WO2004081046A2 - Human monoclonal antibodies against apociii and their use in the therapy of atherosclerosis - Google Patents

Human monoclonal antibodies against apociii and their use in the therapy of atherosclerosis Download PDF

Info

Publication number
WO2004081046A2
WO2004081046A2 PCT/EP2004/002640 EP2004002640W WO2004081046A2 WO 2004081046 A2 WO2004081046 A2 WO 2004081046A2 EP 2004002640 W EP2004002640 W EP 2004002640W WO 2004081046 A2 WO2004081046 A2 WO 2004081046A2
Authority
WO
WIPO (PCT)
Prior art keywords
apociii
antibody
human
antibodies
atherosclerosis
Prior art date
Application number
PCT/EP2004/002640
Other languages
French (fr)
Other versions
WO2004081046A3 (en
Inventor
Pascal Mettens
Rene Meykens
Philippe Monteyne
Torbjörn SCHIÖTT
Leif Strandberg
Original Assignee
Glaxosmithkline Biologicals S.A.
Bioinvent International A.B.
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 Glaxosmithkline Biologicals S.A., Bioinvent International A.B. filed Critical Glaxosmithkline Biologicals S.A.
Publication of WO2004081046A2 publication Critical patent/WO2004081046A2/en
Publication of WO2004081046A3 publication Critical patent/WO2004081046A3/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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • 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
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]

