alterations.
  • the antibody may comprise a VH domain derived from
  • An antibody may comprise VH domain framework regions FR1 , FR2, FR3 and FR4, wherein FR1 , FR2 and FR3 each align with human germline V gene segment IGHV1 -3 with 1 , 2, 3, 4 or 5 amino acid alterations, and a FR4 that aligns with human germline J gene segment IGHJ6 with 1 , 2, 3, 4 or 5 amino acid alterations.
  • An antibody of the invention may comprise an antibody VL domain which
  • (i) is derived from recombination of a human light chain V gene segment and a human light chain J gene segment, wherein
  • V segment is IGLV2-23, and/or
  • the J gene segment is IGLJ3; or
  • (ii) comprises framework regions FR1 , FR2, FR3 and FR4, wherein
  • FR1 aligns with human germline V gene segment IGLV2-23 with up to 5 amino acid alterations, e.g., 1 , 2, 3, 4 or 5 amino acid alterations,
  • FR2 aligns with human germline V gene segment IGLV2-23 with up to 5 amino acid alterations, e.g., 1 , 2, 3, 4 or 5 amino acid alterations,
  • FR3 aligns with human germline V gene segment IGLV2-23 with up to 5 amino acid alterations, e.g., 1 , 2, 3, 4 or 5 amino acid alterations, and/or
  • FR4 aligns with human germline J gene segment IGLJ3 with up to 5 amino acid alterations, e.g., 1 , 2, 3, 4 or 5 amino acid alterations.
  • a FR1 , FR2, FR3 or FR4 segment may align perfectly with the germline gene segment, i.e., with no amino acid alterations.
  • An antibody according to the invention may comprise an antibody VH domain which is the VH domain of antibody 666, antibody 667, antibody 668 or antibody 669, or which has an amino acid sequence at least 90 % identical to the antibody VH domain sequence of antibody 666, antibody 667, antibody 668 or antibody 669.
  • the VH domain is the VH domain of an antibody in the antibody 666 lineage, an antibody in the antibody 667 lineage, an antibody in the antibody 668 lineage or an antibody in the antibody 669 lineage.
  • the antibody may comprise an antibody VL domain which is the VL domain of antibody 666, antibody 667, antibody 668 or antibody 669, or which has an amino acid sequence at least 90 % identical to the antibody VL domain sequence of antibody 666, antibody 667, antibody 668 or antibody 669.
  • the VL domain is the VL domain of an antibody in the antibody 666 lineage, an antibody in the antibody 667 lineage, an antibody in the antibody 668 lineage or an antibody in the antibody 669 lineage.
  • An antibody VH domain having the HCDRs of antibody 666, antibody 667, antibody 668 or antibody 669, or having a variant of those CDRs may be paired with an antibody VL domain having the LCDRs of the same antibody, or having a variant of those CDRs.
  • the VH domain of any of antibody 666, antibody 667, antibody 668 or antibody 669, or a variant of that VH domain may be paired with a VL domain of the same antibody, or a VL domain variant of the same antibody.
  • the antibody may comprise the antibody 667 VH domain and the antibody 667 VL domain.
  • the antibody may comprise the antibody 668 VH domain and the antibody 668 VL domain.
  • Antibodies may include constant regions, optionally human heavy and/or light chain constant regions.
  • An exemplary isotype is IgG, e.g., human lgG1 .
  • nucleic acid molecules encoding sequences of the antibodies described herein, host cells containing such nucleic acids, and methods of producing the antibodies by culturing the host cells and expressing and optionally isolating or purifying the antibodies. The expressed antibody is thereby obtained.
  • VH and VL domains of antibodies described herein may similarly be produced and are aspects of the present invention.
  • Figure 1 illustrates the life cycle of malarial Plasmodium parasites such as P. falciparum.
  • Figure 2 is an immunisation schedule for Kymice to produce anti-malarial antibodies.
  • Figure 3 is a schematic diagram of antigen binding cross-blocking between antibodies.
  • Figure 4 is a table of data relating to selected anti-malarial antibodies.
  • Figure 5 illustrates a model to test protection of mice from mosquito transmission of
  • Figure 6 shows the synthetic nucleotide sequence for Pf CSP (SEQ ID NO: 41 , encoding SEQ ID NO: 42).
  • Figure 7 shows the amino acid sequence of Pf CSP (SEQ ID NO: 42) used for reverse translation of a synthetic nucleotide sequence.
  • the illustrated sequence includes a hexa- histidine C-terminal tag.
  • the sequence without the his tag is Plasmodium falciparum CSP SEQ ID NO: 43.
  • Figure 8 shows the results of epitope mapping experiments for antibodies 667 and 668 and reference data from other test antibodies.
  • A Dose-dependent ELISA binding towards recombinant CSP from P. falciparum.
  • B Dose-dependent ELISA binding towards EP070034 (NANP peptide).
  • C Dose-dependent ELISA binding towards Pf16 (C-terminal peptide of CSP).
  • Figure 9 shows results obtained for Antibody 667 and Antibody 668 in a murine malaria challenge model as described in Examples 4a and 4b. Protection data is displayed in terms of liver-stage parasite burden, determined by the number of P. berghei 18s RNA copies detected in mouse liver homogenates.
  • Figure 10 shows results obtained in the in vivo mouse protection assay described in
  • Figure 1 1 shows further results from the work described in Examples 4a and 4b, in terms of parasitemia following sporozoite challenge of mice receiving passive transfer of anti-CSP mAbs. 17 days after blood feeding on Pb-Pf FULL CSP infected mice, the proportion of infected mosquitoes was -85% (26 out of 30 infected salivary glands). Based on this calculation, it was determined that 6 mosquitoes were needed to challenge mice with 5 infected mosquito bites. Upon passive transfer of monoclonal antibodies, mice were anesthetised with 2% Avertin and mosquitoes allowed to feed on mice for -10 minutes. After feeding, we determined the number of mosquitoes positive for a blood meal. Upper panel data are for mAb667; lower panel data are for mAb668.
  • Figure 12 shows data from the gliding motility assay described in Example 5.
  • P. falciparum sporozoites were pre-incubated with the indicated antibodies for 30 min and then added to wells in the presence of bovine serum albumin and incubated at 37 degrees C for 1 hr. They were then fixed and trails were stained with anti-CSP antibody and counted using a fluorescence microscope. 100 sporozoites were counted and shown is the number associated with the indicated number of circular trails, indicative of their motility.
  • Figure 13 shows ILSDA data from the work described in Example 6. 25k Pf sporozoites were mixed with the indicated mAbs at 20 ⁇ g/ml and inoculated into CPHH. CPHH were harvested after 96 hours incubation.
  • Pf 18s rRNA copy numbers were measured by qRT- PCR. Y axis is copies of Pf 18s per well compared to standards. Error bars represent standard deviation. 3 separate wells were tested for each mAb.
  • the tested anti-CSP antibodies are Ab640, Ab643, Ab646, Ab649, Ab662, Ab667 (second from right), Ab668 (far right) and the reference anti-CSP antibody 2A10 (second from left).
  • Figure 14 shows data on inhibition of hepatocyte traversal as described in Example 7.
  • Graph represents traversal inhibition conferred by anti-CSP Antibodies 640, 643, 646, 649, 667 and 668 and antibodies to another antigen (Antibodies 605, 612, 628, 629, 632) tested in this assay at 10 ⁇ g/mL (right hand bar) and 100 ⁇ g/mL (left hand bar). The means of two independent experiments are shown. Only anti-CSP mAbs 667 and 668 appear to mediate traversal.
  • Figure 15 shows data on inhibition of sporozoite invasion as described in Example 7.
  • Graph represents invasion inhibition conferred by anti-CSP Antibodies 640, 643, 646, 649, 667 and 668 and antibodies to another antigen (Antibodies 605, 612, 628, 629, 632) tested in this assay at 10 ⁇ g/mL. The means of two independent experiments are shown.
  • Figure 16 shows in vivo protection mediated by passive transfer of antibodies 667 and 668 in challenge of FRG humanized liver (Hu-Hep) mice with Pf-infected mosquito bites as described in Example 8.
  • Ab667 and Ab668 exhibit robust and dose dependent protection from Pf sporozoite liver infection.
  • Figure 17 shows amino acid sequences of the VH and VL domains of Antibody 667 and Antibody 668 with CDR sequences boxed.
  • the HCDR1 SEQ ID NO: 13
  • HCDR2 SEQ ID NO: 14
  • HCDR3 SEQ ID NO: 15
  • the LCDR1 SEQ ID NO: 18
  • LCDR2 SEQ ID NO: 19
  • LCDR3 SEQ ID NO: 20
  • the HCDR1 SEQ ID NO: 23
  • HCDR2 SEQ ID NO: 24
  • HCDR3 SEQ ID NO: 25
  • the LCDR1 SEQ ID NO: 28
  • LCDR2 SEQ ID NO: 29
  • LCDR3 SEQ ID NO: 30
  • Figure 18 is an antibody lineage tree for Ab666, showing the inferred relationship between Ab666 and Ab666-1 , Ab666-2, Ab666-3 and Ab666-4.
  • Figure 19 is an antibody lineage tree for Ab667, showing the inferred relationship between Ab667 and Ab667-1 , Ab667-2 and Ab667-3.
  • Figure 20 is an antibody lineage tree for Ab668, showing the inferred relationship between Ab668 and Ab668-1 .
  • Figure 21 is an antibody lineage tree for Ab669, showing the inferred relationship between Ab669 and Ab669-1 , Ab669-2, Ab669-3, Ab669-4, Ab669-5 and Ab669-6.
  • sequence listing includes VH and VL domain sequences for antibodies that were identified as being in the same lineages as antibody 666, 667, 668 and 669 and having different CDR sequences, namely antibodies 666-3, 666-4 (antibody 666 lineage), 667-1 , 667-2, 667-3 (antibody 667 lineage), 668-1 (antibody 668 lineage) 669-1 , 669-2, 669-3, 669-4, 669-5 and 669-6
  • antibody 669 lineage (antibody 669 lineage). Further sets of antibody CDRs are shown in Table 5, Table 6 and Table 7.
  • CDR sequences within antibody variable domains may for example be determined according to the Kabat system, Chothia system, IMGT system, or others.
  • CDR sequence tables we therefore show CDRs for the antibodies, including their CDRs as determined by the Kabat and Chothia systems respectively.
  • Antibodies according to the present invention are intended for medical use, particularly as anti-malarials, and for diagnostic use.
  • Desirable properties for antibodies according to examples of the present invention include one or more of:
  • an antibody of the present invention may have any one or more, for example all, of these properties.
  • An antibody according to the invention may be a neutralising antibody, neutralising one or more functions of CSP.
  • Anti-CSP antibodies according to the invention may inhibit sporozoite functions in assays in vitro, including gliding motility (Example 5), liver stage development (Example 6) and hepatocyte traversal and/or invasion (Example 7).
  • An antibody of the invention may inhibit the pre-erythrocytic stage of a Plasmodium parasite infection, e.g., P. falciparum infection, thereby reducing infection by mosquito transmitted malaria.
  • An antibody may provide protection from challenge in a malaria challenge mouse model (e.g., in which mice are infected with recombinant Plasmodium berghei expressing Plasmodium falciparum CPS) and the equivalent human challenge model.
  • a malaria challenge mouse model e.g., in which mice are infected with recombinant Plasmodium berghei expressing Plasmodium falciparum CPS
  • An illustrative protocol for such a mouse model is set out in Example 4. Protection from challenge may be quantified as % reduction in liver-stage parasite load determined by the number of P. berghei 18s RNA copies detected in mouse liver homogenates in such a model where the murine P.
  • Protection from challenge may be at least 50 %, at least 60 %, at least 70 %, at least 80 %, at least 90 %, at least 95 %, at least 98 % or at least 99 %.
  • Full protection from challenge may be obtained, i.e., liver-stage parasite load may be eliminated or entirely prevented through treatment.
  • Examples 4 and 8 herein describe in detail two different in vivo mouse challenge models that were used to assess the ability of passively-transferred anti-CSP mAbs to protect mice from sporozoite challenge: a transgenic (Pb-Pf CSP) sporozoite challenge and a human liver (Hu-Hep) mouse challenge model.
  • a transgenic (Pb-Pf CSP) sporozoite challenge and a human liver (Hu-Hep) mouse challenge model.
  • Protocols for challenge models are also described in:
  • Circumsporozoite Protein as a Model for Vaccine Down-Selection in Mice Using Sterile Protection as an Endpoint. Porter et al., Clin Vaccine Immunol. 20(6):803-810 2013.
  • Antibodies according to the present invention bind CSP.
  • exemplary antibodies were raised in KymiceTM immunised with CSP from P. falciparum.
  • the nucleotide and amino acid sequences of CSP are shown in Figures 6 and 7.
  • An antibody may bind the four amino acid repeat region (NANP) of CSP. It may bind one or more residues encoded by nucleotides 368 to 862 as shown in Figure 6.
  • the NANP region is the region of the CSP polypeptide that is characterised by multiple tandem repeats of the amino acid sequence NANP (SEQ ID NO: 47) i.e., Asn - Ala - Asn - Pro).
  • An antibody may bind a polypeptide or peptide comprising or consisting of a NANP repeat region (NANP)n where n is 1 to 40, e.g., 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 20, 30 or 40, e.g., (NANP)4.
  • NANP NANP repeat region
  • Any suitable method may be used to determine whether an antibody binds to an antigen such as CSP, a fragment thereof (e.g., the NANP region or a fragment comprising the NANP region), or a polypeptide or peptide comprising a NANP repeat region.
  • a method may comprise surface plasmon resonance (SPR), bio-layer interferometry, or an ELISA to determine specificity of antibodies.
  • An antibody may be said to bind its antigen if the level of binding to antigen is at least 2.5 fold greater, e.g., at least 10 fold greater, than binding to a control antigen. Binding between an antibody and its cognate antigen is often referred to as specific binding.
  • Precise identification of the residues bound by an antibody can usually be obtained using x-ray crystallography. This technique may be used to determine that an antibody of the invention binds one or more residues of the NANP repeat region of CSP.
  • the antibody epitope may comprise or consist of residues within the NANP repeat region.
  • the invention provides antibodies that compete with antibody 666, 667, 668 or 669 for binding to CSP or a fragment thereof.
  • An antibody may compete with a human lgG1 or with an scFv antibody comprising the VH and VL domain of an antibody selected from 666, 667, 668 or 669, or a human lgG1 or with an scFv antibody comprising the heavy and light chain CDRs of antibody 666, 667, 668 or 669.
  • Competition between antibodies may be assayed in vitro, for example using ELISA and/or by tagging a specific reporter molecule to one antibody, which can be detected in the presence of one or more other untagged antibodies, to enable identification of antibodies that bind the same epitope or an
  • full length CSP In assay methods and methods of administration described herein relating to the CSP antigen, it is optional to use full length CSP.
  • These CSP fragments or synthetic peptides may be provided as isolated sequences, contained within longer polypeptide sequences, and/or provided with one or more tags or detectable labels, as appropriate.
  • Kd ability of an antibody to bind its target antigen, and the specificity and affinity of that binding (Kd, Koff and/or Kon) can be determined by any routine method in the art, including SPR.
  • Kd is intended to refer to the equilibrium dissociation constant of a particular antibody-antigen interaction.
  • Affinity of antibody-antigen binding may be determined, e.g., by surface plasmon resonance. Affinity may also be determined by bio-layer interferometry. An example of affinity determination by bio-layer interferometry is provided in Example 3 herein.
  • An antibody may bind to CSP with an affinity (Kd) of 1 mM or less, usually 1 nM or less, e.g., 0.9 nM or less, 0.8 nM or less, 0.7 nM or less, 0.5 nM or less, or 0.4 nM or less.
  • Kd affinity
  • Determining affinity (Kd) by bio-layer interferometry may comprise:
  • Pf CSP dilution series of antigen
  • a baseline measurement may be established using dissociation buffer after loading the biosensors (e.g., for 30 seconds).
  • Parallel references may be set up by using unloaded bare sensor and sensor contacted with the dilution series of the antigen, and the parallel reference used for normalisation in data processing.
  • antigen is immobilised on the biosensors and contacted with a dilution series of the antibody (e.g., starting at 33 nM or 133 nM, 1 :3 diluted down).
  • a dilution series of the antibody e.g., starting at 33 nM or 133 nM, 1 :3 diluted down.
  • suitable concentrations for the dilution series can be determined depending on the strength of the affinity being measured. Any suitable bio-layer interferometry machine may be used.
  • SPR is carried out at 25°C.
  • An alternative is 37 ⁇ .
  • SPR may be performed at physiological pH, such as about pH 7 or at pH 7.6 (e.g., using Hepes buffered saline at pH 7.6 (also referred to as HBS-EP)).
  • SPR may be carried out at a physiological salt level, eg, 150 mM NaCI.