Definitions

  • the present invention relates to novel therapies, and prophylactic treatments of dyslipidaemia, such as atherosclerotic diseases. Accordingly there is provided, methods of treating or preventing atherosclerosis by passive vaccination through administration to a patient of antibodies that are capable of binding to specific epitopes of Apolipoprotein C-III (ApoCIII).
  • the antibodies of the present invention are potent in the prevention, or reduction, of atherosclerotic plaque formation over prolonged periods of time, thereby reducing the potential of atheroslerosis leading to coronary or cerebrovascular disease.
  • the use of the antibodies of the present invention in medicine.
  • Preferred epitopes of ApoCIII which consist of the targets for the passive immunotherapy aspects of the present invention, are encompased within the regions between amino acid numbers 12-35 and between amino acid numbers 45-76 (particuarly 45-65) of the mature form of human ApoCIII, although other regions of ApoCIII may also be targeted by the passive immunotherapy of the present invention.
  • Atherosclerosis is the leading cause of death and disability in the developed world, and is the major cause of coronary and cerebrovascular deaths, with approximately 7.2 and 4.6 million deaths per year worldwide respectively (Atherosclerosis is generally described in Harrison's Principles of Internal Medicine (14 th Edition, McGraw Hill, pl345-1352), Berliner, J.
  • Atherosclerosis of the coronary arteries commonly causes angina pectoris and myocardial infarction.
  • atherosclerosis of the arteries supplying the central nervous system frequently provokes transient cerebral ischemia and strokes.
  • atherosclerosis can cause intermittent claudication and gangrene and can jeopardise limb viability.
  • Involvement of the splanchnic circulation can cause mesenteric ischemia and bowel infarction.
  • Atherosclerosis can affect the kidney directly (eg causing renal artery stenosis), and in addition, the kidney is a frequent site of atheroembolic disease.
  • Atherogenesis in humans typically occurs over many years, usually many decades.
  • the slow build up of atherogenic plaques in the lining of the vasculature can lead to chronic clinical expressions through blood flow restriction (such as stable effort-induced angina pectoris or predictable and reproducible intermittent claudication).
  • a much more dramatic acute clinical event such as a myocardial infarction or cerebrovascular accident can occur after plaque rupture.
  • the way in which atherosclerosis affects an arterial segment also varies, an additional feature of the heterogeneity and complexity of this disease.
  • Atheromas are usually thought of as stenotic lesions, or plaques, which can limit blood flow, however, atherosclerosis can also cause ectasia and development of aneurysmal disease with an increase in lumen caliber. This expression of atherosclerosis frequently occurs in the aorta, creating a predisposition to rupture or dissection rather than to stenosis or occlusion.
  • LDL Low-density lipoprotein
  • the lipoprotein particles undergo chemical modification, including both oxidation and non-enzymatic glycation. These oxidised and glycated lipoproteins then contribute to many of the subsequent events of lesion development.
  • the chemical modifications attract macrophages within the vessel walls, which internalise the oxidised LDL and become foam cells which initiate lesions called plaques. It is the atherosclerotic plaques which are responsible for the clinical manifestations of atherosclerosis, either they limit blood flow, or allow aneurism, or may even rupture provoking the coronary or cerebrovascular attacks.
  • the development of atherosclerosis is a long process which may occur over decades, which is initiated by an imbalance between atherogenic and protective lipoproteins.
  • cholesterol associated with high-density lipoproteins or HDL (so called “good” cholesterol) and low-density lipoproteins or LDL (so called “bad” cholesterol) levels in the circulation are thought to be markers of increased probability of atherosclerosis (Harrison's Principles of Internal Medicine (14 th Edition, McGraw Hill, pl345-1352)).
  • Cholesterol, cholesterol esters, triacylglycerols and other lipids are transported in body fluids by a series of lipoproteins classified according to their increasing density: chylomicrons, Very Low, Low, Intermediate and High density lipoproteins (CM, VLDL, LDL, IDL and HDL respectively).
  • lipoprotein-complexes consist of a core of hydrophobic lipids surrounded by polar lipids and then by a shell of Apolipoproteins.
  • Apolipoproteins there are at least twelve types of apolipoproteins known, A-I, A-II, A-IV, A-V, B, CI, CII, CIII, D, E, H and J.
  • the different types of lipoproteins may have different functions, for example LDL (which are rich in cholesterol esters) are thought to be associated with the transport of cholesterol to peripheral tissues for new membrane synthesis.
  • apolipoprotein C-III is a 79 amino acid protein produced in the liver and intestine (Brewer et al., J. Biol. Chem. (1974), 249 : 4975-4984; Protter, A.A., et al., 1984, DNA, 3:449-456; Fruchart, J.C. et al,
  • ApoCIIIO is not glycosylated, however ApoCIII 1 and ApoCIII2 are glycosylated and have respectively one and two sialic acid residues (Ito et al, 1989 J.lipd. Res. Nov 30:11 1781-1787).
  • the sugar moiety consists of disaccharide ⁇ -D galactosyl (1-3) ⁇ -N-Acetyl-D-Galactosamine attached to threonine 74 of protein chain by O-glycosidic binding (Assman et al, 1989, BBA 541:234-240).
  • ApoCIIIO ApoCIII 1 and ApoCIII2 represent 14%, 59% and 27% of total ApoCIII respectively. Mutagenesis of the glycosylation site of human ApoCIII does not affect its secretion and lipid binding (Roghani et al., 1988 JBC 34:17925-32).
  • Mature Human ApoCIII has the following ammo acid sequence: tSEAEDASLLSFMQGYMKHATKTAKDALSSVQESQVAQQARGWVTDGFSSL KDYWSTVKDKFSEFWDLDPEVRPTSAVAA 79 (SEQ ID.NO. 1).
  • Plasma concentration of ApoCi ⁇ is positively correlated with levels of plasma triglycerides (Schonfeld et al, Metabolism (1979) 28 : 1001-1010; Kaslyap et al, J. Lip. Res. (1981) 22 : 800-810). Liver perfusion studies demonstrate that ApoCIII inhibits the hepatic uptake of triglyceride-rich lipoproteins (TRL) and their remnants (Shelburne et al, J. Clin. Inves., (1980) 65 : 652-658, Windier et al, J. Lip. Res.
  • TRL triglyceride-rich lipoproteins
  • ApoCIII can exist in different physiological forms, for example, oxidised and non-oxidised forms, and that allelic variants and mutants of ApoCIII may exist.
  • the antibodies of the present invention may recognise any of these forms of ApoCIII. It is a preferred embodiment that the antibodies recognise ApoCIII having the sequence of SEQ ID No 1, or comprising one or more of the sequences as set out in any of SEQ ID Nos 2-48. In particular, these antibodies will recognise the non-oxidised form of ApoCIII.
  • the passive immunotherapies of the present invention target an epitope found within the region between amino acid number 1 and 79, or more preferably an epitope found within the region between amino acid number 1 and 17, 12 and 35, or an epitope found within the region between amino acids 45 and 76 of the human ApoCIII molecule as it exists in the circulation of a human, in addition it is preferred that the immunotherapy targets the epitope that is found within the region between amino acid 12 to 21 or 45 to 65 of human ApoCIII.
  • the sequence of the region between amino acid number 12 and 35 of the human ApoCIII is as follows:
  • MQGYMKHATKTAKDALSSVQESQV (SEQ ID NO.2).
  • the sequence of the region between amino acid number 12 and 21 of the human ApoCIII is as follows: MQGYMKHATK (SEQ ID NO.3)
  • DGFSSLKDYWSTVKDKFSEFWDLDPEVRPTSA (SEQ ID NO: 4) The sequence of the region between amino acid number 45 and 65 of the human ApoCIII is as follows:
  • DGFSSLKDYWSTVKDKFSEFW (SEQ ID NO: 5)
  • the present invention also provides the following fragments of the above peptides within which contain an epitope of ApoCIII which may be targeted by the passive immunotherapies of the present invention:
  • the present invention provides antibodies effective in the prophylaxis or therapy of dyslipidaemia or atherosclerosis which target the epitopes listed in SEQ ID NO.s 1-48, of ApoCIII, and also provides for methods of treatment of atherosclerosis by passive administration of the antibodies of the present invention to individuals in need thereof.
  • the antibodies of the invention recognise the epitopes listed in SEQ ID NO: 1, 3, 6-22.
  • the antibodies of the present invention are functional in the treatment of atherosclerosis, and in a preferred form of the present invention they abrogate the inhibition exerted by ApoCIII on the binding of ApoB to its receptor, and/or the activity of lipoprotein lipase. Such activities may readily be assayed by the man skilled in the art for example by methods described in Fruchard et al, supra; and McConathy et al., supra.
  • the antibodies of the present invention are provided for use in medicine, and for use in the treatment or prevention of atherosclerosis.
  • the antibodies of the present invention will be generally administered for both initial and boosting doses. It is expected that the boosting doses will be adequately spaced, or preferably given at such times where the levels of circulating antibody fall below a desired level.
  • the antibody preparations of the present invention may be used to protect or treat a mammal susceptible to, or suffering from atherosclerosis, by means of administering said antibodies via a systemic route. These administrations may include injection via the intramuscular, intraperitoneal, intradermal or subcutaneous routes.
  • the antibodies will preferably be administered in a pharmaceutical composition, together with a pharmaceutically acceptable carrier.
  • Fully human monoclonal antibodies capable of binding to epitopes of SEQ ID NO.s 1 to 48 (preferably SEQ ID NO: 1, 2 or 3) in the context of the human ApoCIII molecule, and their use in immunotherapy.
  • Antibodies typically comprise two heavy chains linked together by disulphide bonds and two light chains. Each light chain is linked to a respective heavy chain by disulphide bonds. Each heavy chain has at one end a variable domain followed by a number of constant domains. Each light chain has a variable region at one end and a constant domain at its other end. The light chain variable domain is aligned with the variable domain of the heavy chain. The light chain constant domain is aligned with the first constant domain of the heavy chain.
  • the constant domains in the light and heavy chains are not involved directly in binding the antibody to antigen.
  • the variable domains in each pair of light and heavy chains form the antigen binding site.
  • the domains on the light and heavy chains have the same general structure and each domain comprises a framework of four regions, whose sequences are relatively conserved,connected by three complementarity determining regions (CDRs).
  • CDRs complementarity determining regions
  • the four framework regions largely adopt a beta sheet conformation and the CDRs form loops connecting, and in some cases forming part of, the beta-sheet structure.
  • the CDRs are held in close proximity by the framework regions and, with the CDRs from the other domain, contribute to the formation of the antibody binding site.
  • antibodies of the present invention are fully human antibodies.
  • the antibodies should not be recognisable by the host human immune system as foreign, or should elicit a far reduced immune response when administered to a human than the immune response mounted by a human against a humanised or a chimaeric antibody which has regions deived from eg a rodent or primate.
  • CDR sequences of preferred human antibodies of the present invention are shown in SEQ ID Nos 49 to 78 in Table 1 on the following page. Most preferred antibodies are those having a CDR as shown in SEQ ID Nos
  • nucleotide molecules encoding the preferred antibodies of the present invention also form part of the present invention. Additionally, nucleotide molecules encoding the CDR of any of SEQ ID NOs 49 to 78, specifically 49 to 66, also form part of the present invention, together with expression vectors comprising the nucleic acid molecules.
  • the nucleotide molecules may be DNA or RNA.
  • the present invention also provides host cells and stable cell-lines containing the vectors herein described.
  • expression vector is meant one which is capable, in an appropriate host, of expressing apolypeptide encoded by the nucleic acid.
  • vectors are plasmids, such as bacterial, viral, mammalian and yeast vector plasmids.
  • Viral vectors include retroviral and adeno viral vectors.
  • Typical prokaryotic vector plasmids are pUC18, pUC19, pBR322 and pBR329 available from Biorad Laboratories (Richmond, CA, USA) and pTrc99A and pKK223-3 available from Pharmacia (Piscataway, NJ, USA).
  • a typical mammalian cell vector plasmid is pSVL available from Pharmacia, Piscataway, NJ, USA.
  • Useful yeast cell plasmid vectors are pRS403-406 and pRS413-416, generally available from Statagenen Cloning Systems, La Jolla, CA, USA Many expression systems are known, including bacterial (e.g E. Coli and
  • Bacillus subtilis Bacillus subtilis
  • yeasts e.g. Saccharomyces cerevisiae
  • filamentous fungi e.g. Aspergillus
  • plant cells animal cells and insect cells.
  • Typical bacterial host cells are cells from a strain of E. Coli, such as HB101F. ' Typical mammalian cells are COS7, NSO and CHO cells, generally available, as well as HEK293EBNA (Invitrogen AB) and PERC.6 (Crucell, the Netherlands).
  • the present invention also provides a method of producing the antibodies as described herein.
  • a fully human monoclonal antibody that recognises the region 12-35 of human ApoCIII is ATH1C3-1.
  • Fully human monoclonal antibodies that recognise the region 45-65 of human ApoCIII are ATH3-2 and ATH3-4.
  • Fully human monoclonal antibodies that recognise the region 1-76 of human ApoCIII are the antibodies mentioned above, together with: ATH1C23-4 and ATH1C23-21, which recognise epitopes which do not fall within the sequences 12-35 and 45-65.
  • the present invention also provides other antibodies that have a similar amino acid sequence in its hypervariable regions, and or similar CDR, so that the antibody is capable of competing with the fully human antibody for binding to ApoCIII.
  • a competition assay at least 40%, preferably at least 50, 60, 70, 80 or 90%, of the "similar" antibodies will bind to ApoCIII in the presence of antibodies with non- modified CDRs.
  • the CDRs of an antibody according to the invention are the light chain CDRs LI to L3 and the heavy chain CDRs HI to H3 as described in any of the SEQ ID Nos 49 to 78.
  • the amino acid sequences of these CDRs may be modified, however.
  • the amino acid sequence of each CDR may be modified by amino acid substitutions, insertions and/or deletions as described below.
  • Each CDR may therefore include one or two amino acid substitutions, insertions and/or deletions.
  • There may be up to three amino acid substitutions, insertions and/or deletions in light chain CDRL3 or heavy chain CDRH3.
  • Up to four amino acid substitutions, insertions and/or deletions may be present in light chain CDRL1.
  • Up to six amino acid substitutions, insertions and/or deletions may be present in heavy chain CDRH2.
  • the amino acid sequence of each CDR is substantially homologous to that of each CDR set out above.
  • the degree of sequence identity is at least 50% and more preferably it is at least 75%. Sequence identities of at least 90, 91, 92, 93, 94, 95, 96, 97, 98 or 99% are most preferred.
  • amino acids may often be substituted for other amino acids which have similar properties without substantially altering or adversely affecting certain properties of a protein. These are sometimes referred to as "conservative" amino acid changes.
  • amino acids glycine, valine, leucine or isoleucine can often be substituted for one another.
  • amino acids which can often be substituted for one another include: phenylalanine, tyrosine and tryptophan (amino acids having aromatic side chains); lysine, arginine and histidine (amino acids having basic side chains); aspartate and glutamate (amino acids having acidic side chains); asparagine and glutamine (amino acids having amide side chains) and cysteine and methionine (amino acids having sulphur containing side chains).
  • derivative can also include a variant of an amino acid sequence comprising one or more such "conservative" changes relative to said sequence.
  • antibody herein is used to refer to a molecule having a useful antigen binding specificity, ie will recognise and bind to ApoCIII. Those skilled in the art will readily appreciate that this term may also cover polypeptides which are fragments of or derivatives of antibodies yet which can show the same or a closely similar functionality. Such antibody fragments or derivatives are intended to be encompassed by the term antibody as used herein. Examples of such antibody fragments or derivatives are single chain Fv molecules (scFv), Fab, single domain antibodies(dAbs), F(ab') 2 , Fv and other fragments that retain the antigen-binding site.
  • scFv single chain Fv molecules
  • Fab single domain antibodies(dAbs)
  • F(ab') 2 single domain antibodies
  • monoclonal antibody is used herein to encompass any isolated antibodies such as conventional monoclonal antibody hybridomas, but also to encompass isolated monospecific antibodies produced by any cell, such as for example a sample of identical human immunoglobulins expressed in a mammalian cell line.
  • the monoclonal antibodies of the present invention are capable of being used in passive prophylaxis or therapy, by administration of the antibodies into a patient, for the amelioration of atherogenic disease.
  • the monoclonal antibodies of the present invention may be generated by screening a phage-display library to produce a scFv sequence which corresponds to a CDR and producing fully human IgG antibodies having this CDR region using known techniques, eg. as described in WO01/75091.
  • Hybridomas secreting the monoclonal antibodies of the present invention are also provided.
  • these hybridomas are of eukaryotic or insect origin.
  • the hybridomas are eukaryotic, in order to reduce the presence of endotoxin and also to provide for appropriate eukaryotic processing of the antibodies.
  • compositions comprising the antibodies, described above, also form an aspect of the present invention. Also provided are the use of the antibodies in medicine, and in the manufacture of medicaments for the treatment of atherosclerosis .
  • the administration of the antibodies of the present invention will be administered (preferably intra-venously) to the patients in need thereof.
  • the frequency of administration may be determined clinically by following the decline of antibody titres in the serum of patients over time, but in any event may be at a frequency of 1 to 52 times per year, and most preferably between 1 and 12 times per year.
  • Quantities of antibody may vary according to the severity of the disease, or half- life of the antibody in the serum, but preferably will be in the range of 1 to 10 g/kg of patient, and preferably within the range of 1 to 5 mg/kg of patient, and most preferably 1 to 2 mg/kg of patient.
  • the immunogens, immunogenic compositions, vaccines or monoclonal antibodies of the present invention may be administered to a patient who is suffering from, or is at risk to, atherosclerotic disease, and are effective in re-establishing the correct equilibrium of the "bad" lipoproteins (apo B containing lipoproteins) to the "good " lipoproteins (apo A-I containing lipoproteins) balance, and minimise the circulation time of apo B containing lipoproteins.
  • the inventors believe that these functions minimise the possibility of deposit and oxidation of apo B containing lipoproteins within the blood vessel walls, and hence, reduce the risk of atherosclerotic plaque formation or growth.
  • the antibodies are administered to a patient who is considered to be at high risk of developing atherosclerotic disease, at an early time point before disease is fully, or partially, established.
  • the present invention therefore, provides the use of the anti- ApoCIII monoclonal antibodies of the present invention, as defined above, in the manufacture of pharmaceutical compositions for the prophylaxis or therapy of atherosclerosis. Accordingly, the anti- ApoCIII monoclonal antibodies of the present invention are provided for use in medicine, and in the medical treatment or prophylaxis of atherosclerosis. There is also provided a method of treatment or prophylaxis of atherosclerosis comprising the administration to a patient suffering from or susceptible to atherosclerosis, of an antibody of the present invention.
  • a method of prophylaxis or treatment of atherosclerosis comprises a reduction of total circulating triglyceride levels in a patient, by the administration of an antibody of the present invention to the patient.
  • a method of reducing the amount of circulating VLDL and LDL in a patient, by the administration of the antibodies of the present invention to the patient comprises reducing the amount of circulating VLDL and LDL in a patient, by the administration of the antibodies of the present invention to the patient.
  • the average circulation time of ApoB containing lipoproteins may be investigated in an in vivo animal model by the measuring the clearance rate of labelled ApoB containing lipoproteins from the plasma of the mammal (half-life of labelled ApoB containing lipoproteins).
  • a preferred antibody for these method of treatment aspects of the present invention recognises any one of the ApoCIII epitopes SEQ ID NO: 1-48.
  • a particularly preferred antibody has a CDR of any of SEQ ID No 49 to 78, or a modification thereof as described herein.
  • Particularly preferred antibodies have CDRs 49 to 54, 55 to 60, 61 to 66, 67 to 72, or 73 to 78, as shown for ATH1C23- 4.MH, ATH1C23-21.MH, ATH32.MH, ATH34.MH, ATH1C3-1.MH, in Table 1.
  • human antibodies which recognise any ApoCIII molecule, or fragments, mutants, homologues, analogues or chemically or biologically modified versions thereof are also included.
  • targetting of ApoCIII may be used to downregulate the negative effects of the "bad” cholesterol (LDL), whilst not having a negative effect on the "good” cholesterol (HDL).
  • Preferred antibody isotypes of the present invention are IgGl and IgG4.
  • IgG4 isotypes are particularly preferred because they are thought to have lower affinity for Fc ⁇ receptors and thus be less efficient in mediating antibody- dependent complement-mediated cytolysis (ADCC; Adair hrrmunol Rev 1992;5-
  • Preferred methods of treating individuals suffering from Atherosclerosis having elevated levels of circulating ApoCIII in their plasma comprise reducing the levels of circulating ApoCIII, by the administration of a monoclonal Ab that is capable of blocking the activity of ApoCIII, by binding to the epitope of any of SEQ
  • Also provided by the present invention is a method of treatment or prophylaxis of atherosclerosis by reducing the number of ApoCIII molecules which are associated with an ApoB molecule in situ in the context of a lipoprotein by administration of a monoclonal antibody of the present invention.
  • a monoclonal antibody of the present invention In a normal individual there is approximately one ApoB present in an LDL particle, the ApoB being associated with between 1-5 ApoCIII molecules. In diseased individuals the number of ApoCIII molecules may increase to up to 25.
  • a method of treatment or prophylaxis of atherosclerosis by reducing the ratio of ApoCIII molecules per ApoB molecules in the LDL in an individual with atherosclerosis from a high disease state level (approximately 20 to 25:1) to a reduced therapeutic level preferably below 15:1, more preferably below 10:1 and more preferably below 5:1, preferably below 3:1, and most preferably approximately 1 : 1 ApoC:ApoB.
  • Levels of ApoCIII contained within ApoB-containing lipoproteins may be measured by nephelometry or electro-immunodiffusion (normal range is 2 to 3 mg/dL).
  • a combination therapy for treatment or prophylaxis of atherosclerosis comprising the passive immunotherapy of the present invention in combination with any other therapy or combination of therapies for treatment or prophylaxis of atherosclerosis, such as immunotherapy directed towards modified ApoCIII, oxidised ApoA, oxidised ApoB (as described in WO02/080954), oxidised LDL (WO02/50550) or cholesterol ester transfer protein (CETP; W099/15655), or other known therapies.
  • immunotherapy directed towards modified ApoCIII, oxidised ApoA, oxidised ApoB (as described in WO02/080954), oxidised LDL (WO02/50550) or cholesterol ester transfer protein (CETP; W099/15655) or other known therapies.
  • the ApoCIII peptides (1-79, 12-35 and 45-65) were synthesised by the solid phase method (Merrifield, 1986) on an automated synthesiser Model ABI 433A (Applied Biosystems Inc.) using Boc/Bzl strategy on a Boc-Ala-PAM resin for total ApoCIII and MBHA resin for the others fragments.
  • Other ApoCIII peptides may be synthesised according to the same method. Each amino acid was coupled twice by dicyclohexylcarbodiimide/hydroxybenzotriazole without capping.
  • the reaction vessel was recharged with p-cresol (0.75 g), p-thiocresol (0.25 g) and 10 ml of anhydrous HF and the mixture was allowed to react at 0°C for 1.5 h. Hydrogen fluoride was removed by evaporation and the residue was triturated with diethyl ether. The residue was filtered off, washed with diethyl ether and extracted with 200 ml of 10% aqueous acetic acid and lyophilised.
  • Peptides synthesised as described in Example 1 may be used to select and screen e.g. the n-CoDeRTM scFv library to identify scFv fragments, which are capable of binding to the peptides.
  • Selection of scFv or Fab phage display libraries can be performed in many different ways, employing various techniques to extract target binding phages, e.g immobilisation of target antigens to solid surfaces or capture of biotinylated target antigens on streptavidin coated magnetic beads such as Dynabeads (Dynal, Norway)
  • peptides (1-79), (12-35) and (45-65) were used to perform three consecutive selections against the target peptides of Example 1 , as described below
  • Selection 1 For each of the 3 target peptides approximately 10 13 n- CoDeRTM phages in PBS containing 3% BSA, 0.05% Tween 20 and 0.02% sodium azide, were incubated for 1 h withlxlO "7 M of the 3 different biotinylated target peptides in 1.8 ml. Biotinylated target antigens were then captured on streptavidin coated magnetic Dynabeads. Non-specific phages were removed by washing with the buffer described above. Bound phages were eluted with trypsin digestion and used to infect Escherichia coli HB101F' for phage amplification before selection 2.
  • Selection 2 The amplified phage pool from selection on peptide(l-79) was divided into two parts and used for selection on a) 2xl0 "8 M biotinylated peptide (1-79) in 1 ml buffer, as above. In addition, lxlO "7 M of non-biotinylated peptide (12-35) and peptide (45-65) were used for counter-selection, thereby increasing the probability to find fragments binding to other regions of peptide (1-79) than regions (12-35) nor (45-65).
  • Selection 3 Selection was performed on non-biotinylated target antigens immobilised in microtiter plate wells, 2 pmole/well. Each selections were performed in 100 ⁇ l in 8 microtiter wells per target.
  • the amplified phage pool from selection on peptide (1-79) was divided into three parts and used for selection on a) peptide (1-79) with counter-selection on lxlO "6 M biotinylated peptide (12-35) captured on Dynabead and competition by lxlO "6 M non-biotinylated peptide (12-35).
  • Candidate target binding scFv clones were then identified in a screening process briefly described below:
  • the selected phage pools from selection 3 were then converted to scFv format by enzymatic cleavage of the phagemid, deleting the phage gene III .
  • the resulting plasmids encoding soluble scFv were transformed into E.coli. Single bacterial colonies were picked for growth in LB-medium, and expression of soluble scFv was induced by addition of IPTG.
  • the resulting scFv stocks were assayed for binding to the biotinylated peptides, loaded in avidin pre-coated wells, in Luminescence ELISA. Positive clones were further assayed in Luminescence ELISA against the target peptides, directly coated to the wells.
  • the chosen clones may preferably be converted to IgGl or IgG4 format using techniques well known in the art, for example as described in Henderikx et al., (2002; Am. J. Pathol. 160, 1597-1608).
  • the clones may be transferred to a IgGl or IgG4 vector and the functionality of the clones tested by transient expression in, for example, Cos 7 cells.
  • Functional clones may then be transfected into NSO cells and bulk pools expanded.