  • SPR may be carried out at a detergent level of no greater than 0.05 % by volume, e.g., in the presence of P20 (polysorbate 20; eg, Tween-20®) at 0.05 % and EDTA at 3 mM.
  • the SPR is carried out at 25 ⁇ or 37°C in a buffer at pH 7.6, 150 mM NaCI, 0.05 % detergent (e.g., P20) and 3 mM EDTA.
  • the buffer can contain 10 mM Hepes.
  • the SPR is carried out at 25 °C or 37 ⁇ in HBS-EP.
  • HBS-EP is available from Teknova Inc (California; catalogue number H8022).
  • the affinity of the antibody is determined using SPR by
  • SPR can be carried out using any standard SPR apparatus, such as by Biacore® using the ProteOn XPR36® (Bio-Rad®).
  • Regeneration of the capture surface can be carried out with 10 mM glycine at pH 1 .7. This removes the captured antibody and allows the surface to be used for another interaction.
  • the binding data can be fitted to 1 :1 model inherent using standard techniques, e.g., using a model inherent to the ProteOn XPR36® analysis software.
  • Antibodies according to the present invention are immunoglobulins or molecules comprising immunoglobulin domains, whether natural or partly or wholly synthetically produced.
  • Antibodies may be IgG, IgM, IgA, IgD or IgE molecules or antigen-specific antibody fragments thereof (including, but not limited to, a Fab, F(ab')2, Fv, disulphide linked Fv, scFv, single domain antibody, closed conformation multispecific antibody, disulphide- linked scfv, diabody), whether derived from any species that naturally produces an antibody, or created by recombinant DNA technology; whether isolated from serum, B-cells, hybridomas, transfectomas, yeast or bacteria.
  • Antibodies can be humanised using routine technology.
  • antibody covers any polypeptide or protein comprising an antibody antigen-binding site.
  • An antigen-binding site is the part of an antibody that binds to and is complementary to the epitope of its target antigen (NANP, eg, comprised by CSP).
  • NANP target antigen
  • epitope refers to a region of an antigen that is bound by an antibody.
  • Epitopes may be defined as structural or functional. Functional epitopes are generally a subset of the structural epitopes and have those residues that directly contribute to the affinity of the interaction. Epitopes may also be conformational, that is, composed of non- linear amino acids.
  • epitopes may include determinants that are chemically active surface groupings of molecules such as amino acids, sugar side chains, phosphoryl groups, or sulfonyl groups, and, in certain embodiments, may have specific three-dimensional structural characteristics, and/or specific charge characteristics.
  • the antigen binding site is a polypeptide or domain that comprises one or more CDRs of an antibody and is capable of binding the antigen.
  • the polypeptide comprises a CDR3 (e.g., HCDR3).
  • the polypeptide comprises CDRs 1 and 2 (e.g., HCDR1 and 2) or CDRs 1 -3 of a variable domain of an antibody (e.g., HCDRsl -3).
  • An antibody antigen-binding site may be provided by one or more antibody variable domains.
  • the antibody binding site is provided by a single variable domain, e.g., a heavy chain variable domain (VH domain) or a light chain variable domain (VL domain).
  • the binding site comprises a VH/VL pair or two or more of such pairs.
  • an antibody antigen-binding site may comprise a VH and a VL.
  • the antibody may be a whole immunoglobulin, including constant regions, or may be an antibody fragment.
  • An antibody fragment is a portion of an intact antibody, for example comprising the antigen binding and/or variable region of the intact antibody. Examples of antibody fragments include:
  • Fab fragment a monovalent fragment consisting of the VL, VH, CL and CH1 domains
  • CDR complementarity determining region
  • Single-chain antibodies e.g., scFv
  • Multispecific antibodies may be formed from antibody fragments.
  • An antibody of the invention may employ any such format, as appropriate.
  • An antibody normally comprises an antibody VH and/or VL domain.
  • Isolated VH and VL domains of antibodies are also part of the invention.
  • the antibody variable domains are the portions of the light and heavy chains of antibodies that include amino acid sequences of complementarity determining regions (CDRs; ie., CDR1 , CDR2, and CDR3), and framework regions (FRs).
  • CDRs complementarity determining regions
  • FRs framework regions
  • a VH domain comprises a set of HCDRs
  • a VL domain comprises a set of LCDRs.
  • VH refers to the variable domain of the heavy chain.
  • VL refers to the variable domain of the light chain.
  • Each VH and VL is typically composed of three CDRs and four FRs, arranged from amino- terminus to carboxy-terminus in the following order: FR1 , CDR1 , FR2, CDR2, FR3, CDR3, FR4.
  • the amino acid positions assigned to CDRs and FRs may be defined according to Kabat (Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md., 1987 and 1991 )) or according to IMGT nomenclature.
  • An antibody may comprise an antibody VH domain comprising a VH CDR1 , CDR2 and CDR3 and a framework.
  • VL domain comprising a VL CDR1 , CDR2 and CDR3 and a framework.
  • antibody VH and VL domains and CDRs according to the present invention are as listed in the appended sequence listing that forms part of the present disclosure. All VH and VL sequences, CDR sequences, sets of CDRs and sets of HCDRs and sets of LCDRs disclosed herein represent aspects and embodiments of the invention.
  • a "set of CDRs" comprises CDR1 , CDR2 and CDR3.
  • a set of HCDRs refers to
  • HCDR1 , HCDR2 and HCDR3 and a set of LCDRs refers to LCDR1 , LCDR2 and LCDR3. Unless otherwise stated, a "set of CDRs" includes HCDRs and LCDRs.
  • An antibody the invention may comprise one or more CDRs as described herein, e.g. a CDR3, and optionally also a CDR1 and CDR2 to form a set of CDRs.
  • the CDR or set of CDRs may be a CDR or set of CDRs of any of antibodies 666, 667 668 or 669, or may be a variant thereof as described herein.
  • the invention provides antibodies comprising an HCDR1 , HCDR2 and/or HCDR3 of any of antibodies 666, 667, 668 and 669 and/or an LCDR1 , LCDR2 and/or LCDR3 of any of these antibodies, e.g. a set of CDRs.
  • the antibody may comprise a set of VH CDRs of one of these antibodies.
  • it may also comprise a set of VL CDRs of one of these antibodies, and the VL CDRs may be from the same or a different antibody as the VH CDRs.
  • a VH domain comprising a disclosed set of HCDRs, and/or a VL domain comprising a disclosed set of LCDRs, are also provided by the invention.
  • a VH domain is paired with a VL domain to provide an antibody antigen- binding site, although as discussed further below a VH or VL domain alone may be used to bind antigen.
  • the antibody 667 VH domain may be paired with the antibody 667 VL domain, so that an antibody antigen-binding site is formed comprising both the antibody 667 VH and VL domains.
  • Analogous embodiments are provided for the other VH and VL domains disclosed herein.
  • the antibody 667 VH is paired with a VL domain other than the antibody 667 VL. Light-chain promiscuity is well established in the art. Again, analogous embodiments are provided by the invention for the other VH and VL domains disclosed herein.
  • VH of any of antibodies 666 to 669 may be paired with the VL of any of antibodies 666 to 669.
  • antibody 667 VH may be paired with antibody 668 VL
  • antibody 668 VH may be paired with antibody 667 VL.
  • An antibody VH domain may optionally be paired with a VH domain of another antibody in the same lineage.
  • the VH domain of antibody 666 may be paired with the VL domain of any of antibodies 666-1 , 666-2, 666-3 and 666-4
  • the VH domain of antibody 666-1 may be paired with the VL domain of any of antibodies 666, 666-2, 666-3 and antibody 666-4, and so on.
  • An antibody may comprise one or more CDRs, e.g. a set of CDRs, within an antibody framework.
  • the framework regions may be of human germline gene segment sequences.
  • the antibody may be a human antibody having a VH domain comprising a set of HCDRs in a human germline framework.
  • the antibody also has a VL domain comprising a set of LCDRs, e.g. in a human germline framework.
  • Figure 4 identifies human germline gene segment sequences that align to the variable domains of antibodies 666, 667, 668 and 669, indicating that these antibodies were derived from recombination of the identified gene segments.
  • an antibody “gene segment”, e.g., a VH gene segment, D gene segment, or JH gene segment refers to oligonucleotide having a nucleic acid sequence from which that portion of an antibody is derived, e.g., a VH gene segment is an oligonucleotide comprising a nucleic acid sequence that corresponds to a polypeptide VH domain from FR1 to part of CDR3.
  • Human V, D and J gene segments recombine to generate the VH domain, and human V and J segments recombine to generate the VL domain.
  • the D domain or region refers to the diversity domain or region of an antibody chain.
  • J domain or region refers to the joining domain or region of an antibody chain.
  • Somatic hypermutation may result in an antibody VH or VL domain having framework regions that do not exactly match or align with the corresponding gene segments, but sequence alignment can be used to identify the closest gene segments and thus identify from which particular combination of gene segments a particular VH or VL domain is derived.
  • the antibody amino acid sequence may be aligned with the amino acid sequence encoded by the gene segment, or the antibody nucleotide sequence may be aligned directly with the nucleotide sequence of the gene segment.
  • An antibody of the invention may comprise an antibody VH domain derived from recombination of a human heavy chain V gene segment, a human heavy chain D gene segment and a human heavy chain J gene segment, wherein
  • V segment is IGHV1 -3;
  • the D gene segment is IGHD3-10, IGHD3-9, IGHD4-1 1 or IGHD2-2; and/or the J gene segment is IGHJ6.
  • IGHV1 -3 is a particularly preferred V gene segment in antibodies and VH domains of the present invention.
  • the selection of IGHV1 -3 for recombination to generate antibody VH domains is believed to represent a particularly effective "choice" when the human immune repertoire is challenged with CSP antigen, as shown in the present Examples.
  • Populations of transgenic mice immunised with CSP may generate anti-NANP repeat antibodies utilising the IGHV1 -3 gene segment, these antibodies being effective for neutralising CSP.
  • the antibody of the invention comprises an antibody VH domain derived from recombination of a human heavy chain V gene segment, a human heavy chain D gene segment and a human heavy chain J gene segment, wherein the V segment is IGHV1 -3 and the antibody neutralises NANP (e.g., neutralises CSP comprising (NANP) 4 ).
  • NANP neutralises CSP comprising (NANP) 4
  • neutralisation is with an IC50 of 500M or less.
  • An antibody of the invention may comprise an antibody VL domain derived from recombination of a human light chain V gene segment and a human light chain J gene segment, wherein
  • V segment is IGLV2-23, and/or
  • the J gene segment is IGLJ3.
  • An antibody of the invention may be a human antibody or a chimaeric antibody comprising human variable regions and non-human (e.g., mouse) constant regions.
  • the antibody of the invention for example has human variable regions, and optionally also has human constant regions.
  • antibodies optionally include constant regions or parts thereof, e.g., human antibody constant regions or parts thereof.
  • a VL domain may be attached at its C-terminal end to antibody light chain kappa or lambda constant domains.
  • an antibody VH domain may be attached at its C-terminal end to all or part (e.g. a CH1 domain or Fc region) of an immunoglobulin heavy chain constant region derived from any antibody isotype, e.g. IgG, IgA, IgE and IgM and any of the isotype sub-classes, such as lgG1 or lgG4.
  • Constant regions of antibodies of the invention may alternatively be non-human constant regions. For example, when antibodies are generated in transgenic animals
  • chimaeric antibodies may be produced comprising human variable regions and non-human (host animal) constant regions. Some transgenic animals generate fully human antibodies. Others have been engineered to generate antibodies comprising chimaeric heavy chains and fully human light chains. Where antibodies comprise one or more non-human constant regions, these may be replaced with human constant regions to provide antibodies more suitable for administration to humans as therapeutic compositions, as their immunogenicity is thereby reduced. Generating and modifying antibodies
  • Antibodies may be generated using transgenic mice (eg, the KymouseTM, Velocimouse® , Omnimouse® , Xenomouse®, HuMab Mouse® or MeMo Mouse®), rats (e.g., the Omnirat®), camelids, sharks, rabbits, chickens or other non-human animals immunised with CSP or a fragment thereof or a synthetic peptide comprising NANP tandem repeats (as described elsewhere herein), followed optionally by humanisation of the constant regions and/or variable regions to produce human or humanised antibodies.
  • display technologies can be used, such as yeast, phage or ribosome display, as will be apparent to the skilled person.
  • Standard affinity maturation e.g., using a display technology, can be performed in a further step after isolation of an antibody lead from a transgenic animal, phage display library or other library. Representative examples of suitable technologies are described in
  • a KymouseTM, VELOCIMMUNE® or other mouse or rat can be challenged with the antigen of interest, and lymphatic cells (such as B-cells) are recovered from the mice that express antibodies.
  • lymphatic cells such as B-cells
  • the lymphatic cells may be fused with a myeloma cell line to prepare immortal hybridoma cell lines, and such hybridoma cell lines are screened and selected to identify hybridoma cell lines that produce antibodies specific to the antigen of interest.
  • DNA encoding the variable regions of the heavy chain and light chain may be isolated and linked to desirable isotypic constant regions of the heavy chain and light chain.
  • Such an antibody protein may be produced in a cell, such as a CHO cell.
  • DNA encoding the antigen-specific chimaeric antibodies or the variable domains of the light and heavy chains may be isolated directly from antigen-specific lymphocytes.
  • high affinity chimaeric antibodies are isolated having a human variable region and a mouse constant region.
  • the antibodies are characterised and selected for desirable characteristics, including affinity, selectivity, epitope, etc.
  • the mouse constant regions are optionally replaced with a desired human constant region to generate the fully human antibody of the invention, for example wild-type or modified lgG1 or lgG4 (for example, SEQ ID NO: 751 , 752, 753 in US201 1/0065902 (which is incorporated by reference herein in its entirety). While the constant region selected may vary according to specific use, high affinity antigen-binding and target specificity characteristics reside in the variable region.
  • Variable domain amino acid sequence variants of any of the VH and VL domains or CDRs whose sequences are specifically disclosed herein may be employed in accordance with the present invention, as discussed.
  • An antibody may comprise a set of H and/or L CDRs of any of the disclosed antibodies with one or more amino acid mutations within the disclosed set of H and/or L CDRs.
  • the mutation may be an amino acid substitution, deletion or insertion.
  • there may be one or more amino acid substitutions within the disclosed set of H and/or L CDRs.
  • An antibody may comprise a VH domain that has at least 60, 70, 80, 85, 90, 95, 98 or 99 % amino acid sequence identity with a VH domain of any of the antibodies shown in the appended sequence listing, and/or comprising a VL domain that has at least 60, 70, 80, 85, 90, 95, 98 or 99 % amino acid sequence identity with a VL domain of any of those antibodies.
  • Algorithms that can be used to calculate % identity of two amino acid sequences include e.g. BLAST, FASTA, or the Smith-Waterman algorithm, e.g. employing default parameters.
  • Particular variants may include one or more amino acid sequence alterations (addition, deletion, substitution and/or insertion of an amino acid residue) .
  • Alterations may be made in one or more framework regions and/or one or more CDRs. Variants are optionally provided by CDR mutagenesis.
  • the alterations normally do not result in loss of function, so an antibody comprising a thus-altered amino acid sequence may retain an ability to bind CSP. It may retain the same quantitative binding ability as an antibody in which the alteration is not made, e.g. as measured in an assay described herein.
  • the antibody comprising a thus-altered amino acid sequence may have an improved ability to bind CSP.
  • Alteration may comprise replacing one or more amino acid residue with a non- naturally occurring or non-standard amino acid, modifying one or more amino acid residue into a non-naturally occurring or non-standard form, or inserting one or more non- naturally occurring or non-standard amino acid into the sequence. Examples of numbers and locations of alterations in sequences of the invention are described elsewhere herein.
  • Naturally occurring amino acids include the 20 "standard" L-amino acids identified as G, A, V, L, I, M, P, F, W, S, T, N, Q, Y, C, K, R, H, D, E by their standard single-letter codes.
  • Nonstandard amino acids include any other residue that may be incorporated into a polypeptide backbone or result from modification of an existing amino acid residue.
  • Non-standard amino acids may be naturally occurring or non-naturally occurring.
  • variant refers to a peptide or nucleic acid that differs from a parent polypeptide or nucleic acid by one or more amino acid or nucleic acid deletions, substitutions or additions, yet retains one or more specific functions or biological activities of the parent molecule.
  • Amino acid substitutions include alterations in which an amino acid is replaced with a different naturally-occurring amino acid residue. Such substitutions may be classified as “conservative", in which case an amino acid residue contained in a polypeptide is replaced with another naturally occurring amino acid of similar character either in relation to polarity, side chain functionality or size. Such conservative substitutions are well known in the art.
  • substitutions encompassed by the present invention may also be "non- conservative", in which an amino acid residue which is present in a peptide is substituted with an amino acid having different properties, such as naturally-occurring amino acid from a different group (e.g., substituting a charged or hydrophobic amino; acid with alanine), or alternatively, in which a naturally-occurring amino acid is substituted with a non- conventional amino acid.
  • amino acid substitutions are conservative.
  • polynucleotide or polypeptide refers to a polynucleotide or polypeptide that can vary in primary, secondary, or tertiary structure, as compared to a reference polynucleotide or polypeptide, respectively (e.g., as compared to a wild- type polynucleotide or polypeptide).
  • “Modified variants” can include conservative or non- conservative amino acid changes, as described below.
  • Polynucleotide changes can result in amino acid substitutions, additions, deletions, fusions and truncations in the polypeptide encoded by the reference sequence.
  • Some aspects use include insertion variants, deletion variants or substituted variants with substitutions of amino acids, including insertions and substitutions of amino acids and other molecules) that do not normally occur in the peptide sequence that is the basis of the variant, for example but not limited to insertion of ornithine which do not normally occur in human proteins.
  • conservative substitution refers to substituting an amino acid residue for a different amino acid residue that has similar chemical properties (e.g., acidic, basic, positively or negatively charged, polar or nonpolar, etc.).
  • Conservative amino acid substitutions include replacement of a leucine with an isoleucine or valine, an aspartate with a glutamate, or a threonine with a serine.
  • Conservative substitution tables providing functionally similar amino acids are well known in the art.
  • the following six groups each contain amino acids that are conservative substitutions for one another: 1 ) Alanine (A), Serine (S), Threonine (T); 2) Aspartic acid (D), Glutamic acid (E); 3) Asparagine (N), Glutamine (Q); 4) Arginine (R), Lysine (K); 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V); and 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W). (See also Creighton, Proteins, W. H.
  • substitutions suitable for amino acids on the exterior of a protein or peptide for example, but not limited to, the following substitutions can be used: substitution of Y with F, T with S or K, P with A, E with D or Q, N with D or G, R with K, G with N or A, T with S or K, D with N or E, I with L or V, F with Y, S with T or A, R with K, G with N or A, K with R, A with S, K or P.
  • substitutions encompassed suitable for amino acids on the interior of a protein or peptide for example one can use suitable conservative substitutions for amino acids is on the interior of a protein or peptide (i.e. the amino acids are not exposed to a solvent), for example but not limited to, one can use the following conservative substitutions: where Y is substituted with F, T with A or S, I with L or V, W with Y, M with L, N with D, G with A, T with A or S, D with N, I with L or V, F with Y or L, S with A or T and A with S, G, T or V.
  • non-conservative amino acid substitutions are also encompassed within the term of variants.
  • the invention includes methods of producing antibodies containing VH and/or VL domain variants of the antibody VH and/or VL domains shown in the appended sequence listing.
  • Such antibodies may be produced by a method comprising
  • parent antibody VH domain is the VH domain of any of antibodies 666, 667, 668 and 669 or a VH domain comprising the heavy chain complementarity determining regions of any of those antibodies, (ii) optionally combining the VH domain thus provided with a VL domain, to provide a VH/VL combination, and
  • Desired characteristics include binding to CSP, for example binding to the four amino acid repeat region NANP of the circumsporozoite protein (CSP) of Plasmodium parasites such as Plasmodium falciparum and optionally other malarial Plasmodium species as mentioned herein. Antibodies with comparable or higher affinity for binding CSP may be identified. Other desired characteristics include inhibiting the pre-erythrocytic stage of infection by Plasmodium, e.g, P. falciparum, which may be determined in an assay as described herein.
  • CSP circumsporozoite protein
  • the VL domain may be a VL domain of any of antibodies 666, 667, 668 or 669, or may be a variant provided by way of addition, deletion, substitution or insertion of one or more amino acids in the amino acid sequence of a parent VL domain, wherein the parent VL domain is the VL domain of any of antibodies 666, 667, 668 and 669 or a VL domain comprising the light chain complementarity determining regions of any of those antibodies.
  • Methods of generating variant antibodies may optionally comprise producing copies of the antibody or VH/VL domain combination. Methods may further comprise expressing the resultant antibody. It is possible to produce nucleotide sequences corresponding to a desired antibody VH and/or VL domain, optionally in one or more expression vectors. Suitable methods of expression, including recombinant expression in host cells, are set out in detail herein.
  • nucleic acids and methods of expression Isolated nucleic acid may be provided, encoding antibodies according to the present invention.
  • Nucleic acid may be DNA and/or RNA. Genomic DNA, cDNA, mRNA or other RNA, of synthetic origin, or any combination thereof can encode an antibody.
  • the present invention provides constructs in the form of plasmids, vectors, transcription or expression cassettes which comprise at least one polynucleotide as above.
  • nucleotide sequences are included in the sequence listing.
  • Reference to a nucleotide sequence as set out herein encompasses a DNA molecule with the specified sequence, and encompasses a RNA molecule with the specified sequence in which U is substituted for T, unless context requires otherwise.
  • the present invention also provides a recombinant host cell that comprises one or more nucleic acids encoding the antibody.
  • Methods of producing the encoded antibody may comprise expression from the nucleic acid, e.g., by culturing recombinant host cells containing the nucleic acid.
  • the antibody may thus be obtained, and may be isolated and/or purified using any suitable technique, then used as appropriate.
  • a method of production may comprise formulating the product into a composition including at least one additional component, such as a pharmaceutically acceptable excipient.
  • Suitable host cells include bacteria, mammalian cells, plant cells,
  • filamentous fungi filamentous fungi, yeast and baculovirus systems and transgenic plants and animals.
  • Vectors may contain appropriate regulatory sequences, including promoter sequences, terminator sequences, polyadenylation sequences, enhancer sequences, marker genes and other sequences as appropriate.
  • Nucleic acid encoding an antibody can be introduced into a host cell.
  • Nucleic acid can be introduced to eukaryotic cells by various methods, including calcium phosphate transfection, DEAE-Dextran, electroporation, liposome-mediated transfection and transduction using retrovirus or other virus, e.g. vaccinia or, for insect cells, baculovirus.
  • Introducing nucleic acid in the host cell, in particular a eukaryotic cell may use a viral or a plasmid based system.
  • the plasmid system may be maintained episomally or may be incorporated into the host cell or into an artificial chromosome. Incorporation may be either by random or targeted integration of one or more copies at single or multiple loci.
  • suitable techniques include calcium chloride transformation, electroporation and transfection using bacteriophage. The introduction may be followed by expressing the nucleic acid, e.g., by culturing host cells under conditions for expression of the gene, then optionally isolating or purifying the antibody.
  • Nucleic acid of the invention may be integrated into the genome (e.g. chromosome) of the host cell. Integration may be promoted by inclusion of sequences that promote recombination with the genome, in accordance with standard techniques.
  • the present invention also provides a method that comprises using nucleic acid described herein in an expression system in order to express an antibody.
  • Antibodies may be monoclonal or polyclonal, but are preferably provided as monoclonal antibodies for therapeutic use. They may be provided as part of a mixture of other antibodies, optionally including antibodies of different binding specificity, such as one or more antibodies that bind to different antigens of a Plasmodium parasite, e.g., P.
  • Antibodies according to the invention, and encoding nucleic acid will usually be provided in isolated form.
  • the antibodies, VH and/or VL domains, and nucleic acids may be provided purified from their natural environment or their production environment.
  • Isolated antibodies and isolated nucleic acid will be free or substantially free of material with which they are naturally associated, such as other polypeptides or nucleic acids with which they are found in vivo, or the environment in which they are prepared (e.g. cell culture) when such preparation is by recombinant DNA technology in vitro.
  • an isolated antibody or nucleic acid (1 ) is free of at least some other proteins with which it would normally be found, (2) is essentially free of other proteins from the same source, e.g., from the same species, (3) is expressed by a cell from a different species, (4) has been separated from at least about 50 percent of polynucleotides, lipids, carbohydrates, or other materials with which it is associated in nature, (5) is operably associated (by covalent or noncovalent interaction) with a polypeptide with which it is not associated in nature, or (6) does not occur in nature.
  • Antibodies or nucleic acids may be formulated with diluents or adjuvants and still for practical purposes be isolated - for example they may be mixed with carriers if used to coat microtitre plates for use in immunoassays, and may be mixed with pharmaceutically acceptable carriers or diluents when used in therapy. As described elsewhere herein, other active ingredients may also be included in therapeutic preparations.
  • Antibodies may be glycosylated, either naturally in vivo or by systems of heterologous eukaryotic cells such as CHO cells, or they may be (for example if produced by expression in a prokaryotic cell) unglycosylated.
  • the invention encompasses antibodies having a modified glycosylation pattern.
  • modification to remove undesirable glycosylation sites may be useful, or e.g., removal of a fucose moiety to increase antibody dependent cellular cytotoxicity (ADCC) function (see Shields et al. (2002) JBC 277:26733).
  • ADCC antibody dependent cellular cytotoxicity
  • an isolated product constitutes at least about 5%, at least about 10%, at least about 25%, or at least about 50% of a given sample.
  • An antibody may be substantially free from proteins or polypeptides or other contaminants that are found in its natural or production environment that would interfere with its therapeutic, diagnostic, prophylactic, research or other use.
  • An antibody may have been identified, separated and/or recovered from a
  • the isolated antibody may be free of association with all other components from its production
  • Contaminant components of its production environment such as that resulting from recombinant transfected cells, are materials that would typically interfere with research, diagnostic or therapeutic uses for the antibody, and may include enzymes, hormones, and other proteinaceous or non-proteinaceous solutes.
  • the antibody will be purified: (1 ) to greater than 95% by weight of antibody as determined by, for example, the Lowry method, and in some embodiments, to greater than 99% by weight; (2) to a degree sufficient to obtain at least 15 residues of N-terminal or internal amino acid sequence by use of a spinning cup sequenator, or (3) to homogeneity by SDS-PAGE under non-reducing or reducing conditions using Coomassie blue or silver stain.
  • Isolated antibody includes the antibody in situ within recombinant cells since at least one component of the antibody's natural environment will not be present. Ordinarily, however, an isolated antibody or its encoding nucleic acid will be prepared by at least one purification step.
  • compositions comprising the antibodies described herein.
  • Therapeutic compositions comprising nucleic acid encoding such antibodies are also provided. Encoding nucleic acids are described in more detail elsewhere herein and include DNA and RNA, e.g., mRNA.
  • compositions may contain suitable carriers, excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • suitable carriers excipients, and other agents that are incorporated into formulations to provide improved transfer, delivery, tolerance, and the like.
  • a multitude of appropriate formulations can be found in the formulary known to all pharmaceutical chemists: Remington's Pharmaceutical Sciences, Mack Publishing
  • formulations include, for example, powders, pastes, ointments, jellies, waxes, oils, lipids, lipid (cationic or anionic) containing vesicles (such as
  • compositions may comprise the antibody or nucleic acid in combination with medical injection buffer and/or with adjuvant.
  • Antibodies, or their encoding nucleic acids may be formulated for the desired route of administration to a patient, e.g., in liquid (optionally aqueous solution) for injection.
  • compositions of the invention include, but are not limited to, intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • the composition may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • the pharmaceutical composition can be also delivered in a vesicle, in particular a liposome (see Langer (1990) Science 249:1527-1533 ; Treat et al. (1989) in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez Berestein and Fidler (eds.), Liss, New York, pp. 353-365 ; Lopez-Berestein, ibid., pp. 317-327 ; see generally ibid.).
  • a liposome see Langer (1990) Science 249:1527-1533 ; Treat et al. (1989) in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez Berestein and Fidler (eds.), Liss, New York, pp. 353-365 ; Lopez-Berestein, ibid., pp. 317-327 ; see generally ibid.).
  • the pharmaceutical composition can be delivered in a controlled release system.
  • a pump may be used (see Langer, supra; Sefton (1987) CRC Crit. Ref. Biomed. Eng. 14:201 ).
  • polymeric materials can be used; see, Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974 ).
  • a controlled release system can be placed in proximity of the composition's target, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 1 15-138, 1984 ).
  • the injectable preparations may include dosage forms for intravenous,
  • injectable preparations may be prepared by methods publicly known.
  • the injectable preparations may be prepared, e.g., by dissolving, suspending or emulsifying the antibody or its salt described above in a sterile aqueous medium or an oily medium conventionally used for injections.
  • aqueous medium for injections there are, for example, physiological saline, an isotonic solution containing glucose and other auxiliary agents, etc., which may be used in combination with an appropriate solubilizing agent such as an alcohol (e.g., ethanol), a polyalcohol (e.g., propylene glycol, polyethylene glycol), a nonionic surfactant [e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil)], etc.
  • an alcohol e.g., ethanol
  • a polyalcohol e.g., propylene glycol, polyethylene glycol
  • a nonionic surfactant e.g., polysorbate 80, HCO-50 (polyoxyethylene (50 mol) adduct of hydrogenated castor oil