Abstract

The present invention relates to novel therapies and treatments of atherosclerotic diseases. Accordingly there is provided, methods of treating or preventing atherosclerosis by passive vaccination through administration to a patient of a human antibody that is capable of binding to the specific fragments of ApoCIII. Specific human monoclonal antibodies and their use in therapy of atherosclerosis is provided. There is further provided the use of the antibodies of the present invention in medicine.

Description

Novel composition
The present invention relates to novel therapies, and prophylactic treatments of dyslipidaemia, such as atherosclerotic diseases. Accordingly there is provided, methods of treating or preventing atherosclerosis by passive vaccination through administration to a patient of antibodies that are capable of binding to specific epitopes of Apolipoprotein C-III (ApoCIII). The antibodies of the present invention are potent in the prevention, or reduction, of atherosclerotic plaque formation over prolonged periods of time, thereby reducing the potential of atheroslerosis leading to coronary or cerebrovascular disease. There is further provided the use of the antibodies of the present invention in medicine.
Preferred epitopes of ApoCIII which consist of the targets for the passive immunotherapy aspects of the present invention, are encompased within the regions between amino acid numbers 12-35 and between amino acid numbers 45-76 (particuarly 45-65) of the mature form of human ApoCIII, although other regions of ApoCIII may also be targeted by the passive immunotherapy of the present invention. Atherosclerosis is the leading cause of death and disability in the developed world, and is the major cause of coronary and cerebrovascular deaths, with approximately 7.2 and 4.6 million deaths per year worldwide respectively (Atherosclerosis is generally described in Harrison's Principles of Internal Medicine (14th Edition, McGraw Hill, pl345-1352), Berliner, J. et al., 1995, Circulation, 91:2488-2496; Ross, R., 1993; Nature, 362:801). The name in Greek refers to the thickening (sclerosis) of the arterial intima and accumulation of lipid (athere) in lesions.
Although many generalised or systemic risk factors predispose to its development, such as a high cholesterol diet and smoking, this disease may affect different distinct regions of the circulation. For example, atherosclerosis of the coronary arteries commonly causes angina pectoris and myocardial infarction. Whilst, atherosclerosis of the arteries supplying the central nervous system frequently provokes transient cerebral ischemia and strokes. In the peripheral circulation, atherosclerosis can cause intermittent claudication and gangrene and can jeopardise limb viability. Involvement of the splanchnic circulation can cause mesenteric ischemia and bowel infarction. Atherosclerosis can affect the kidney directly (eg causing renal artery stenosis), and in addition, the kidney is a frequent site of atheroembolic disease.
Atherogenesis in humans typically occurs over many years, usually many decades. The slow build up of atherogenic plaques in the lining of the vasculature can lead to chronic clinical expressions through blood flow restriction (such as stable effort-induced angina pectoris or predictable and reproducible intermittent claudication). Alternatively, a much more dramatic acute clinical event, such as a myocardial infarction or cerebrovascular accident can occur after plaque rupture. The way in which atherosclerosis affects an arterial segment also varies, an additional feature of the heterogeneity and complexity of this disease. Atheromas are usually thought of as stenotic lesions, or plaques, which can limit blood flow, however, atherosclerosis can also cause ectasia and development of aneurysmal disease with an increase in lumen caliber. This expression of atherosclerosis frequently occurs in the aorta, creating a predisposition to rupture or dissection rather than to stenosis or occlusion.
The genesis of atherogenic plaques has been studied in depth. In normal human adults, the intimal layer of arteries contains some resident smooth muscle cells embedded in extracellular matrix and is covered with a monolayer of vascular endothelial cells. Initial stages of atherogenesis involve the development of "fatty streaks" in the walls of the blood vessel resulting from accumulation and deposit of lipoproteins in regions of the intimal layer of the artery. Low-density lipoprotein (LDL) particles, rich in cholesterol, is an example of an atherogenic lipoprotein which is capable of deposition in the vessel walls to form such fatty streaks.
Once deposited within the vessel wall, the lipoprotein particles undergo chemical modification, including both oxidation and non-enzymatic glycation. These oxidised and glycated lipoproteins then contribute to many of the subsequent events of lesion development. The chemical modifications attract macrophages within the vessel walls, which internalise the oxidised LDL and become foam cells which initiate lesions called plaques. It is the atherosclerotic plaques which are responsible for the clinical manifestations of atherosclerosis, either they limit blood flow, or allow aneurism, or may even rupture provoking the coronary or cerebrovascular attacks. The development of atherosclerosis is a long process which may occur over decades, which is initiated by an imbalance between atherogenic and protective lipoproteins. For example, cholesterol associated with high-density lipoproteins or HDL (so called "good" cholesterol) and low-density lipoproteins or LDL (so called "bad" cholesterol) levels in the circulation are thought to be markers of increased probability of atherosclerosis (Harrison's Principles of Internal Medicine (14th Edition, McGraw Hill, pl345-1352)). Cholesterol, cholesterol esters, triacylglycerols and other lipids are transported in body fluids by a series of lipoproteins classified according to their increasing density: chylomicrons, Very Low, Low, Intermediate and High density lipoproteins (CM, VLDL, LDL, IDL and HDL respectively). These lipoprotein-complexes consist of a core of hydrophobic lipids surrounded by polar lipids and then by a shell of Apolipoproteins. Currently, there are at least twelve types of apolipoproteins known, A-I, A-II, A-IV, A-V, B, CI, CII, CIII, D, E, H and J. There are at least two functions of these apolipoproteins which are common to all lipoprotein complexes, first they are responsible for the solubilisation of the hydrophobic lipid cores that they carry, and second they are also involved in the regulation of cholesterol lipoprotein uptake by specific cells. The different types of lipoproteins may have different functions, for example LDL (which are rich in cholesterol esters) are thought to be associated with the transport of cholesterol to peripheral tissues for new membrane synthesis.
One of these apolipoproteins, apolipoprotein C-III (ApoCIII), is a 79 amino acid protein produced in the liver and intestine (Brewer et al., J. Biol. Chem. (1974), 249 : 4975-4984; Protter, A.A., et al., 1984, DNA, 3:449-456; Fruchart, J.C. et al,
1996, Drugs Affecting Lipid Metabolism, (Eds. Gotto, A.M. et al.), Kluwer Academic Publishers and Fordazione Giovanni Lorenzini, Netherlands, p631-638; Claveny, V. et al., Arteriosclerosis, Thrombosis and Vascular Biology, 15, 7, 963-971; US patent No. 4,801,531; McConathy, W.J. et al. 1992, Journal of Lipid Research, 33, 995- 1003). ApoCIII is a component of CM, VLDL, LDL (Lenich et al., C, J. Lip. Res. (1988) 29, 755-764), and also HDL, and exists as three isoforms : ApoCIIIO, ApoCIII 1 and ApoCIII2. ApoCIIIO is not glycosylated, however ApoCIII 1 and ApoCIII2 are glycosylated and have respectively one and two sialic acid residues (Ito et al, 1989 J.lipd. Res. Nov 30:11 1781-1787). The sugar moiety consists of disaccharide β-D galactosyl (1-3) α-N-Acetyl-D-Galactosamine attached to threonine 74 of protein chain by O-glycosidic binding (Assman et al, 1989, BBA 541:234-240). In human normolipidemic plasma ApoCIIIO, ApoCIII 1 and ApoCIII2 represent 14%, 59% and 27% of total ApoCIII respectively. Mutagenesis of the glycosylation site of human ApoCIII does not affect its secretion and lipid binding (Roghani et al., 1988 JBC 34:17925-32).
Mature Human ApoCIII has the following ammo acid sequence: tSEAEDASLLSFMQGYMKHATKTAKDALSSVQESQVAQQARGWVTDGFSSL KDYWSTVKDKFSEFWDLDPEVRPTSAVAA79 (SEQ ID.NO. 1).
Plasma concentration of ApoCiπ is positively correlated with levels of plasma triglycerides (Schonfeld et al, Metabolism (1979) 28 : 1001-1010; Kaslyap et al, J. Lip. Res. (1981) 22 : 800-810). Liver perfusion studies demonstrate that ApoCIII inhibits the hepatic uptake of triglyceride-rich lipoproteins (TRL) and their remnants (Shelburne et al, J. Clin. Inves., (1980) 65 : 652-658, Windier et al, J. Lip. Res.
(1985) 26 : 556-563). Also in vitro experiments show that ApoCIII inhibit the activity of both lipoprotein lipase (LPL) and hepatic lipase (Brown and Bakinsky, Biochim. Biophs. Acta. (1972) 46 : 375-382; Krauss et al, Circ. Res. (1973) 33 : 403-411; Wang et al, J. Clin. Inves. (1985) 75 : 384-390; Mc Conathy et al, J. Lip. Res. (1972) 33 : 995-1003; Kinnemen and Enholm, FEBS (1976) 65 : 354-357). Moreover, ApoCIII is said to be involved in inhibition of LDL binding to LDL receptors (Fruchart et al. supra), via ApoB.
The role of ApoCIII in plasma TRL metabolism has been more defined by the results of recent studies in transgenic animals (Aalto-Setala et al, J. Clin. Invest. (1992) 90:5 1889-1900.). Plasma accumulation of TRL in mice overexpressing ApoCIII has been shown to be associated with reduced plasma VLDL and chylomicron clearance (Harrold et al, J. Lip. Res. (1996) 37 : 754-760) also the inhibitory effect of C apolipoproteins on the LDL receptor of apo B-containing lipoproteins was demonstrated (Clavey et al, Arth. Thromb. and Vase. Biol. (1995) 15 : 963-971).
Previously, vaccines in the field of immunotherapy of atherosclerosis have focused on the use of cholesterol as an immunogen to reduce serum cholesterol levels (Bailey, J.M. et al, 1994, Biochemical Society Transactions, 22, 433S; Alving, C. and Swartz, G.M., 1991, Crit. Rev. Immunol., 10, 441-453; Alving, C. and Wassef, N.M., 1999, Immunology Today, 20, 8, 362-366). Others have attempted to alter the activity of the Cholesterol Ester Transfer Protein (CETP) by vaccination (WO 99/15655).
Alternatively, some authors have described vaccines using oxidised LDL as the immunogen, in order to inhibit plaque formation after balloon injury in hypercholesterolemic rabbits (Nilsson, J. et al, 1997, JACC, 30, 7, 1886-1891). It has been found, surprisingly, that atherosclerosis may be prevented or ameliorated by passive immunotherapy, by reducing or blocking the function of ApoCIII. In particular, the passive immunotherapies of the present invention can be advantageously carried out using specific human antibodies which target epitopes of ApoCIII. The use of the specific antibodies against ApoCIII can focus the immune response to parts of the human ApoCIII molecule without triggering a general response to the whole molecule. Without wishing to be bound by theory, this can be used as a means of distinguishing parts of ApoCIII that are surface exposed on LDL and not HDL, thus focusing the immune response against carriers of "bad cholesterol", whilst not affecting the positive role of ApoCIII in HDL. It will be appreciated that ApoCIII can exist in different physiological forms, for example, oxidised and non-oxidised forms, and that allelic variants and mutants of ApoCIII may exist. The antibodies of the present invention may recognise any of these forms of ApoCIII. It is a preferred embodiment that the antibodies recognise ApoCIII having the sequence of SEQ ID No 1, or comprising one or more of the sequences as set out in any of SEQ ID Nos 2-48. In particular, these antibodies will recognise the non-oxidised form of ApoCIII.
The passive immunotherapies of the present invention target an epitope found within the region between amino acid number 1 and 79, or more preferably an epitope found within the region between amino acid number 1 and 17, 12 and 35, or an epitope found within the region between amino acids 45 and 76 of the human ApoCIII molecule as it exists in the circulation of a human, in addition it is preferred that the immunotherapy targets the epitope that is found within the region between amino acid 12 to 21 or 45 to 65 of human ApoCIII. The sequence of the region between amino acid number 12 and 35 of the human ApoCIII is as follows:
MQGYMKHATKTAKDALSSVQESQV (SEQ ID NO.2). The sequence of the region between amino acid number 12 and 21 of the human ApoCIII is as follows: MQGYMKHATK (SEQ ID NO.3)
The sequence of the region between amino acid number 45 and 76 of the human ApoCIII is as follows:
DGFSSLKDYWSTVKDKFSEFWDLDPEVRPTSA (SEQ ID NO: 4) The sequence of the region between amino acid number 45 and 65 of the human ApoCIII is as follows:
DGFSSLKDYWSTVKDKFSEFW (SEQ ID NO: 5) The present invention also provides the following fragments of the above peptides within which contain an epitope of ApoCIII which may be targeted by the passive immunotherapies of the present invention:
Figure imgf000007_0001
Figure imgf000008_0001
The sequence of the region between amino acid number 1 and 17 of the human ApoCIII is as follows: iSEAEDASLLSFMQGYMKπ (SEQ ID NO: 48)
The present invention provides antibodies effective in the prophylaxis or therapy of dyslipidaemia or atherosclerosis which target the epitopes listed in SEQ ID NO.s 1-48, of ApoCIII, and also provides for methods of treatment of atherosclerosis by passive administration of the antibodies of the present invention to individuals in need thereof. Most preferably the antibodies of the invention recognise the epitopes listed in SEQ ID NO: 1, 3, 6-22.
Most preferably the antibodies of the present invention are functional in the treatment of atherosclerosis, and in a preferred form of the present invention they abrogate the inhibition exerted by ApoCIII on the binding of ApoB to its receptor, and/or the activity of lipoprotein lipase. Such activities may readily be assayed by the man skilled in the art for example by methods described in Fruchard et al, supra; and McConathy et al., supra. The antibodies of the present invention are provided for use in medicine, and for use in the treatment or prevention of atherosclerosis.
The antibodies of the present invention will be generally administered for both initial and boosting doses. It is expected that the boosting doses will be adequately spaced, or preferably given at such times where the levels of circulating antibody fall below a desired level.
The antibody preparations of the present invention may be used to protect or treat a mammal susceptible to, or suffering from atherosclerosis, by means of administering said antibodies via a systemic route. These administrations may include injection via the intramuscular, intraperitoneal, intradermal or subcutaneous routes. The antibodies will preferably be administered in a pharmaceutical composition, together with a pharmaceutically acceptable carrier.
In one aspect of the present invention are provided fully human monoclonal antibodies capable of binding to epitopes of SEQ ID NO.s 1 to 48 (preferably SEQ ID NO: 1, 2 or 3) in the context of the human ApoCIII molecule, and their use in immunotherapy.
Antibodies typically comprise two heavy chains linked together by disulphide bonds and two light chains. Each light chain is linked to a respective heavy chain by disulphide bonds. Each heavy chain has at one end a variable domain followed by a number of constant domains. Each light chain has a variable region at one end and a constant domain at its other end. The light chain variable domain is aligned with the variable domain of the heavy chain. The light chain constant domain is aligned with the first constant domain of the heavy chain.
The constant domains in the light and heavy chains are not involved directly in binding the antibody to antigen. The variable domains in each pair of light and heavy chains form the antigen binding site. The domains on the light and heavy chains have the same general structure and each domain comprises a framework of four regions, whose sequences are relatively conserved,connected by three complementarity determining regions (CDRs). The four framework regions largely adopt a beta sheet conformation and the CDRs form loops connecting, and in some cases forming part of, the beta-sheet structure. The CDRs are held in close proximity by the framework regions and, with the CDRs from the other domain, contribute to the formation of the antibody binding site. The preparation of altered antibodies in which the variable region of a rodent antibody is combined with the constant region of a human antibody is now well known in the the art (Oi and Morrison 1986 Biotechniques 4, 214-212). Humanised antibodies in which the CDRs are derived from a source different from that of the framework of the antibody's variable domains are disclosed in EP-A-0239400. The CDRs may be derived from a rodent or primate monoclonal antibody, or by screening a human phage-display library using known techniques (eg WO01/75091, W098/32845). If the CDRs are obtained from a non-human source, and are introduced into the scaffold of a human antibody, such hybrid antibodies are known "humanised" antibodies. If the CDR regions are obtained by use of a human phage display library, and are introduced into the scaffold of a human antibody, then the resulting antibodies are fully human. Preferably, antibodies of the present invention are fully human antibodies. The antibodies should not be recognisable by the host human immune system as foreign, or should elicit a far reduced immune response when administered to a human than the immune response mounted by a human against a humanised or a chimaeric antibody which has regions deived from eg a rodent or primate.
The CDR sequences of preferred human antibodies of the present invention are shown in SEQ ID Nos 49 to 78 in Table 1 on the following page. Most preferred antibodies are those having a CDR as shown in SEQ ID Nos
49 to 66. Nucleotide molecules encoding the preferred antibodies of the present invention also form part of the present invention. Additionally, nucleotide molecules encoding the CDR of any of SEQ ID NOs 49 to 78, specifically 49 to 66, also form part of the present invention, together with expression vectors comprising the nucleic acid molecules. The nucleotide molecules may be DNA or RNA. The present invention also provides host cells and stable cell-lines containing the vectors herein described.
By "expression vector" is meant one which is capable, in an appropriate host, of expressing apolypeptide encoded by the nucleic acid. Examples of vectors are plasmids, such as bacterial, viral, mammalian and yeast vector plasmids. Viral vectors include retroviral and adeno viral vectors.
Typical prokaryotic vector plasmids are pUC18, pUC19, pBR322 and pBR329 available from Biorad Laboratories (Richmond, CA, USA) and pTrc99A and pKK223-3 available from Pharmacia (Piscataway, NJ, USA). A typical mammalian cell vector plasmid is pSVL available from Pharmacia, Piscataway, NJ, USA.
Useful yeast cell plasmid vectors are pRS403-406 and pRS413-416, generally available from Statagenen Cloning Systems, La Jolla, CA, USA Many expression systems are known, including bacterial (e.g E. Coli and
Bacillus subtilis), yeasts (e.g. Saccharomyces cerevisiae), filamentous fungi (e.g. Aspergillus), plant cells, animal cells and insect cells.
Typical bacterial host cells are cells from a strain of E. Coli, such as HB101F. ' Typical mammalian cells are COS7, NSO and CHO cells, generally available, as well as HEK293EBNA (Invitrogen AB) and PERC.6 (Crucell, the Netherlands).