  • oily medium there are employed, e.g., sesame oil, soybean oil, etc., which may be used in combination with a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc.
  • a solubilizing agent such as benzyl benzoate, benzyl alcohol, etc.
  • the injection thus prepared can be filled in an appropriate ampoule.
  • a pharmaceutical composition of the present invention can be delivered
  • a pen delivery device readily has applications in delivering a pharmaceutical composition of the present invention.
  • a pen delivery device can be reusable or disposable.
  • a reusable pen delivery device generally utilizes a replaceable cartridge that contains a pharmaceutical composition. Once all of the pharmaceutical composition within the cartridge has been administered and the cartridge is empty, the empty cartridge can readily be discarded and replaced with a new cartridge that contains the pharmaceutical composition. The pen delivery device can then be reused. In a disposable pen delivery device, there is no replaceable cartridge.
  • the disposable pen delivery device comes prefilled with the pharmaceutical composition held in a reservoir within the device. Once the reservoir is emptied of the pharmaceutical composition, the entire device is discarded.
  • Numerous reusable pen and autoinjector delivery devices have applications in the subcutaneous delivery of a pharmaceutical composition of the present invention. Examples include, but certainly are not limited to AUTOPENTM (Owen Mumford, Inc., Woodstock, UK),
  • DISETRONICTM pen (Disetronic Medical Systems, Burghdorf, Switzerland), HUMALOG MIX 75/25TM pen, HUMALOGTM pen, HUMALIN 70/30TM pen (Eli Lilly and Co., Indianapolis, Ind.), NOVOPENTM!, II and III (Novo Nordisk, Copenhagen, Denmark), NOVOPEN JUNIORTM (Novo Nordisk, Copenhagen, Denmark), BDTM pen (Becton Dickinson, Franklin Lakes, N.J.), OPTIPENTTM, OPTIPEN PROTM, OPTIPEN STARLETTM, and OPTICLIKTTM (Sanofi- Aventis, Frankfurt, Germany), to name only a few.
  • Examples of disposable pen delivery devices having applications in subcutaneous delivery of a pharmaceutical composition of the present invention include, but certainly are not limited to the SOLOSTARTM pen (Sanofi- Aventis), the FLEXPENTM (Novo Nordisk), and the KWIKPENTM (Eli Lilly).
  • the pharmaceutical compositions for oral or parenteral use described above are prepared into dosage forms in a unit dose suited to fit a dose of the active ingredients.
  • dosage forms in a unit dose include, for example, tablets, pills, capsules, injections (ampoules), suppositories, etc.
  • the amount of the aforesaid antibody contained is generally about 5 to about 500 mg per dosage form in a unit dose; especially in the form of injection, the aforesaid antibody may be contained in about 5 to about 100 mg and in about 10 to about 250 mg for the other dosage forms.
  • the antibody, nucleic acid, or composition comprising it may be contained in a medical container such as a phial, syringe, IV container or an injection device.
  • a medical container such as a phial, syringe, IV container or an injection device.
  • the antibody, nucleic acid or composition is in vitro, and may be in a sterile container.
  • a kit is provided comprising the antibody, packaging and instructions for use in a therapeutic method as described herein.
  • compositions comprising an antibody or nucleic acid of the invention and one or more pharmaceutically acceptable excipients, examples of which are listed above.
  • “Pharmaceutically acceptable” refers to approved or approvable by a regulatory agency of the USA Federal or a state government or listed in the U.S.
  • a pharmaceutically acceptable carrier, excipient, or adjuvant can be administered to a patient, together with an agent, e.g., any antibody or antibody chain described herein, and does not destroy the pharmacological activity thereof and is nontoxic when administered in doses sufficient to deliver a therapeutic amount of the agent.
  • compositions according to the present invention optionally include one or more additional active ingredients.
  • Further therapeutic agents may be anti-malarial agents such as other antibodies that bind antigens of P. falciparum or other malarial Plasmodium species.
  • such compositions contain multiple antibodies (or encoding nucleic acids) in a combined preparation, e.g., a single formulation comprising the anti-CSP antibody and one or more other antibodies to the same or different antigens of malarial Plasmodium species such as P. falciparum.
  • a composition may include an antibody to Pfs25, an antigen of P. falciparum.
  • antibodies to Plasmodium antigens are antibodies to MSP-1 , MSP-2, TRAP, CelTOS, AMA1 , Rh5 or LSA1 , e.g., from P. falciparum.
  • Other antimalarial agents include chemoprophylactic drugs such as mefloquinine, doxycycline, atovaquone, proguanil, quinine, artemether, lumefantrine, clindamycin, primaquine, artemisinin, sulphadoxine-pyrimethamine and chloroquine. Atovaquone and proguanil are usually administered in combination.
  • anti-malarial drug combinations include artemether with lumefantrine, quinine plus doxycycline, and quinine plus clindamycin.
  • Other therapeutic agents that it may be desirable to administer with antibodies or nucleic acids according to the present invention include antibiotics such as amikacin, and analgaesic agents. Any such agent or combination of agents may be administered in combination with, or provided in compositions with antibodies or nucleic acids according to the present invention, whether as a combined or separate preparation.
  • the antibody or nucleic acid according to the present invention may be administered separately and sequentially, or concurrently and optionally as a combined preparation, with another therapeutic agent or agents such as those mentioned.
  • compositions can be administered separately or simultaneously. Separate administration refers to the two compositions being administered at different times, e.g. at least 10, 20, 30, or 10-60 minutes apart, or 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 12 hours apart. One can also administer compositions at 24 hours apart, or even longer apart. Alternatively, two or more compositions can be administered simultaneously, e.g. less than 10 or less than 5 minutes apart. Compositions administered simultaneously can, in some aspects, be administered as a mixture, with or without similar or different time release mechanism for each of the components.
  • Antibodies, and their encoding nucleic acids can be used as anti-malarial agents.
  • Mammals may be humans, including humans at risk of malaria or humans diagnosed with malaria.
  • the human or other mammal to whom the composition is administered may be one who has been, or is suspected of having been, infected with a malarial Plasmodium parasite, and/or who exhibits one or more symptoms of malaria.
  • An antibody molecule of the present invention may be provided for use in a method of: treating or preventing malaria in a mammal;
  • the method comprises administering the antibody or composition to a mammal.
  • the invention also provides a method of:
  • the method comprising administering an antibody of the invention, or a composition comprising the antibody or its encoding nucleic acid, to the mammal
  • Administration is normally in a "therapeutically effective amount", this being an amount that produces the desired effect for which it is administered, sufficient to show benefit to a patient.
  • the exact amount will depend on the purpose of the treatment, and will be ascertainable by one skilled in the art using known techniques (see, for example, Lloyd (1999) The Art, Science and Technology of Pharmaceutical Compounding).
  • Benefit to the patient may be at least amelioration of at least one symptom of malaria, e.g., reduction in liver-stage parasite load, or protection against malaria or infection by malarial Plasmodium species such as P. falciparum.
  • Prescription of treatment e.g. decisions on dosage etc, is within the responsibility of general practitioners and other medical doctors and may depend on the severity of the symptoms and/or progression of a disease being treated.
  • therapeutically effective amount or suitable dose of antibody or nucleic acid can be determined by comparing its in vitro activity and in vivo activity in an animal model. Methods for extrapolation of effective dosages in mice and other test animals to humans are known.
  • one or more doses may be administered.
  • a single dose may be effective to achieve a long-term benefit.
  • the method may comprise administering a single dose of the antibody, its encoding nucleic acid, or the composition.
  • multiple doses may be administered, usually sequentially and separated by a period of days, weeks or months.
  • the terms “treat,” “treatment,” “treating,” or “amelioration” refer to therapeutic treatments, wherein the object is to reverse, alleviate, ameliorate, inhibit, slow down or stop the progression or severity of a condition associated with a disease or disorder.
  • the term “treating” includes reducing or alleviating at least one adverse effect or symptom of a condition, disease or disorder.
  • Treatment is generally “effective” if one or more symptoms or clinical markers are reduced.
  • treatment is “effective” if the progression of a disease is reduced or halted. That is, “treatment” includes not just the improvement of symptoms or markers, but also a cessation of, or at least slowing of, progress or worsening of symptoms compared to what would be expected in the absence of treatment.
  • Beneficial or desired clinical results include, but are not limited to, alleviation of one or more symptom(s), diminishment of extent of disease, stabilised (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, remission (whether partial or total), and/or decreased mortality, whether detectable or undetectable.
  • treatment also includes providing relief from the symptoms or side- effects of the disease (including palliative treatment). For treatment to be effective a complete cure is not contemplated. The method can in certain aspects include cure as well.
  • treatment may be preventative treatment, optionally the prevention or reduction of infection by Plasmodium such as P. falciparum, and/or the prevention of malaria, or reduction of at least one symptom of malaria.
  • a further aspect of the invention relates to diagnostic uses of the antibodies described herein.
  • the antibodies offer advantages for point of care diagnostics in mammals, and are especially envisaged for use in humans, where rapid diagnosis of malaria and/or Plasmodium infection is of significant clinical benefit.
  • An antibody according to the invention may be used to detect Plasmodium, e.g., Plasmodium falciparum.
  • an antibody may comprise or be conjugated to a detectable label, such as a radioisotope or a fluorescent label.
  • An antibody according to the invention may be used for determining the presence or absence of CSP or Plasmodium in a sample.
  • a method of determining the presence or absence of CSP or Plasmodium in a sample may comprise
  • Detection of binding indicates the presence of CSP or Plasmodium in the sample, whereas absence of binding indicates the absence of CSP or Plasmodium in the sample.
  • Methods may further comprise quantifying the level of binding, for example by comparing the level of binding with a control.
  • Control assays may be run in parallel, or the level of binding may be compared with previously obtained control values.
  • Suitable controls include negative control samples in which no CSP or Plasmodium is present, thus establishing a level of background signal, which can be subtracted from the signal obtained from the test assay.
  • Positive controls may also be conducted, by contacting the antibody with a sample known to contain a pre-determined amount of CSP or Plasmodium.
  • Determining the level of binding in the test sample compared with the level of binding in one or more positive controls is one method of quantifying the amount of CSP or Plasmodium in the sample.
  • the sample may be one that has been obtained from a mammal who has been, or is suspected of having been, infected with a malarial Plasmodium parasite, and/or who exhibits one or more symptoms of malaria. It may be a blood sample, e.g., serum or whole blood. Peripheral blood samples are routinely obtainable from mammals, including human patients, in a clinical setting.
  • kits may comprise the antibody, or a composition as described herein, and optionally one or more buffering solutions.
  • the antibody may comprise or be conjugated to a detectable label.
  • Labelled antibodies allow for detection of the antibody, facilitating detection of binding of the antibody to its target.
  • an antibody may be used in combination with a secondary antibody or other labelled agent, the secondary antibody comprising or carrying a detectable label.
  • a kit may comprise a first reagent comprising an antibody according to the present invention, plus a second reagent comprising a detector molecule that binds to the first reagent.
  • the detector molecule may be an antibody that comprises or is conjugated to a detectable label.
  • mammals may be humans or other mammalian subjects such as wild animals, livestock or test (laboratory) animals.
  • the mammal may be one who has been, or is suspected of having been, infected with a malarial Plasmodium parasite, and/or who exhibits one or more symptoms of malaria.
  • Such mammals represent potential subjects for treatment with antibodies and compositions of the invention, and subjects from whom it may be of value to obtain samples for performing the diagnostic methods described herein.
  • a mammal may be a non-human mammal comprising a human immunoglobulin variable region that is capable of generating antibodies comprising human variable regions.
  • the KymouseTM is a transgenic mouse containing a humanised antibody heavy chain locus and a humanised antibody light chain locus. It is described in Lee et al., Nature Biotechnology 32(4): 356-367 2014 and in WO201 1004192.
  • KymiceTM A number of different KymiceTM have been developed, including mice with humanised kappa light chain variable regions, humanised lambda light chain variable regions, and mice in which the heavy chain variable regions and both lambda and kappa light chain variable regions are fully humanised and comprise the full repertoire of human immunoglobulin gene segments, reflecting the full diversity of the human antibody repertoire.
  • mice such as Velocimouse® , Omnimouse® , Xenomouse®, HuMab Mouse® and MeMo Mouse®, and rats such as the Omnirat®.
  • these animals Owing to their ability to mirror aspects of the human antibody immune response, these animals - especially the KymouseTM - are of particular value in a pre-clinical setting for the development of vaccines and for testing compositions to determine efficacy of immunisation against infection by malarial Plasmodium parasites. They find use in many aspects of the invention, such as in testing anti-malarial vaccines as described in more detail elsewhere herein. Plasmodium
  • Plasmodium is a genus of parasitic protozoa. Infection with Plasmodium results in the disease malaria, which can be fatal.
  • the malarial Plasmodium parasite has two hosts in its life cycle - a Dipteran insect host and a vertebrate host. The life-cycle is complex, involving a sequence of different stages both in the vector and the vertebrate host.
  • sporozoites which are injected by the insect vector into the vertebrate host's blood; latent hypnozoites, which may rest undetected in the liver for up to 30 years; merosomes and merozoites, which infect the red cells (erythrocytes) of the blood; trophozoites, which grow in the red cells, and schizonts, which divide in red blood cells. Schizonts produce merozoites, which leave to infect more red cells.
  • the sexual forms, gametocytes are taken up by other insect hosts during feeding. Gametocytes develop into gametes in the insect midgut, and then fertilise each other to form motile zygotes, which escape the gut.
  • Plasmodium may be any Plasmodium species, such as P. falciparum, P. vivax, P. ovale, or P. malariae. It may be a naturally occuring Plasmodium species, or a laboratory-engineered Plasmodium such as P. berghei that has been engineered to express CSP from Plasmodium falciparum. Such engineered protozoa are useful in animal models, especially with mice as described herein.
  • Antibodies according to the present invention may target or bind Plasmodium sporozoites. Antibodies may inhibit or reduce the risk of Plasmodium infection, specifically the pre-erythrocytic or sporozoite stage of infection. This provides the advantage of targeting the Plasmodium at an early stage of entry to the vertebrate host body, offering the possibility of preventing infection from taking place. Where a body is already infected with Plasmodium, inhibition of sporozoites nevertheless has the advantage of reducing progression of
  • Plasmodium infection and of inhibiting sporozoites in the blood stream thereby lessening the risk of transmission of Plasmodium from that individual to another via insect feeding e.g., mosquito bite or via contact with infected blood.