The present invention also provides a method of producing the antibodies as described herein.
Table 1
Figure imgf000012_0001
A fully human monoclonal antibody that recognises the region 12-35 of human ApoCIII is ATH1C3-1. Fully human monoclonal antibodies that recognise the region 45-65 of human ApoCIII are ATH3-2 and ATH3-4. Fully human monoclonal antibodies that recognise the region 1-76 of human ApoCIII are the antibodies mentioned above, together with: ATH1C23-4 and ATH1C23-21, which recognise epitopes which do not fall within the sequences 12-35 and 45-65.
The sequences of the hypervariable regions and the complementarity determining regions (CDRs) of the fully human antibodies of the present invention are fully encompassed within the present invention.
Also encompassed within the scope of the present invention are "similar" human antibodies to the above identified monoclonal antibodies. For example, the present invention also provides other antibodies that have a similar amino acid sequence in its hypervariable regions, and or similar CDR, so that the antibody is capable of competing with the fully human antibody for binding to ApoCIII. Thus, in a competition assay at least 40%, preferably at least 50, 60, 70, 80 or 90%, of the "similar" antibodies will bind to ApoCIII in the presence of antibodies with non- modified CDRs. Suitably, the CDRs of an antibody according to the invention are the light chain CDRs LI to L3 and the heavy chain CDRs HI to H3 as described in any of the SEQ ID Nos 49 to 78.
The amino acid sequences of these CDRs may be modified, however. The amino acid sequence of each CDR may be modified by amino acid substitutions, insertions and/or deletions as described below. Each CDR may therefore include one or two amino acid substitutions, insertions and/or deletions. There may be up to three amino acid substitutions, insertions and/or deletions in light chain CDRL3 or heavy chain CDRH3. Up to four amino acid substitutions, insertions and/or deletions may be present in light chain CDRL1. Up to six amino acid substitutions, insertions and/or deletions may be present in heavy chain CDRH2. Preferably the amino acid sequence of each CDR is substantially homologous to that of each CDR set out above. Preferably the degree of sequence identity is at least 50% and more preferably it is at least 75%. Sequence identities of at least 90, 91, 92, 93, 94, 95, 96, 97, 98 or 99% are most preferred.
It will nevertheless be appreciated by the skilled person that high degrees of sequence identity are not necessarily required since various amino acids may often be substituted for other amino acids which have similar properties without substantially altering or adversely affecting certain properties of a protein. These are sometimes referred to as "conservative" amino acid changes. Thus the amino acids glycine, valine, leucine or isoleucine can often be substituted for one another. Other amino acids which can often be substituted for one another include: phenylalanine, tyrosine and tryptophan (amino acids having aromatic side chains); lysine, arginine and histidine (amino acids having basic side chains); aspartate and glutamate (amino acids having acidic side chains); asparagine and glutamine (amino acids having amide side chains) and cysteine and methionine (amino acids having sulphur containing side chains). Thus the term "derivative" can also include a variant of an amino acid sequence comprising one or more such "conservative" changes relative to said sequence.
The term "antibody" herein is used to refer to a molecule having a useful antigen binding specificity, ie will recognise and bind to ApoCIII. Those skilled in the art will readily appreciate that this term may also cover polypeptides which are fragments of or derivatives of antibodies yet which can show the same or a closely similar functionality. Such antibody fragments or derivatives are intended to be encompassed by the term antibody as used herein. Examples of such antibody fragments or derivatives are single chain Fv molecules (scFv), Fab, single domain antibodies(dAbs), F(ab')2, Fv and other fragments that retain the antigen-binding site.
The term "monoclonal antibody" is used herein to encompass any isolated antibodies such as conventional monoclonal antibody hybridomas, but also to encompass isolated monospecific antibodies produced by any cell, such as for example a sample of identical human immunoglobulins expressed in a mammalian cell line. The monoclonal antibodies of the present invention are capable of being used in passive prophylaxis or therapy, by administration of the antibodies into a patient, for the amelioration of atherogenic disease.
The monoclonal antibodies of the present invention may be generated by screening a phage-display library to produce a scFv sequence which corresponds to a CDR and producing fully human IgG antibodies having this CDR region using known techniques, eg. as described in WO01/75091.
Hybridomas secreting the monoclonal antibodies of the present invention are also provided. Preferably, these hybridomas are of eukaryotic or insect origin. Most preferably, the hybridomas are eukaryotic, in order to reduce the presence of endotoxin and also to provide for appropriate eukaryotic processing of the antibodies.
Pharmaceutical compositions comprising the antibodies, described above, also form an aspect of the present invention. Also provided are the use of the antibodies in medicine, and in the manufacture of medicaments for the treatment of atherosclerosis .
In the passive treatments of atherosclerosis as provided herein, the administration of the antibodies of the present invention will be administered (preferably intra-venously) to the patients in need thereof. The frequency of administration may be determined clinically by following the decline of antibody titres in the serum of patients over time, but in any event may be at a frequency of 1 to 52 times per year, and most preferably between 1 and 12 times per year. Quantities of antibody may vary according to the severity of the disease, or half- life of the antibody in the serum, but preferably will be in the range of 1 to 10 g/kg of patient, and preferably within the range of 1 to 5 mg/kg of patient, and most preferably 1 to 2 mg/kg of patient.
The immunogens, immunogenic compositions, vaccines or monoclonal antibodies of the present invention may be administered to a patient who is suffering from, or is at risk to, atherosclerotic disease, and are effective in re-establishing the correct equilibrium of the "bad" lipoproteins (apo B containing lipoproteins) to the "good " lipoproteins (apo A-I containing lipoproteins) balance, and minimise the circulation time of apo B containing lipoproteins. Not wishing to be bound by theory, the inventors believe that these functions minimise the possibility of deposit and oxidation of apo B containing lipoproteins within the blood vessel walls, and hence, reduce the risk of atherosclerotic plaque formation or growth. Preferably, the antibodies are administered to a patient who is considered to be at high risk of developing atherosclerotic disease, at an early time point before disease is fully, or partially, established.
The present invention, therefore, provides the use of the anti- ApoCIII monoclonal antibodies of the present invention, as defined above, in the manufacture of pharmaceutical compositions for the prophylaxis or therapy of atherosclerosis. Accordingly, the anti- ApoCIII monoclonal antibodies of the present invention are provided for use in medicine, and in the medical treatment or prophylaxis of atherosclerosis. There is also provided a method of treatment or prophylaxis of atherosclerosis comprising the administration to a patient suffering from or susceptible to atherosclerosis, of an antibody of the present invention.
A method of prophylaxis or treatment of atherosclerosis is provided which comprises a reduction of total circulating triglyceride levels in a patient, by the administration of an antibody of the present invention to the patient. In particular there is provided a method of reducing the amount of circulating VLDL and LDL in a patient, by the administration of the antibodies of the present invention to the patient.
Also provided is a method of prophylaxis or treatment of atherosclerosis by the administration to a patient of an antibody which is capable of reducing the average circulation time of ApoB containing lipoproteins. In tins regard the average circulation time of ApoB containing lipoproteins, may be investigated in an in vivo animal model by the measuring the clearance rate of labelled ApoB containing lipoproteins from the plasma of the mammal (half-life of labelled ApoB containing lipoproteins).
A preferred antibody for these method of treatment aspects of the present invention recognises any one of the ApoCIII epitopes SEQ ID NO: 1-48. A particularly preferred antibody has a CDR of any of SEQ ID No 49 to 78, or a modification thereof as described herein. Particularly preferred antibodies have CDRs 49 to 54, 55 to 60, 61 to 66, 67 to 72, or 73 to 78, as shown for ATH1C23- 4.MH, ATH1C23-21.MH, ATH32.MH, ATH34.MH, ATH1C3-1.MH, in Table 1. In addition, human antibodies which recognise any ApoCIII molecule, or fragments, mutants, homologues, analogues or chemically or biologically modified versions thereof are also included.
Surprisingly, targetting of ApoCIII may be used to downregulate the negative effects of the "bad" cholesterol (LDL), whilst not having a negative effect on the "good" cholesterol (HDL).
Preferred antibody isotypes of the present invention are IgGl and IgG4.
IgG4 isotypes are particularly preferred because they are thought to have lower affinity for Fcγreceptors and thus be less efficient in mediating antibody- dependent complement-mediated cytolysis (ADCC; Adair hrrmunol Rev 1992;5-
39.)
Preferred methods of treating individuals suffering from Atherosclerosis having elevated levels of circulating ApoCIII in their plasma comprise reducing the levels of circulating ApoCIII, by the administration of a monoclonal Ab that is capable of blocking the activity of ApoCIII, by binding to the epitope of any of SEQ
ID NO: 1-48 and thereby abrogating the ApoCIII-mediated inhibition of lipoprotein lipase and/or the binding of ApoB to its receptor, to said patient.
Also provided by the present invention is a method of treatment or prophylaxis of atherosclerosis by reducing the number of ApoCIII molecules which are associated with an ApoB molecule in situ in the context of a lipoprotein by administration of a monoclonal antibody of the present invention. In a normal individual there is approximately one ApoB present in an LDL particle, the ApoB being associated with between 1-5 ApoCIII molecules. In diseased individuals the number of ApoCIII molecules may increase to up to 25. Accordingly, there is provided by the present invention a method of treatment or prophylaxis of atherosclerosis by reducing the ratio of ApoCIII molecules per ApoB molecules in the LDL in an individual with atherosclerosis from a high disease state level (approximately 20 to 25:1) to a reduced therapeutic level preferably below 15:1, more preferably below 10:1 and more preferably below 5:1, preferably below 3:1, and most preferably approximately 1 : 1 ApoC:ApoB. Levels of ApoCIII contained within ApoB-containing lipoproteins may be measured by nephelometry or electro-immunodiffusion (normal range is 2 to 3 mg/dL).
Also provided by the present invention is a combination therapy for treatment or prophylaxis of atherosclerosis comprising the passive immunotherapy of the present invention in combination with any other therapy or combination of therapies for treatment or prophylaxis of atherosclerosis, such as immunotherapy directed towards modified ApoCIII, oxidised ApoA, oxidised ApoB (as described in WO02/080954), oxidised LDL (WO02/50550) or cholesterol ester transfer protein (CETP; W099/15655), or other known therapies.
The present invention is illustrated, but not limited, by the following examples:
EXAMPLES Example 1, Peptide synthesis
The ApoCIII peptides (1-79, 12-35 and 45-65) were synthesised by the solid phase method (Merrifield, 1986) on an automated synthesiser Model ABI 433A (Applied Biosystems Inc.) using Boc/Bzl strategy on a Boc-Ala-PAM resin for total ApoCIII and MBHA resin for the others fragments. Other ApoCIII peptides may be synthesised according to the same method. Each amino acid was coupled twice by dicyclohexylcarbodiimide/hydroxybenzotriazole without capping. Side chain protecting groups were as follows: Arg(Ts), Asp(Ochex), Glu(Ochex), Lys(2-Cl-Z), His(Dnp), Ser(Bzl), Thr(Bzl), Met(0)and Tyr(Br-Z). According to the sequence, the group Dnp on His was removed from the peptide, prior to the cleavage from its support by treatment with 10% β-mercaptoethanol, 5% diisopropylethylamine in
DCM for 2 h and in NMP for 2 h. The peptidyl resin was then treated with 50% TFA in DCM for 20 min to remove the amino-terminal Boc. The peptide was cleaved from the resin and simultaneously deprotected according to a low and high HF procedure: the resin (Ig) was treated with anhydrous HF (2.5 mL) in the presence of p-cresol (0.75 g), p-thiocresol (0.25 g) and dimethylsulfide (6.5 mL) at 0°C. After 3 h hydrogen fluoride and dimethylsulfide were removed by vacuum evaporation and the residual scavengers and by products were extracted with diethyl ether. The reaction vessel was recharged with p-cresol (0.75 g), p-thiocresol (0.25 g) and 10 ml of anhydrous HF and the mixture was allowed to react at 0°C for 1.5 h. Hydrogen fluoride was removed by evaporation and the residue was triturated with diethyl ether. The residue was filtered off, washed with diethyl ether and extracted with 200 ml of 10% aqueous acetic acid and lyophilised. The crude product was analysed by reversed-phase HPLC on a Vydac C18 column (4,6 x 250 mm, 5μ, 100 A) using 60 min linear gradient from 0 to 100% Buffer B (Buffer A: 0.05% TFA in H2O and Buffer B: 0.05% TFA, 60% CH3CN in H2O) at flow rate of 0.7 ml/min and detection was performed at 215 nm. Synthetic peptides were purified by RP-HPLC and were characterised and analysed by HPLC, the molecular mass determined by spectrometry. Example 2, Monoclonal Antibody production
Methods of production of recombinant antibodies, by screening phage display libraries, such as scFv or Fab libraries, are well known in the art (McCafferty et al 1990 Nature 348, 552-554; Barbas et al 1991 Proc.Natl.Acad.Sci. USA 88, 7978- 7982; Clarkson et al 191, Nature 352, 624-628; Soderlind et al 2000 Nature Biotech. 8, 852-856). The framework regions of antibodies derived from the n-CoDeR™ scFv library used in the present example are shown below. CDR regions (H1-H3 and Ll- L3) are indicated.
Heavy chain region EVQLLESGGGLVQPGGSLRLSCAASGFT— HI —KGLEWV— H2 —FTISRDN SKNTLYLQMNSLRAEDTAVYYC— H3 — WGQGTLVTVSS
Light chain region
QSVLTQPPSASGTPGQRVTIS— LI — WYQQLPGTAPKLLIY— L2 — GVP DRFSGSKSGTSASLAISGLRSEDEADYY— L3 — FGGGTKLTVLG
Peptides synthesised as described in Example 1 may be used to select and screen e.g. the n-CoDeR™ scFv library to identify scFv fragments, which are capable of binding to the peptides. Selection of scFv or Fab phage display libraries can be performed in many different ways, employing various techniques to extract target binding phages, e.g immobilisation of target antigens to solid surfaces or capture of biotinylated target antigens on streptavidin coated magnetic beads such as Dynabeads (Dynal, Norway)
Briefly, peptides (1-79), (12-35) and (45-65) were used to perform three consecutive selections against the target peptides of Example 1 , as described below
Selection 1: For each of the 3 target peptides approximately 1013 n- CoDeR™ phages in PBS containing 3% BSA, 0.05% Tween 20 and 0.02% sodium azide, were incubated for 1 h withlxlO"7 M of the 3 different biotinylated target peptides in 1.8 ml. Biotinylated target antigens were then captured on streptavidin coated magnetic Dynabeads. Non-specific phages were removed by washing with the buffer described above. Bound phages were eluted with trypsin digestion and used to infect Escherichia coli HB101F' for phage amplification before selection 2.
Selection 2: The amplified phage pool from selection on peptide(l-79) was divided into two parts and used for selection on a) 2xl0"8 M biotinylated peptide (1-79) in 1 ml buffer, as above. In addition, lxlO"7 M of non-biotinylated peptide (12-35) and peptide (45-65) were used for counter-selection, thereby increasing the probability to find fragments binding to other regions of peptide (1-79) than regions (12-35) nor (45-65).
b) 2xl0"8 M biotinylated peptide (1-79), as above. In addition, lxlO"7 M of non-biotinylated peptide (12-35) and peptide(45-65) were used for competition, thereby increasing the probability to find fragments binding to other regions of peptide (1-79) than regions (12-35) nor (45-65).
Amplified phage pools from selection on peptide (45-65), were further selected on the biotinylated using competition with non-biotinylated target peptides.
Selection 3: Selection was performed on non-biotinylated target antigens immobilised in microtiter plate wells, 2 pmole/well. Each selections were performed in 100 μl in 8 microtiter wells per target. The amplified phage pool from selection on peptide (1-79) was divided into three parts and used for selection on a) peptide (1-79) with counter-selection on lxlO"6 M biotinylated peptide (12-35) captured on Dynabead and competition by lxlO"6 M non-biotinylated peptide (12-35). b) peptide (1-79) with counter-selection on lxlO"6 M biotinylated peptide (45-65) captured on Dynabead and competition by lxlO"6
M non-biotinylated peptide (45-65). c) Peptide (1-79) with counter-selection on lxl 0"6 M biotinylated peptide (12-35 captured on Dynabeads, lxlO"6 M biotinylated peptide (45-65) captured on Dynabeads, and lxlO"6 M non- biotinylated peptides (12-35) and (45-65) Amplified phage pools from selection on peptide (12-35), were selected on peptide (12-35) and counter-selected on Dynabead coupled peptide (45-65) as well as competed non-biotinylated peptide.
Amplified phage pools from selection on peptide (45-65), were selected on peptide (45-65) and counter-selected on Dynabead coupled peptide (12-35) as well as competed non-biotinylated peptide.
Candidate target binding scFv clones were then identified in a screening process briefly described below:
The selected phage pools from selection 3 were then converted to scFv format by enzymatic cleavage of the phagemid, deleting the phage gene III . The resulting plasmids encoding soluble scFv were transformed into E.coli. Single bacterial colonies were picked for growth in LB-medium, and expression of soluble scFv was induced by addition of IPTG. The resulting scFv stocks were assayed for binding to the biotinylated peptides, loaded in avidin pre-coated wells, in Luminescence ELISA. Positive clones were further assayed in Luminescence ELISA against the target peptides, directly coated to the wells.
Positive clones from all groups were sequenced, and identical scFv clones were excluded. Chosen clones were expressed and purify by e.g protein A affinity chromatography. correct binding of the purified scFv clones were confirmed in ELISA
The chosen clones may preferably be converted to IgGl or IgG4 format using techniques well known in the art, for example as described in Henderikx et al., (2002; Am. J. Pathol. 160, 1597-1608).
The clones may be transferred to a IgGl or IgG4 vector and the functionality of the clones tested by transient expression in, for example, Cos 7 cells.
Functional clones may then be transfected into NSO cells and bulk pools expanded.
Figure imgf000022_0001