  • compositions may further comprise one or more additional antigens or antigen fragments from one or more malarial Plasmodium parasites, optionally P. falciparum.
  • Efficacy may be measured in pre-clinical models, such as in mice, and in human clinical trials.
  • One way to measure the ability of a composition to confer protection against a malarial Plasmodium parasite is to administer the composition to a mammal and determine the efficacy of immunisation of that mammal resulting from the administration.
  • Measures of efficacy of immunisation include strength of immune response generated, e.g., antibody titre, and quality of antibodies generated, e.g., in terms of their binding specificity, neutralising ability, or sequence and/or epitope-binding diversity.
  • Efficacy of immunisation can also be determined by subsequently challenging the individual with a malarial Plasmodium parasite and observing the degree to which the individual develops, or is protected from, Plasmodium infection and/or symptoms of malaria. It is useful to measure efficacy of immunisation in populations of individuals, such as cohorts of mice, and to determine protection against challenge in terms of length of survival of the individuals following challenge. It will be appreciated that individuals, or populations of individuals, will usually be compared with control individuals or populations of individuals that receive the same challenge without having previously been immunised with the candidate composition. Control individuals may be placed in the same experimental conditions except that they do not receive the administration of candidate composition. They may for example receive a mock injection - injection of buffer/saline in place of an injection of the test composition. Efficacy of immunisation, and protection from challenge, are thus usually compared with control data.
  • Antibodies according to the present invention may be used to determine the efficacy of immunisation against a malarial Plasmodium parasite. They may be used as diagnostic reagents for determining the level of CSP or Plasmodium in a sample, this being indicative of the level of P. falciparum (or of other Plasmodium species that express CSP, such as P. berghei engineered to express P. falciparum CSP) in the sample, and thus the level of infection in the mammal from whom the sample is obtained, particularly the pre-erythrocytic or sporozoite stage of the parasite. Methods of detecting CSP or Plasmodium with antibodies are described in detail elsewhere herein.
  • a method of determining efficacy of immunisation of a mammal against a malarial Plasmodium parasite may comprise:
  • CSP CSP
  • Plasmodium parasite to a mammal, allowing time for development of an immune response in the mammal;
  • a malarial Plasmodium parasite for example by exposing the mammal to biting by a mosquito infected with the parasite, and allowing time for development of an immune response in the mammal, to provide a challenged mammal; obtaining a sample from the challenged mammal;
  • a method of determining efficacy of immunisation of a mammal against a malarial Plasmodium parasite may comprise:
  • the level of binding being negatively correlated with the efficacy of immunisation.
  • binding may be quantified relative to binding in samples obtained from control individuals, or relative to positive and/or negative control samples.
  • the CSP (or fragment thereof, or NANP repeat peptide) or the Plasmodium may be administered by any method suitable to generate an immune response in the mammal. Common methods of delivering vaccines include by intramuscular, subcutaneous or intravenous injection, and any such method may be used here. Other possible methods of administration are described elsewhere herein, and may be applied to methods of administering the CSP (or fragment thereof, or NANP repeat peptide) or the Plasmodium.
  • Administering the candidate vaccine composition comprising the antigen or antigens may comprise injecting live or killed Plasmodium, or antigen or antigens purified from
  • the antigen or antigens may be administered in combination with an adjuvant.
  • mice are a suitable test animal for vaccine development.
  • the mammal in these methods may thus be a mouse.
  • a suitable Plasmodium parasite is P. berghei, which can be engineered to express P. falciparum CSP or a fragment thereof comprising the NANP repeat region, ensuring that the P. berghei is bound by antibodies according to the present invention.
  • transgenic mice containing a human antibody repertoire are employed, this provides a model of infection of humans by P. falciparum, the antibody response in the mice representing a model antibody response generated by the human immune system.
  • the KymouseTM is especially suitable for use in such models. It is possible to use other transgenic mice or other test animals engineered to express antibodies with human variable regions, and examples of these are mentioned elsewhere herein.
  • the generation of antibodies according to the present invention provides an indication that the mouse generates an effective immune response.
  • generation of antibodies according to the present invention is an indication that the human generates an effective immune response against P. falciparum.
  • Antibodies according to the invention may thus be used as a measure of vaccine efficacy in anti-malarial vaccine trials, whether pre-clinical (e.g., in transgenic animals expressing antibodies with human variable domains) or clinical (in humans).
  • a method of determining efficacy of immunisation of a mammal against a malarial Plasmodium parasite may comprise:
  • CSP CSP
  • a fragment thereof comprising the NANP repeat region, or a NANP repeat peptide, and optionally one or more other antigens of the Plasmodium parasite to a mammal, allowing time for development of an immune response in the mammal, and obtaining a sample from the mammal;
  • a method of determining efficacy of immunisation of a mammal against a malarial Plasmodium parasite may comprise:
  • a sample obtained from a mammal wherein the mammal has received an administration of CSP, a fragment thereof comprising the NANP repeat region, or a NANP repeat peptide, and optionally one or more other antigens of the Plasmodium parasite, and wherein the sample has been obtained after allowing time for development of an immune response;
  • Exemplary antibodies are antibodies comprising the antibody 667 or antibody 668 CDRs, and variants thereof, as described herein.
  • Assaying for the presence of an antibody molecule may comprise identifying whether any B cells in the mammal encode antibodies according to the present invention, for example by sequencing nucleic acid from the B cells.
  • the administered composition may be a vaccine or a candidate vaccine, and may comprise one or more adjuvants.
  • the methods described here may be used to determine efficacy of vaccination of a mammal, for example to confirm, predict or test for protection of a vaccinated human individual against infection by a malarial Plasmodium parasite.
  • the methods may also be used in the context of pre-clinical or clinical trials involving a candidate vaccine composition.
  • the method may further comprise challenging the mammal with a malarial
  • Plasmodium parasite by exposing the mammal to biting by a Plasmodium-infected mosquito, and determining the degree of protection against challenge, as described.
  • an antibody VH domain comprising complementarity determining regions (CDRs) HCDR1 , HCDR2 and HCDR3, and
  • an antibody VL domain comprising complementarity determining regions LCDR1 ,
  • HCDR3 is an HCDR3 of antibody 666, antibody 667, antibody 668 or antibody 669, or comprises the antibody 666, 667, 668 or 669 HCDR3 with 1 , 2, 3, 4 or 5 amino acid alterations.
  • 666, 667, 668 or 669 or comprises the antibody 666, 667, 668 or 669 HCDR2 with 1 , 2, 3, 4 or 5 amino acid alterations.
  • HCDR1 is an HCDR1 of antibody 666, 667, 668 or 669, or comprises the antibody 666, 667, 668 or 669 HCDR1 with 1 , 2, 3, 4 or 5 amino acid alterations.
  • antibody heavy chain CDRs are those of antibody 666, antibody 667, antibody 668 or antibody 669, or comprise the antibody 666, 667, 668 or 669 heavy chain CDRs with 1 , 2, 3, 4 or 5 amino acid alterations.
  • an antibody VH domain comprising complementarity determining regions HCDR1 , HCDR2 and HCDR3, and
  • an antibody VL domain comprising complementarity determining regions LCDR1 , LCDR2 and LCDR3,
  • LCDR3 is a LCDR3 of antibody 666, antibody 667, antibody 668 or antibody 669, or comprises the antibody 666, 667, 668 or 669 LCDR3 with 1 , 2, 3, 4 or 5 amino acid alterations.
  • LCDR2 is an LCDR2 of antibody 666, 667, 668 or 669, or comprises the antibody 666, 667, 668 or 669 LCDR2 with 1 , 2, 3, 4 or 5 amino acid alterations.
  • antibody light chain CDRs are those of antibody 666, antibody 667, antibody 668 or antibody 669, or comprise the antibody 666, 667, 668 or 669 light chain CDRs with 1 , 2, 3, 4 or 5 amino acid alterations.
  • (i) is derived from recombination of a human heavy chain V gene segment, a human heavy chain D gene segment and a human heavy chain J gene segment, wherein
  • V segment is IGHV1 -3;
  • the D gene segment is IGHD3-10, IGHD3-9, IGHD4-1 1 or IGHD2-2; and/or the J gene segment is IGHJ6, or
  • (ii) comprises framework regions FR1 , FR2, FR3 and FR4, wherein
  • FR1 aligns with human germline V gene segment IGHV1 -3 with up to 1 , 2, 3, 4 or 5 amino acid alterations
  • FR2 aligns with human germline V gene segment IGHV1 -3 with up to 1 , 2, 3, 4 or 5 amino acid alterations
  • FR3 aligns with human germline V gene segment IGHV1 -3 with up to 1 , 2, 3, 4 or 5 amino acid alterations, and/or
  • FR4 aligns with human germline J gene segment IGHJ6 with up to 1 , 2, 3, 4 or 5 amino acid alterations.
  • An isolated antibody comprising an antibody VH domain which
  • (i) is derived from recombination of a human heavy chain V gene segment IGHV1 -3, a human heavy chain D gene segment and a human heavy chain J gene segment, or
  • (ii) comprises framework regions FR1 , FR2, FR3 and FR4, wherein FR1 , FR2 and FR3 each align with human germline V segment IGHV1 -3 with up to 1 , 2, 3, 4 or 5 amino acid alterations,
  • the antibody inhibits the pre-erythrocytic stage of Plasmodium infection in a mammal, reduces risk of malaria in a mammal, reduces one or more symptoms of malaria in a mammal, reduces progression of Plasmodium infection in a mammal, and/or reduces transmission, or reduces risk of transmission, of Plasmodium between mammals.
  • (i) is derived from recombination of a human light chain V gene segment and a human light chain J gene segment, wherein
  • V segment is IGLV2-23, and/or
  • the J gene segment is IGLJ3; or
  • (ii) comprises framework regions FR1 , FR2, FR3 and FR4, wherein
  • FR1 aligns with human germline V gene segment IGLV2-23 with up to 1 , 2, 3, 4 or 5 amino acid alterations
  • FR2 aligns with human germline V gene segment IGLV2-23 with up to 1 , 2, 3, 4 or 5 amino acid alterations
  • FR3 aligns with human germline V gene segment IGLV2-23 with up to 1 , 2, 3, 4 or 5 amino acid alterations, and/or
  • FR4 aligns with human germline J gene segment IGLJ3 with up to 1 , 2, 3, 4 or 5 amino acid alterations.
  • An antibody according to any of the preceding clauses comprising an antibody VL domain derived from recombination of a human light chain V gene segment and a human light chain J gene segment, wherein
  • V segment is IGLV2-23, and optionally
  • the J gene segment is IGLJ3.
  • An antibody according to any of the preceding clauses comprising an antibody VH domain which is the VH domain of antibody 666, antibody 667, antibody 668 or antibody 669, or which has an amino acid sequence at least 90 % identical to the antibody VH domain sequence of antibody 666, antibody 667, antibody 668 or antibody 669.
  • An antibody according to any of the preceding clauses comprising an antibody VL domain which is the VL domain of antibody 666, antibody 667, antibody 668 or antibody 669, or which has an amino acid sequence at least 90 % identical to the antibody VL domain sequence of antibody 666, antibody 667, antibody 668 or antibody 669.
  • an antibody VH domain which is selected from the VH domain of antibody 666, antibody 667, antibody 668 or antibody 669, or which has an amino acid sequence at least 90 % identical to the antibody VH domain sequence of antibody 666, antibody 667, antibody 668 or antibody 669, and
  • an antibody VL domain which is the VL domain of said selected antibody, or which has an amino acid sequence at least 90 % identical to the antibody VL domain sequence of said selected antibody.
  • composition comprising an isolated antibody according to any of the preceding clauses and a pharmaceutically acceptable excipient.
  • composition comprising isolated nucleic acid encoding an antibody according to any of the preceding clauses and a pharmaceutically acceptable excipient.
  • composition according to clause 28 or clause 29 formulated for intravenous or subcutaneous administration.
  • 33. A composition according to clause 32, wherein the one or more additional antibodies bind one or more antigens of a malarial Plasmodium parasite.
  • the method comprises administering the antibody or composition to a mammal.
  • the method comprising administering atherapeutically effective amount of an antibody according to any of clauses 1 to 27 or of a composition according to any of clauses 28 to 34 to the mammal.
  • a method of producing an antibody or an antibody VH or VL domain comprising culturing host cells according to clause 40 under conditions for expression of the antibody or antibody VH or VL domain, and obtaining the expressed antibody or domain.
  • 42. A method according to clause 41 , further comprising isolating and/or purifying the antibody or antibody VH or VL domain.
  • a method according to clause 43 comprising formulating the antibody, or antibody VH or VL domain, into a composition for subcutaneous or intravenous administration.
  • a method for producing an antibody according to clause 1 comprising (i) providing, by way of addition, deletion, substitution or insertion of one or more amino acids in the amino acid sequence of a parent antibody VH domain, an antibody VH domain that is an amino acid sequence variant of the parent antibody VH domain,
  • parent antibody VH domain is the VH domain of any of antibodies 666, 667, 668 and 669 or a VH domain comprising the heavy chain complementarity determining regions of any of those antibodies,
  • VL domain is provided by way of addition, deletion, substitution or insertion of one or more amino acids in the amino acid sequence of a parent VL domain, wherein the parent VL domain is the VL domain of any of antibodies 666, 667, 668 and 669 or a VL domain comprising the light chain
  • a method of determining the presence or absence of CSP or Plasmodium in a sample comprising
  • a diagnostic kit for the use or method as set out in any of clauses 49 to 54 comprising an antibody according to any of clauses 1 to 27, and optionally one or more buffering solutions.
  • a first reagent comprising the antibody according to any of clauses 1 to 27, and a second reagent comprising a detector molecule that binds to the first reagent.
  • a method of determining efficacy of immunisation of a mammal against a malarial Plasmodium parasite comprising:
  • CSP CSP
  • Plasmodium parasite to a mammal
  • a method of determining efficacy of immunisation of a mammal against a malarial Plasmodium parasite comprising:
  • a sample obtained from a mammal wherein the mammal has received an administration of CSP, a fragment thereof comprising the NANP repeat region, or a synthetic NANP repeat peptide, and optionally one or more other antigens of the Plasmodium parasite, and wherein the sample has been obtained after allowing time for development of an immune response;
  • administering the antigen or antigens comprises injecting live or killed Plasmodium, or antigen or antigens purified from
  • Plasmodium or recombinantly expressed into the mammal, optionally together with an adjuvant.
  • a method according to any of clauses 61 to 63 further comprising challenging the mammal with a malarial Plasmodium parasite by exposing the mammal to biting by a Plasmodium-infected mosquito, and determining the degree of protection against challenge.
  • KymiceTM a transgenic mouse platform capable of generating antibodies with human variable domains, were immunised with CSP to generate anti-CSP antibodies.
  • the KymouseTM platform used in this study is composed of two strains of mice,
  • Kymouse HK and Kymouse HL both of which carry human variable domains and mouse constant domains.
  • the antibody repertoire of these mice are composed of human V, D and J segments.
  • the endogenous mouse variable genes have been silenced and make up a very small portion of the repertoire (less than 0.5 % of all heavy chain variable regions are of mouse origin).
  • the mice display normal B-cell signalling and development. These mice respond robustly to antigen challenge and produce high affinity antibodies with human-like HCDR3 lengths.
  • Kymouse HK includes engineered IgH and IgK loci. It includes inserted human VH- gene segments and human V-kappa gene segments. It has normal mouse heavy and light constant regions.
  • Kymouse HL includes engineered IgH and IgL loci. It includes inserted human VH- gene segments and V-lambda gene segments. It has normal mouse heavy regions and human light constant regions.
  • CSP used for this experiment was sourced from Gennova through the Malaria Vaccine Initiative (MVI). Protein was provided in lyophilised form.
  • the antigen is a 58 kDa protein from P. falciparum strain 3D7; GenBank accession number AL034558.
  • a synthetic nucleotide sequence encoding the full-length, mature translated protein sequence for PfCSP (3D7 strain; GenBank accession number AL034558) was commercially synthesised with codons optimised for maximizing expression of the heterologous gene in E. coli. This synthetic gene encoded the predicted full-length mature protein with a carboxy-terminal hexa-histidine tag, without the signal sequence and putative GPI anchor sequence.
  • E. coli BL21 (DE3). These host cells contain a chromosomal copy of T7 RNA polymerase gene under the control of lacUV5 promoter which is induced by addition of lactose analogue such as isopropyl ⁇ -D-thiogalactopyranoside (IPTG). IPTG induces production of T7 RNA polymerase allowing transcription of the target DNA in the plasmid.
  • BL21 (DE3) strain is deficient in both Ion and ompT proteases, thus improving stability of the recombinant protein expressed in these host cells.
  • the purified antigen has been show to elicit a strong T-cell immune response by Elispot. Nucleotide and amino acid sequences of CSP are shown in Figures 6 and 7. The antigen has also previously been shown to be immunogenic in immunisations in animals (Kastenmijller K, Espinosa DA, Trager L,
  • Post-immunisation antibody responses in the KymiceTM were identified from blasting B cells using the IRCTM sequencing technology.
  • IRCTM technology has been described by Atreca, Inc. in published patent applications. This platform captures full length sequences, enabling detection of somatic mutations across the entire antibody variable region. Natively paired heavy and light chain IgG variable regions are analysed, and constant region isotype assignments can be made.
  • IRC corrects for sequence errors introduced at all steps of a single-cell analysis process and provides unbiased output by correcting for biases from PCR amplification and other sources.
  • IRC can be applied to any B cell type, other isotypes, and any species for which constant region sequences are known, including blasted B cells (e.g.
  • IRC immunosorbent assay for human plasmablasts, mouse splenocytes, etc.
  • antigen sorted B cells e.g. antigen- binding memory B cells.
  • IRC was used here to provide the signal peptide, full variable region, and a portion of the constant region of natively-paired heavy and light chain sequences from individual B cells of the immunised Kymice.
  • Candidate antibody lineages for expression and testing were chosen based on sequence-features such as the degree of lineage dominance, somatic hypermutation levels, and apparent convergence of lineages across multiple CSP-immunised Kymice. Forty-eight antibody sequences representing thirty-seven diverse, putative antibody lineages were produced via gene synthesis and recombinant expression as fully human antibodies.
  • DNA sequences for paired heavy chain (HC) and light chain (LC) IgG variable regions were synthesised and subcloned into expression vector pLEV123 (LakePharma, Inc.). Heavy chain variable region sequences were fused to the signal peptide
  • MDPKGSLSWRILLFLSLAFELSYG SEQ ID NO: 51
  • human lgG1 constant regions The light chain variable region sequences were fused to the signal peptide
  • METDTLLLWVLLLWVPGSTG (SEQ ID NO: 53) for the kappa light chain, followed by the compatible human kappa or lambda light chain constant regions.
  • HEK293 cells were seeded in shake flasks one day before transfection, and were grown using serum-free chemically defined media.
  • the DNA expression plasmids were scaled up and transiently transfected into 10-30 ml of suspension HEK293 cells using LakePharma's standard operating procedure for transient transfection. After 20 hours, cultures were fed and production continued for 5 days. Cells were sampled to obtain the viabilities and viable cell counts, and titers were measured (Octet QKe, ForteBio). On day 5, cells were sampled to obtain the viabilities and viable cell counts, and titers were measured (Octet QKe, ForteBio) before harvesting the cell cultures.
  • Protein A affinity purification The conditioned media from HEK293 cells expressing antibody were harvested from the transient transfection production run by centrifugation. The supernatant was run over a Protein A column and eluted with a low pH buffer. Filtration using a 0.2 ⁇ membrane filter was performed before aliquoting. After purification and filtration, the protein concentration was calculated from the OD280 and the extinction coefficient. Antibodies were formulated in HEPES buffer (200 mM HEPES, 100 mM NaCI, 50 mM NaOac, pH 7.) CE-SDS analysis was performed (LabChip GXII, Perkin Elmer) to ensure antibody quality.
  • ELISA positive lineages Seven of nine ELISA positive lineages were expressed as full-length antibodies with human lgG1 constant region and further characterised using bio-layer interferometry and competitive binding assays to identify distinct binding sites.
  • CSP binding site was determined via ELISA with CSP fragments.
  • Binding affinities against rCSP were measured via ForteBio Octet Red69 using two different assay formats. By changing the Octet assay antibody-antigen orientation (see methods section below for two formats), the dose-dependent responses of Ab667 and Ab668 toward Streptavidin-sensor captured biotin/rCSP were obtained and the kinetics constants were calculated. The kinetic analysis of Ab667 and Ab668 binding toward biotin/rCSP by Octet was used to generate the kinetic constants shown in Table 3 and illustrate the high affinity of antibodies 667 and 668 to the CSP antigen. Loading Sample ID KD (M) kon(1 /Ms) kdis(1 /s) Full R A 2 Full ⁇ ⁇ 2
  • Anti-CSP mAbs 667 and 668 were shown to bind to rPF-CSP and to the NANP repeat region, while lacking binding capacity to the Pf16 motif at the C-terminus region of CSP. See Figure 8.
  • Epitope binning tests antibodies in a pairwise manner; antibodies competing for the same binding region are grouped together into bins.
  • epitope binning was performed on Octet QKe system (ForteBio) using two assay formats. The biosensors were hydrated in sample diluent (0.1 % BSA in PBS and 0.02% Tween 20) and preconditioned in pH 1 .9 Glycine.
  • the first assay format (Assay Format 1 ) used Anti-hFc (AHC) kinetic grade biosensors (ForteBio, #18-5060) to capture rCSP/Ab1 mix (rCSP 100nM, Ab1 300nM, 60 seconds); the sensors were then dipped into high concentration of hFc (150 ug/mL) for 300 seconds to saturate the AHC sensor. Ab1 of 300nM was loaded again for 300 seconds to check for unsaturated Ab1 binding epitope. Lastly, the sensors were dipped into Ab2
  • AHC Anti-hFc
  • a second assay format (Assay Format 2) used Streptavidin (SA) kinetic grade biosensors (ForteBio, #18-5021 ) to immobilize biotinylated rCSP at a concentration of 20 ⁇ for 120 seconds.
  • SA Streptavidin
  • the antigen-loaded SA biosensors were then dipped into Ab1 ("saturating antibody”) at 20 ug/ml for 600 seconds.
  • Ab1 saturatedating antibody
  • a short baseline (30 seconds) was observed using dissociation buffer after the Ab1 loading.
  • the sensor was then dipped into Ab2 ("competing antibody”) at 5 ug/mL and the association was observed for 120 seconds, followed by 120 seconds of dissociation.
  • EPO70034 (NANP repeat region).
  • Peptide Pf16 was reconstituted with DMSO to 20 mg/mL and then diluted with sterile water to obtain a final concentration of 5 mg/mL.
  • EP070034 was reconstituted with sterile water to obtain a final concentration of 5 mg/mL.
  • 96-well plates were coated overnight with the three antigens (including the full-length Pf CSP antigen) at a final concentration of ⁇ g/mL. The plate was blocked with 1 x PBS, 2.5% BSA for 2 hours the next morning at room temperature.
  • Antibody samples were prepared in an 8-point 1 :5 serial dilution series with 1 x PBS, 1 % BSA with a starting concentration of 50 ⁇ g/mL.
  • the HRP- goat anti-human Fc was diluted 1 :5,000 for detection as described above.
  • Affinity determination was performed on an Octet QKe system (ForteBio).
  • Anti-hFc (AHC) kinetic grade biosensors (ForteBio, #18-5060) were hydrated in sample diluent (0.1 % BSA in PBS and 0.02% Tween 20) and preconditioned in pH 1 .9 Glycine.
  • Antibody was immobilised onto AHC biosensors at a concentration of 20 ⁇ g/mL for 60 seconds.
  • the antibody-loaded AHC biosensors were then dipped into an 8-point dilution series of the Pf CSP antigen (starting at 1000 nM, 1 :3 diluted down). Association was observed for 120 seconds, followed by 180 seconds of dissociation.
  • a short baseline (30 seconds) was established using dissociation buffer after AHC loading.
  • Parallel references were set up by using unloaded bare sensor and sensor dipped into an 8-point dilution series of the antigen. During data processing, the parallel reference was used for normalization.
  • the binding affinity of Pf CSP to each antibody was characterized by fitting kinetic sensorgrams to a monovalent binding model (1 :1 binding). This assay set up was used for five related antibodies (AB-000662, AB-000640, AB-000649, AB000646 and AB000643).
  • Example 4a In vivo protection studies using mouse malaria challenge model
  • Candidate antibodies are tested in a mouse malaria challenge model. This in vivo mouse protection assay uses intravenous passive transfer of selected antibodies (300 ⁇ g/mouse) followed by challenge with chimeric P. berghei encoding full-length P.
  • antibodies are injected intravenously. Sporozoites are injected 30 minutes later by intravenous challenge. Parasite liver burden (measured by PCR) is used as a readout. Antibodies that show protection in this IV challenge are then further tested in a second experiment, using mosquito bite challenge. Parasitaemia can be used as a readout to assess sterile protection.
  • Figure 5 illustrates principles of the challenge model.
  • livers are extracted and RNA is isolated from liver homogenates.
  • Real-time PCR targeting the parasite 18S rRNA sequence, is used to measure the relative level of liver-stage parasites.
  • Liver-stage parasite load can be quantified in terms of the number of P. berghei 18S rRNA copies in liver. % reduction of parasite load can be determined relative to control mice who do not receive the candidate antibody or who receive a control antibody (naive mice).
  • mice may be monitored for a period of, e.g., 7 or 14 days, and % mice remaining free of blood-stage parasites is determined for challenged mice and control mice.
  • Example 4a The mouse challenge described in Example 4a was performed to assess the ability of passively-transferred anti-CSP mAbs to protect mice from sporozoite challenge.
  • these in vivo protection studies involving transgenic sporozoites comprised initial screening by IV injection of 300 ug of Ab per animal followed by measurement of liver stage burden.
  • Results are shown in Figure 9 and Figure 10. The data indicate that antibodies 667 and 668 confer significant and very strong protection relative to the 2A10 positive control.
  • mAbs 667 and 668 confer protection from transgenic sporozoite challenge and evaluate whether such protection could be sterilising.
  • a second challenge was performed using three different concentrations of antibody. The outcome was evaluated by determination of parasitemia at various time points post-challenge.
  • mice Upon passive transfer of monoclonal antibodies, mice were anesthetized with 2% Avertin and mosquitoes allowed to feed on mice for -10 minutes. After feeding, the number of mosquitoes positive for a blood meal was determined. On days 4 - 1 1 after challenge, blood smears were taken from mice to determine parasitemia. % sterile protection was determined by dividing the number of uninfected mice by the total number of mice challenged. Pre-patent period, the number of days prior to detection of parasites in the blood, was also recorded.
  • the impact of anti-CSP antibodies on sporozoites' gliding motility was assessed using methods previously described by Ejigiri et al. (Ejigiri et al., Shedding of TRAP by a Rhomboid Protease from the Malaria Sporozoite Surface is Essential for Gliding Motility and Sporozoite Infectivity, PLoS Pathog 8(7), 2012). Briefly, P.
  • falciparum sporozoites were pre-incubated with the indicated antibodies (100 ug/ml) for 30 min and then added to wells in the presence of bovine serum albumin and incubated at 37°C for 1 hr. They were then fixed and trails were stained with anti-CSP antibody and counted using a fluorescence microscope.
  • Anti-CSP antibodies mAb667 and mAb668 abolished gliding motility of the sporozoites in this assay. Data are shown in Figure 12.
  • liver stage development assay tests antibodies for the ability to block sporozoite development in hepatocytes.
  • ILSDA is an excellent candidate assay to identify correlates of humoral protection, particularly against the liver stage of malaria infection.
  • the Inhibition of Liver Stage Development Assay (ILSDA) was performed as previously described by Zou et al. (Zou et al, Towards an optimized inhibition of liver stage development assay (ILDSA) for Plasmodium falciparum, Malaria Journal 12:394, 2013).
  • the NF54 strain of Plasmodium falciparum (Pf) sporozoites obtained from salivary gland dissections
  • mAbs monoclonal antibodies
  • the sporozoites-mAbs mixtures were then introduced into the wells containing cryopreserved human hepatocytes (CPHH; BioReclamation IVT, Baltimore MD) and incubated at 37 °C for 3 hours to allow sporozoites to infect hepatocytes.
  • the anti- CSP mAb 2A10 was used as a positive control.
  • CPHH were washed with fresh hepatocyte culture media to remove non-invaded sporozoites and incubated at 37°C for 96 hours.
  • the RNA from the cells was then harvested for downstream quantitative real-time PCR (qRT-PCR) analysis. Pf 18s rRNA level were quantified to determine the inhibitive ability of the tested mAbs.
  • Example 7 Inhibition of sporozoite traversal and invasion (ISTI)
  • Anti-CSP antibodies mAb667 and mAb668 dramatically reduced both hepatocyte traversal ( Figure 14) and hepatocyte invasion (Figure 15) in this assay.
  • Antibody 666 and antibody 669 were also tested and were confirmed to be functional hits in the ISTI assay.
  • Example 8 Sporozoite challenge into FRG Hu-Hep mice
  • a human liver (Hu-Hep) mouse challenge model was used to assess the ability of passively-transferred anti-CSP mAbs to protect mice from sporozoite challenge.
  • mice were purchased from Yecuris, Inc. and used in passive transfer experiments as previously described (K. Sack et al., Model for in vivo assessment of humoral protection against malaria sporozoite challenge by passive transfer of monoclonal antibodies and immune serum. Infect Immun 82, 808-817 (2014), and Sack et al., submitted). Briefly, mice were intravenously injected with 150 ⁇ g of mAb or non-specific murine IgG (mock) 16-24 hours prior to infection by bite of 50 mosquitos infected with Pf GFP-luciferase parasites (Vaughan et al. 2012) for 10 minutes. Six days after infection, parasite liver burden was assessed by bioluminescent imaging.
  • Percent of mock liver burden was calculated by normalizing all liver burdens to the average of the mock-injected group within each independent experiment.
  • the 3C1 anti-CSP mAb (Zavala et al., J Exp Med 157:1947-1957 1983) had previously been shown in this model to mediate higher inhibition of infection than the 2A10 positive control and was therefore used as the positive control in the present experiment.
  • the phylogenetic family of Ab667 contains Ab666 and a number of further antibodies, as shown in Table 5.
  • DSFYDILTGPVYHYYGMDV WINAGNGYTKYSQKFQD GYTFTNYAMH CSYAGSSTWV DVSKRPS TGTSNDVGVYNHVS
  • the phylogenetic family of Ab668 contains Ab669 and a number of further 5 antibodies, as shown in Table 6.
  • antibodies have the same germline gene usage, high identity light chains and high identity in the HCDR1 and HCDR2. It is believed that antibodies comprising VH domains having these CDRs may be functionally active in a similar way to Ab666, 667, 668 and 669. This may be confirmed through assays as described in the above Examples.
  • Ab666 lineage Ab666, Ab666-1 , Ab666-2, Ab666-3, Ab666-4.
  • Ab667 lineage Ab667, Ab667-1 , Ab667-2, Ab667-3.
  • Ab668 lineage Ab668, Ab668-1 .
  • Ab669 lineage Ab669, Ab669-1 , Ab669-2, Ab669-3, Ab669-4, Ab669-5, Ab669-6.
  • Lineage trees are shown in Figure 18 (Ab666 lineage), Figure 19 (Ab667 lineage), Figure 20 (Ab668 lineage) and Figure 21 (Ab669 lineage).
  • SEQ ID NO: 1 18 Ab669-4 H contig nucleotide sequence

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Biophysics (AREA)
  • Oncology (AREA)
  • Genetics & Genomics (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Pulmonology (AREA)
  • Communicable Diseases (AREA)
  • Biochemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention décrit des anticorps qui se lient à la protéine circumsporozoïte (CSP) de Plasmodium falciparum. De tels anticorps peuvent être utilisés comme agents anti-paludéens, pour conférer la protection contre l'infection par les parasites paludéens tels que P. falciparum par la transmission par un vecteur type insecte. Les anticorps anti-CSP peuvent être utilisés afin d'effectuer un diagnostic du paludisme. Des procédés de détermination de l'efficacité des compositions vaccinales candidates dans le développement et le test de vaccins anti-paludéens sont également décrits.
PCT/GB2017/050786 2016-03-21 2017-03-21 Anticorps anti-paludéens qui se lient à la protéine circumsporozoïte WO2017163049A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/977,762 US11066464B2 (en) 2016-03-21 2018-05-11 Anti-malarial antibodies that bind circumsporozoite protein

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662311059P 2016-03-21 2016-03-21
US62/311,059 2016-03-21

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/977,762 Continuation-In-Part US11066464B2 (en) 2016-03-21 2018-05-11 Anti-malarial antibodies that bind circumsporozoite protein

Publications (1)

Publication Number Publication Date
WO2017163049A1 true WO2017163049A1 (fr) 2017-09-28

Family

ID=58455341

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2017/050786 WO2017163049A1 (fr) 2016-03-21 2017-03-21 Anticorps anti-paludéens qui se lient à la protéine circumsporozoïte

Country Status (1)

Country Link
WO (1) WO2017163049A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11066464B2 (en) 2016-03-21 2021-07-20 Kymab Limited Anti-malarial antibodies that bind circumsporozoite protein
EP3621643A4 (fr) * 2017-05-11 2021-07-21 Atreca, Inc. Anticorps antipaludéens qui se lient à la protéine circumsporozoïte

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011004192A1 (fr) 2009-07-08 2011-01-13 Genome Research Limited Modèles d'animaux et molécules thérapeutiques
US20110065902A1 (en) 2008-12-15 2011-03-17 Regeneron Pharmaceuticals, Inc. High affinity human antibodies to pcsk9
US20120093818A1 (en) 2007-08-23 2012-04-19 Simon Mark Jackson Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (pcsk9)
WO2016033547A1 (fr) * 2014-08-29 2016-03-03 Sorrento Therapeutics, Inc. Anticorps thérapeutiques qui se lient à oprf et oprl

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120093818A1 (en) 2007-08-23 2012-04-19 Simon Mark Jackson Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (pcsk9)
US20110065902A1 (en) 2008-12-15 2011-03-17 Regeneron Pharmaceuticals, Inc. High affinity human antibodies to pcsk9
WO2011004192A1 (fr) 2009-07-08 2011-01-13 Genome Research Limited Modèles d'animaux et molécules thérapeutiques
WO2016033547A1 (fr) * 2014-08-29 2016-03-03 Sorrento Therapeutics, Inc. Anticorps thérapeutiques qui se lient à oprf et oprl

Non-Patent Citations (42)

* Cited by examiner, † Cited by third party
Title
"LIPOSOMES IN THE THERAPY OF INFECTIOUS DISEASE AND CANCER", pages: 317 - 327
"Medical Applications of Controlled Release", 1974, CRC PRES., article "In another embodiment, polymeric materials can be used"
"Remington's Pharmaceutical Sciences", MACK PUBLISHING COMPANY, EASTON, PA
A. N. DOUGLASS; P. G. METZGER; S. H. KAPPE; A. KAUSHANSKY: "Flow Cytometry-Based Assessment of Antibody Function Against Malaria Pre-erythrocytic Infection", METHODS MOL BIOL, vol. 1325, 2015, pages 49 - 58
ANKER, ZAVALA; POLLOK, EUR J IMMUNOL., vol. 20, 1990, pages 2757 - 2761
ANKER; ZAVALA; POLLOK, EUR J IMMUNOL., vol. 20, 1990, pages 2757 - 2761
BRUNA-ROMERO ET AL., INT. J. PARASITOL., vol. 31, 2001, pages 1499 - 1502
CRAIG PRZYSIECKI ET AL: "Sporozoite neutralizing antibodies elicited in mice and rhesus macaques immunized with a Plasmodium falciparum repeat peptide conjugated to meningococcal outer membrane protein complex", FRONTIERS IN CELLULAR AND INFECTION MICROBIOLOGY, vol. 2, 30 November 2012 (2012-11-30), pages 1 - 11, XP055375838, DOI: 10.3389/fcimb.2012.00146 *
DATABASE EMBL [online] 23 February 2004 (2004-02-23), "Homo sapiens (human) partial immunoglobulin lambda-1 variable region", XP002770487, retrieved from EBI accession no. EMBL:CAE18330 Database accession no. CAE18330 *
DATABASE EPO Proteins [online] 31 October 2008 (2008-10-31), "Sequence 253 from Patent WO2007120767.", XP002770486, retrieved from EBI accession no. EPOP:FB656442 Database accession no. FB656442 *
DATABASE Geneseq [online] 3 March 2016 (2016-03-03), "Anti-OprF/OprI antibody heavy chain variable region, SEQ ID 41.", XP002770488, retrieved from EBI accession no. GSP:BCM88055 Database accession no. BCM88055 *
DEL GUIDICE ET AL., J. CLIN. MICROBIOL., vol. 25, no. 1, 1987, pages 91 - 96
DORDO ET AL., J. MOL BIOL, vol. 217, 1999, pages 721 - 739
E-CHIANG LEE ET AL: "Complete humanization of the mouse immunoglobulin loci enables efficient therapeutic antibody discovery", NATURE BIOTECHNOLOGY, vol. 32, no. 4, 16 March 2014 (2014-03-16), pages 356 - 363, XP055131583, ISSN: 1087-0156, DOI: 10.1038/nbt.2825 *
ELIZABETH H. NARDIN ET AL: "Circumsporozoite proteins of human malaria parasites Plasmodium falciparum and Plasmodium vivax", J. EXP. MED., vol. 156, no. 1, 1 July 1982 (1982-07-01), pages 20 - 30, XP055375832 *
ESPINOSA DA1; YADAVA A; ANGOV E; MAURIZIO PL; OCKENHOUSE CF; ZAVALA F.: "Development of a chimeric Plasmodium berghei strain expressing the repeat region of the P. vivax circumsporozoite protein for in vivo evaluation of vaccine efficacy", INFECT IMMUN., vol. 81, no. 8, 28 May 2013 (2013-05-28), pages 2882 - 7
GOODSON, MEDICAL APPLICATIONS OF CONTROLLED RELEASE, vol. 2, 1984, pages 115 - 138
HERINGTON ET AL., NATURE, vol. 328, 1987, pages 257 - 259
HERRINGTON D A ET AL: "SAFETY AND IMMUNOGENICITY IN MAN OF A SYNTHETIC PEPTIDE MALARIA VACCINE AGAINST PLASMODIUM FALCIPARUM SPROZOITES", NATURE, vol. 328, no. 16, 16 July 1987 (1987-07-16), NATURE PUBLISHING GROUP, UNITED KINGDOM, pages 257 - 259, XP000986125, ISSN: 0028-0836, DOI: 10.1038/328257A0 *
IMWONG ET AL., THE LANCET, 2017
J. G. KUBLIN ET AL.: "Complete attenuation of genetically engineered Plasmodium falciparum sporozoites in human subjects", SCI TRANSL MED, vol. 9, 2017
K. SACK ET AL.: "Model for in vivo assessment of humoral protection against malaria sporozoite challenge by passive transfer of monoclonal antibodies and immune serum", INFECT IMMUN, vol. 82, 2014, pages 808 - 817
KABAT: "Sequences of Proteins of Immunological Interest", 1987, NATIONAL INSTITUTES OF HEALTH, BETHESDA, MD.
KASTENMULLER K; ESPINOSA DA; TRAGER L; STOYANOV C; SALAZAR AM; POKALWAR S; SINGH S; DUTTA S; OCKENHOUSE CF; ZAVALA F, INFECT IMMUN., vol. 81, no. 3, 28 December 2012 (2012-12-28), pages 789 - 800
KASTENMULLER K; ESPINOSA DA; TRAGER L; STOYANOV C; SALAZAR AM; POKALWAR S; SINGH S; DUTTA S; OCKENHOUSE CF; ZAVALA F: "Full-length Plasmodium falciparum circumsporozoite protein administered with long-chain poly(l«C) or the Toll-like receptor 4 agonist glucopyranosyl lipid adjuvant-stable emulsion elicits potent antibody and CD4+ T cell immunity and protection in mice", INFECT IMMUN., vol. 81, no. 3, 28 December 2012 (2012-12-28), pages 789 - 800
KUMAR SANJAI ET AL: "A chemiluminescent-western blot assay for quantitative detection ofPlasmodium falciparumcircumsporozoite protein", JOURNAL OF IMMUNOLOGICAL METHODS, vol. 390, no. 1, 8 February 2013 (2013-02-08), ELSEVIER SCIENCE PUBLISHERS B.V.,AMSTERDAM, NL, pages 99 - 105, XP028999151, ISSN: 0022-1759, DOI: 10.1016/J.JIM.2013.02.001 *
LANGER, SCIENCE, vol. 249, 1990, pages 1527 - 1533
LEE ET AL., NATURE BIOTECHNOLOGY, vol. 32, no. 4, 2014, pages 356 - 367
LLOYD, THE ART, SCIENCE AND TECHNOLOGY OF PHARMACEUTICAL COMPOUNDING, 1999
MALARIA JOURNAL, vol. 12, 2013, pages 394
MICHAEL R HOLLINGDALE ET AL: "Inhibition of entry of Plasmodium falciparum and P. vivax sporozoites into cultured cells: an in vitro assay of protective antibodies", THE JOURNAL OF IMMUNOLOGY, vol. 132, no. 2, 1 February 1984 (1984-02-01), US, pages 909 - 913, XP055374688, ISSN: 0022-1767 *
PORTER ET AL.: "Transgenic Parasites Stably Expressing Full-Length Plasmodium falciparum Circumsporozoite Protein as a Model for Vaccine Down-Selection in Mice Using Sterile Protection as an Endpoint", CLIN VACCINE IMMUNOL., vol. 20, no. 6, 2013, pages 803 - 810
POWELL ET AL.: "Compendium of excipients for parenteral formulations", J PHARM SCI TECHNOL, vol. 52, 1998, pages 238 - 311
RAUL HERRERA ET AL: "Reversible Conformational Change in the Plasmodium falciparum Circumsporozoite Protein Masks Its Adhesion Domains", INFECTION AND IMMUNITY, vol. 83, no. 10, 1 October 2015 (2015-10-01), pages 3771 - 3780, XP055375494, ISSN: 0019-9567, DOI: 10.1128/IAI.02676-14 *
ROBERTO ANKER ET AL: "VH and VL region structure of antibodies that recognize the (NANP)s dodecapeptide sequence in the circumsporozoite protein of Plasmodium falciparum", EUR. J. IMMUNOL, vol. 20, 1 January 1990 (1990-01-01), pages 2757 - 2761, XP055374618, DOI: 10.1002/eji.1830201233 *
S. FRENCH; B. ROBSON, J. MOL. EVOL., vol. 19, 1983, pages 171
SEFTON, CRC CRIT. REF. BIOMED. ENG., vol. 14, 1987, pages 201
SHIELDS ET AL., JBC, vol. 277, 2002, pages 26733
TAYLOR ET AL., J. THEOR. BIOL., vol. 119, 1986, pages 205 - 218
TREAT ET AL.: "Liposomes in the Therapy of Infectious Disease and Cancer", 1989, pages: 353 - 365
WARD ET AL., NATURE, vol. 341, 1989, pages 544 - 546
ZAVALA ET AL., J EXP MED, vol. 157, 1983, pages 1947 - 1957

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11066464B2 (en) 2016-03-21 2021-07-20 Kymab Limited Anti-malarial antibodies that bind circumsporozoite protein
EP3621643A4 (fr) * 2017-05-11 2021-07-21 Atreca, Inc. Anticorps antipaludéens qui se lient à la protéine circumsporozoïte

Similar Documents

Publication Publication Date Title
EP3621643A1 (fr) Anticorps antipaludéens qui se lient à la protéine circumsporozoïte
US8414890B2 (en) Human antibodies to human RANKL, encoding nucleic acids and methods of treatment
US8613927B2 (en) High affinity human antibodies to human nerve growth factor
AU2011258342B2 (en) Antibodies to human GDF8
TWI644921B (zh) 抗fel d1之人類抗體及其使用方法
CN104603149B (zh) 与预防和治疗狂犬病感染相关的组合物和方法
US10081675B2 (en) Antibodies targeting M-CSF
WO2015085140A1 (fr) Compositions antipaludiques
US20240101649A1 (en) ANTI-CfaE ANTIBODIES AND METHODS OF USE
WO2017163049A1 (fr) Anticorps anti-paludéens qui se lient à la protéine circumsporozoïte
US11066464B2 (en) Anti-malarial antibodies that bind circumsporozoite protein
US11834497B2 (en) Glucose transporter 4 antibodies, methods of making the same, and uses thereof
US10357554B2 (en) AMA-1 epitopes, antibodies, compositions, and methods of making and using the same
KR20150084007A (ko) 항-프로키네티신 수용체 (prokr) 항체 및 이의 용도
EP3180024B1 (fr) Anticorps anti-ospa et procédés d'utilisation
US20230391885A1 (en) Claudin 18.2 antibodies, methods of making the same, and uses thereof
WO2023081851A1 (fr) Anticorps neutralisants dirigés contre la protéine plasmodium falciparum circumsporozoite et leur utilisation
WO2024026301A1 (fr) Méthode d'immunofocalisation d'une réponse immunitaire
WO2020118293A2 (fr) Anticorps qui se lient à la myociline pliée de manière native

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

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

Ref document number: 17714517

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 17714517

Country of ref document: EP

Kind code of ref document: A1