Claims

Claims:
1. A human antibody, fragment or derivative thereof which recognises ApoCIII, for the treatment of atherosclerosis or dyslipidaemia, or diseases associated therewith or resulting therefrom.
2. An antibody according to claim 1 in which has a CDR corresponding to any of SEQ ID Nos 49 to 78, or modifications thereof, which recognise human ApoCIII or peptides, fragments or mimotopes thereof.
3. An antibody according to claim 1 or 2 in which the antibody has a CDR corresponding to any of SEQ ID Nos 49 to 66, or modifications thereof, which recognise human ApoCIII or peptides, fragments or mimotopes thereof.
4. Use of a human antibody, fragment or derivative thereof according to any of claims 1 to 3, for the manufacture of a medicament for the treatment of atherosclerosis or dyslipidaemia, or diseases associated therewith or resulting therefrom.
5. A pharmaceutical composition comprising an antibody, fragment or derivative thereof according to any of claims 1 to 3 which recognises human ApoCIII or peptides, fragments or mimotopes thereof, together with a pharmaceutically acceptable excipient
6. A human antibody, fragment or derivative thereof, which recognises ApoCIII, for use in medicine.
7. A nucleotide molecule encoding a CDR of SEQ ID No 49 to 78
8. A nucleotide molecule encoding an antibody according to any of claims 1 to 3.
9. A vector containing the nucleic acid molecule of claim 8
10. A host cell containing the vectors of claim 9
11. A stable cell-line comprising the host cell of claim 10
12. A method of producing the antibodies of any of claims 1 to 3, comprising screening a phage display library for fragments capable of binding to ApoCIII, or a peptide, fragment or mimotope thereof, identifying the CDRs of the fragments, and inserting the identified CDRs into a human antibody scaffold to obtain a human antibody capable of binding to ApoCIII, or a peptide, fragment or mimotope thereof.
13. The method according to claim 12 in which the fragments are scFv or Fab fragments.
PCT/EP2004/002640 2003-03-13 2004-03-11 Human monoclonal antibodies against apociii and their use in the therapy of atherosclerosis WO2004081046A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB0305790.8A GB0305790D0 (en) 2003-03-13 2003-03-13 Novel Composition
GB0305790.8 2003-03-13

Publications (2)

Publication Number Publication Date
WO2004081046A2 true WO2004081046A2 (en) 2004-09-23
WO2004081046A3 WO2004081046A3 (en) 2005-02-17

Family

ID=9954729

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2004/002640 WO2004081046A2 (en) 2003-03-13 2004-03-11 Human monoclonal antibodies against apociii and their use in the therapy of atherosclerosis

Country Status (2)

Country Link
GB (1) GB0305790D0 (en)
WO (1) WO2004081046A2 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010127642A1 (en) 2009-05-04 2010-11-11 Centro De Inmunologia Molecular Antibodies that recognize sulphatides and sulphated proteoglycans and the use thereof
US20120315276A1 (en) * 2009-12-10 2012-12-13 Bayer Intellectual Property Gmbh Neutralizing prolactin receptor antibodies and their therapeutic use
WO2014131008A1 (en) * 2013-02-25 2014-08-28 Intrinsic Metabio Solutions, Llc A polipoprotein c3 (apociii) antagonists and methods of their use to remove apociii inhibition of lipoprotein lipase (lpl)
CN106810606A (en) * 2015-11-27 2017-06-09 西藏自治区人民医院 A kind of preparation and application of Apolipoprotein C-III antigen polypeptide and its polyclonal antibody
WO2018007999A1 (en) * 2016-07-08 2018-01-11 Staten Biotechnology B.V. Anti-apoc3 antibodies and methods of use thereof
US10538583B2 (en) 2017-10-31 2020-01-21 Staten Biotechnology B.V. Anti-APOC3 antibodies and compositions thereof
WO2020070678A3 (en) * 2018-10-03 2020-05-14 Staten Biotechnology B.V. Antibodies specific for human and cynomolgus apoc3 and methods of use thereof
WO2021119482A1 (en) 2019-12-13 2021-06-17 Progenity, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract
US11242381B2 (en) 2017-04-21 2022-02-08 Staten Biotechnology B.V. Anti-ApoC3 antibodies and methods of use thereof
US11248041B2 (en) 2017-10-31 2022-02-15 Staten Biotechnology B.V. Anti-ApoC3 antibodies

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001064008A2 (en) * 2000-03-03 2001-09-07 Smithkline Beecham Biologicals S.A. Vaccine for the treatment of artherosclerosis
WO2002098919A2 (en) * 2001-06-05 2002-12-12 Genfit Method for apo ciii measurement in apob and non apob containing particles
WO2003020765A2 (en) * 2001-08-31 2003-03-13 Glaxosmithkline Biologicals S.A. Vaccine therapies and prophylactic treatments of atherosclerotic diseases

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001064008A2 (en) * 2000-03-03 2001-09-07 Smithkline Beecham Biologicals S.A. Vaccine for the treatment of artherosclerosis
WO2002098919A2 (en) * 2001-06-05 2002-12-12 Genfit Method for apo ciii measurement in apob and non apob containing particles
WO2003020765A2 (en) * 2001-08-31 2003-03-13 Glaxosmithkline Biologicals S.A. Vaccine therapies and prophylactic treatments of atherosclerotic diseases

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ENGBERG J ET AL: "PHAGE-DISPLAY LIBRARIES OF MURINE AND HUMAN ANTIBODY FAB FRAGMENTS" NUCLEIC ACID PROTOCOLS HANDBOOK, XX, XX, 2000, pages 449-478, XP001037201 *
MARGET M ET AL: "BYPASSING HYBRIDOMA TECHNOLOGY: HLA-C REACTIVE HUMAN SINGLE-CHAIN ANTIBODY FRAGMENTS (SCFV) DERIVED FROM A SYNTHETIC PHAGE DISPLAY LIBRARY (HUCAL) AND THEIR POTENTIAL TO DISCRIMINATE HLA CLASS I SPECIFICITIES" TISSUE ANTIGENS, MUNKSGAARD, COPENHAGEN, DK, vol. 56, no. 1, July 2000 (2000-07), pages 1-9, XP000971781 ISSN: 0001-2815 *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010127642A1 (en) 2009-05-04 2010-11-11 Centro De Inmunologia Molecular Antibodies that recognize sulphatides and sulphated proteoglycans and the use thereof
US20120315276A1 (en) * 2009-12-10 2012-12-13 Bayer Intellectual Property Gmbh Neutralizing prolactin receptor antibodies and their therapeutic use
WO2014131008A1 (en) * 2013-02-25 2014-08-28 Intrinsic Metabio Solutions, Llc A polipoprotein c3 (apociii) antagonists and methods of their use to remove apociii inhibition of lipoprotein lipase (lpl)
US9783600B2 (en) 2013-02-25 2017-10-10 Imbp Holding, Llc Apolipoprotein C3 (ApoCIII) antagonists and methods of their use to remove ApoCIII inhibition of lipoprotein lipase (LPL)
CN106810606A (en) * 2015-11-27 2017-06-09 西藏自治区人民医院 A kind of preparation and application of Apolipoprotein C-III antigen polypeptide and its polyclonal antibody
US11091539B2 (en) 2016-07-08 2021-08-17 Staten Biotechnology B.V. Anti-ApoC3 antibodies and methods of use thereof
WO2018007999A1 (en) * 2016-07-08 2018-01-11 Staten Biotechnology B.V. Anti-apoc3 antibodies and methods of use thereof
CN109689685A (en) * 2016-07-08 2019-04-26 斯塔滕生物技术有限公司 Anti- APOC3 antibody and its application method
JP2019525772A (en) * 2016-07-08 2019-09-12 スターテン・バイオテクノロジー・ベー・フェー Anti-APOC3 antibody and method of use thereof
US11242381B2 (en) 2017-04-21 2022-02-08 Staten Biotechnology B.V. Anti-ApoC3 antibodies and methods of use thereof
US10538583B2 (en) 2017-10-31 2020-01-21 Staten Biotechnology B.V. Anti-APOC3 antibodies and compositions thereof
US11248041B2 (en) 2017-10-31 2022-02-15 Staten Biotechnology B.V. Anti-ApoC3 antibodies
US11248042B2 (en) 2017-10-31 2022-02-15 Staten Biotechnology B.V. Polynucleotides encoding anti-ApoC3 antibodies
WO2020070678A3 (en) * 2018-10-03 2020-05-14 Staten Biotechnology B.V. Antibodies specific for human and cynomolgus apoc3 and methods of use thereof
WO2021119482A1 (en) 2019-12-13 2021-06-17 Progenity, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract
EP4309722A2 (en) 2019-12-13 2024-01-24 Biora Therapeutics, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract

Also Published As

Publication number Publication date
WO2004081046A3 (en) 2005-02-17
GB0305790D0 (en) 2003-04-16

Similar Documents

Publication Publication Date Title
US20040185044A1 (en) Novel composition
EP1267908B1 (en) Vaccine for the treatment of atherosclerosis
US7601353B2 (en) Peptide-based passive immunization therapy for treatment of atherosclerosis
JP2005508900A6 (en) vaccine
RU2454428C2 (en) Immunotherapeutic treatment
WO2004081046A2 (en) Human monoclonal antibodies against apociii and their use in the therapy of atherosclerosis
JP2006506450A5 (en)
KR20100080507A (en) Treatment of atherosclerosis
US20050287137A1 (en) Novel composition
WO2004080375A2 (en) Vaccine therapy of atherosclerosis
AU2003267905B2 (en) Peptide-based passive immunization therapy for treatment of atherosclerosis
WO2004081045A2 (en) Vaccine related to modified apolipoprotein c-iii
KR20140026390A (en) Cetp fragments
ZA200207001B (en) Vaccine for the treatment of artherosclerosis.
TW201420112A (en) Peptide, pharmaceutical compound and use of the pharmaceutical compound thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): BW GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase