WO2019136516A1 - Procédés de traitement d'une myocardite et/ou d'une cardiomyopathie et réactifs correspondants - Google Patents

Procédés de traitement d'une myocardite et/ou d'une cardiomyopathie et réactifs correspondants Download PDF

Info

Publication number
WO2019136516A1
WO2019136516A1 PCT/AU2019/050006 AU2019050006W WO2019136516A1 WO 2019136516 A1 WO2019136516 A1 WO 2019136516A1 AU 2019050006 W AU2019050006 W AU 2019050006W WO 2019136516 A1 WO2019136516 A1 WO 2019136516A1
Authority
WO
WIPO (PCT)
Prior art keywords
seq
set forth
isolated
antibody
sequence set
Prior art date
Application number
PCT/AU2019/050006
Other languages
English (en)
Inventor
Maria Halasz
Darren Jones
Ulrich GRABMAIER
Ludwig WECKBACH
Barbara WALZOG
Graham Robertson
Original Assignee
Cellmid Limited
Ludwig-Maximilians-Universität München
Ludwig-Maximilians-Universität München, Medizinische Klinik Und Poliklinik I
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
Priority claimed from AU2018900052A external-priority patent/AU2018900052A0/en
Application filed by Cellmid Limited, Ludwig-Maximilians-Universität München, Ludwig-Maximilians-Universität München, Medizinische Klinik Und Poliklinik I filed Critical Cellmid Limited
Publication of WO2019136516A1 publication Critical patent/WO2019136516A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • 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]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding

Definitions

  • the present disclosure is directed to methods and reagents for treatment or prevention of myocarditis and/or cardiomyopathy.
  • the method and reagents of the disclosure may be useful for treating or preventing cardiac hypertrophy and/or preventing cardiac fibrosis and/or improving cardiac function in a subject suffering from cardiomyopathy.
  • the present disclosure relates to the use of an isolated or recombinant antibody or antigen binding fragment thereof, which binds to the N-terminal domain of midkine protein (hereinafter, referred to as "MK”), and inhibits or reduces the function of MK in the treatment or prevention of myocarditis and/or cardiomyopathy.
  • MK midkine protein
  • Myocarditis is an acute or chronic disease of heart muscle involving focal or diffuse inflammatory infiltrate in interstitial myocardium and degeneration, necrosis or lysis of cardiac muscle fiber.
  • the disease can lead to myocardial damage, heart dysfunction and arrhythmia, and in more extreme circumstances involving prolonged cardiac hypertrophy and permanent cardiac muscle injury, it can progress to dilated cardiomyopathy (weakening of the heart muscle), a major cause of heart failure and death.
  • the aetiology of myocarditis can be due to a wide variety of injuries to the myocardium, including injury caused by toxins and drugs (e.g ., cocaine and interleukin 2) or infectious agents, most commonly including viral (e.g., coxsackievirus, adenovirus, HIV and hepatitis C virus), bacterial (e.g., diphtheria, meningococcus, psittacosis and streptococcus), rickettsial (e.g., typhus and Rocky Mountain spotted fever), fungal (e.g., aspergillosis and candidiasis), and parasitic (Chagas disease, toxoplasmosis), as well as giant cell myocarditis, and hypersensitivity reactions to drugs such as antibiotics, sulfonamides, anticonvulsants, and anti-inflammatories. It has also been reported that in autoimmune-prone mice and patients with type 1 diabetes, chronic myocardit
  • myocarditis and cardiomyopathy depend upon the underlying cause and upon the individual patient.
  • existing treatments for viral myocarditis include antibiotics, heart protective agents, and antioxidant (such as high dose vitamin C, vitamin E and coenzyme Q10).
  • more aggressive therapy may be necessary, such as drugs to reduce the heart’s workload (e.g ., ACE inhibitors, Angiotensin III receptor blockers (ARBs) or beta blockers), drugs to eliminate excess fluid (e.g., diuretics), intravenous medication to improve the heart pumping function, placement of a pump in the aorta (intra-aortic balloon pump), use of a temporary artificial heart (e.g., ventricular assist device (VAD)), and consideration of urgent heart transplantation.
  • drugs to reduce the heart’s workload e.g ACE inhibitors, Angiotensin III receptor blockers (ARBs) or beta blockers
  • drugs to eliminate excess fluid e.g., diuretics
  • intravenous medication to improve the heart pumping function
  • placement of a pump in the aorta intra-aortic balloon pump
  • a temporary artificial heart e.g., ventricular assist device (VAD)
  • VAD ventricular assist device
  • Some patients may have chronic and irreversible
  • MK Midkine
  • MK is known to have various biological activities. For example, MK expression is known to be increased in a number of different human cancer cells (Muramatsu (2002) J. Biochem. 132:359-371), and its expression has been found to promote the survival and migration of cancer cells, promote angiogenesis, and contribute to cancer progression.
  • MK is also known to play a central role in inflammatory processes. For example, it is known that neointimal formation after vascular injury and nephritis onset during ischemic injury in the kidney are suppressed in knockout mice deficient in MK genes. Moreover, it is also known that rheumatic injury models and post-operative adhesions are significantly suppressed in such knockout mice (W02000/10608; W02004/078210).
  • MK The three-dimensional structure of MK has been determined by NMR and reported (Iwasaki et al. (1997) EMBO J. 16, p. 6936-6946).
  • MK is composed of: an N-terminal fragment (hereinafter, referred to as an "N-fragment") consisting of amino acid residues 1 to 52; a C-terminal fragment (hereinafter, referred to as a "C-fragment”) consisting of amino acid residues 62 to 121; and a loop region (amino acid residues 53 to 61) (hereinafter, referred to as a "loop”) that links these fragments.
  • N-fragment an N-terminal fragment
  • C-fragment C-terminal fragment
  • loop region amino acid residues 53 to 61
  • Each of the N- and C-fragments is mainly composed of: a portion having a three- dimensional structure consisting of three antiparallel [betaj-sheets (hereinafter, referred to as a "domain”; the domain (consisting of amino acid residues 15 to 52) in the N-fragment is referred to as an "N-domain", and the domain (consisting of amino acid residues 62 to 104) in the C-fragment is referred to as a "C-domain”); and a terminally located portion devoid of the domain that does not assume a particular three-dimensional structure (hereinafter, referred to as a "tail”; the tail (consisting of amino acid residues 1 to 14) in the N-fragment is referred to as an "N-tail", and the tail (consisting of amino acid residues 105 to 121) in the C-fragment is referred to as a "C-tail”).
  • domain the domain (consisting of amino acid residues 15 to 52) in the N-
  • Anti-MK antibodies against the C-domain and the N-domain are known e.g., as disclosed in W02008/059616, WO2012/122590 and W02016/058047. Based on previous findings that MK has a number of biological activities and is implicated in a range of diseases and conditions, these and other anti-MK antibodies may be therapeutically effective for a number of diseases/conditions.
  • the present disclosure is based on the inventors’ finding that treatment of an animal model of myocarditis with an anti-MK antibody which binds specifically to the N-domain of MK, significantly reduced cardiac hypertrophy, fibrosis and heart failure relative to animals which received vehicle control, IgG isotype control or an anti-MK antibody which binds specifically to the C-domain of MK.
  • the present disclosure thus provides a method of treating and/or preventing myocarditis or cardiomyopathy in a subject, said method comprising administering to the subject an isolated or recombinant protein comprising an antigen binding domain of an antibody which binds specifically to an epitope located within the N-domain of MK protein and inhibits or reduces a function of MK.
  • administering the isolated or recombinant protein to the subject prevents cardiac hypertrophy and/or prevents cardiac fibrosis and/or improves heart function in a subject.
  • the subject is suffering from myocarditis and administering the isolated or recombinant protein to the subject prevents development of cardiomyopathy.
  • the myocarditis is the result of injury to the myocardium caused by toxins and/or drugs e.g., including therapeutic drugs.
  • the myocarditis is the result of injury to the myocardium caused by infectious agents, such as e.g., viral, bacterial, rickettsial, fungal, and parasitic agents.
  • the myocarditis is the result of injury to the myocardium caused by an autoimmune disease.
  • the myocarditis is giant cell myocarditis.
  • the myocarditis is the result of a hypersensitivity reaction to a drug, such as an antibiotics, sulfonamides, anticonvulsants, and anti inflammatories.
  • the myocarditis is acute. In one example, the myocarditis is subacute. In one example, the myocarditis is chronic.
  • the cardiomyopathy may be selected from the group consisting of hypertrophic cardiomyopathy, dilated cardiomyopathy, restrictive cardiomyopathy, arrhythmogenic right ventricular dysplasia, metabolic cardiomyopathy and unclassified cardiomyopathy.
  • the cardiomyopathy is hypertrophic cardiomyopathy.
  • the cardiomyopathy is dilated cardiomyopathy.
  • the cardiomyopathy is restrictive cardiomyopathy.
  • the cardiomyopathy is arrhythmogenic right ventricular dysplasia.
  • the cardiomyopathy is metabolic cardiomyopathy (e.g diabetic cardiomyopathy, alcoholic cardiomyopathy, or drug-induced cardiomyopathy).
  • the cardiomyopathy is unclassified cardiomyopathy.
  • the epitope to which the isolated or recombinant protein binds is located within the N-domain of MK as defined by amino acid residues 1-61 of the sequence set forth in SEQ ID NO: 1.
  • the isolated or recombinant protein useful in the method of the disclosure recognizes at least a portion of a high electrostatic potential cluster located at amino acid residues 1-61 of the sequence set forth in SEQ ID NO: 1.
  • the isolated or recombinant protein useful in the method of the disclosure binds specifically to a conformational epitope formed by the amino acid sequence set forth in SEQ ID NO: 1, wherein the epitope includes at least two residues selected from the group consisting of 18W, 20W, 34F, 35R, 36E, 38T, 43T, 45R, 47R and 49R.
  • the isolated or recombinant protein binds an epitope defined by residues 18W, 20W, 35R and 49R.
  • the isolated or recombinant protein binds an epitope defined by residues 18W, 20W, 36E, 38T, 43T and 45R.
  • the isolated or recombinant protein binds an epitope defined by residues 18W, 20W, 34F, 36E, 45R and 47R.
  • an isolated or recombinant protein useful in the method of the disclosure comprises:
  • VH heavy chain variable domain
  • VL light chain variable domain
  • VH comprising the sequence set forth in SEQ ID NO: 2 or a sequence exhibiting 95% or greater identity thereto and a VL comprising the sequence set forth in SEQ ID NO: 3 or a sequence exhibiting 95% or greater identity thereto;
  • VH comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 4, 5 and 6 respectively, and a VL comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 7, 8 and 9 respectively; or
  • the isolated or recombinant protein comprises a VH comprising the sequence set forth in SEQ ID NO: 2 or a sequence exhibiting 95% or greater identity thereto. In one example, the isolated or recombinant protein comprises a VL comprising the sequence set forth in SEQ ID NO: 3 or a sequence exhibiting 95% or greater identity thereto. In one example, the isolated or recombinant protein comprises a VH comprising the sequence set forth in SEQ ID NO: 2 or a sequence exhibiting 95% or greater identity thereto and a VL comprising the sequence set forth in SEQ ID NO: 3 or a sequence exhibiting 95% or greater identity thereto.
  • the isolated or recombinant protein comprises a VH comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 4, 5 and 6 respectively, and a VL comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 7, 8 and 9 respectively.
  • the isolated or recombinant protein comprises three CDRs comprised within the sequence set forth in SEQ ID NO: 2 and three CDRs comprised within the sequence set forth in SEQ ID NO: 3.
  • an isolated or recombinant protein useful in the method of the disclosure comprises:
  • VH heavy chain variable domain
  • VL light chain variable domain
  • VH comprising the sequence set forth in SEQ ID NO: 10 or a sequence exhibiting 95% or greater identity thereto and a VL comprising the sequence set forth in SEQ ID NO: 11 or a sequence exhibiting 95% or greater identity thereto;
  • VH comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 12, 13 and 14 respectively
  • VL comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 15, 16 and 17 respectively
  • the isolated or recombinant protein comprises a VH comprising the sequence set forth in SEQ ID NO: 10 or a sequence exhibiting 95% or greater identity thereto. In one example, the isolated or recombinant protein comprises a VL comprising the sequence set forth in SEQ ID NO: 11 or a sequence exhibiting 95% or greater identity thereto. In one example, the isolated or recombinant protein comprises a VH comprising the sequence set forth in SEQ ID NO: 10 or a sequence exhibiting 95% or greater identity thereto and a VL comprising the sequence set forth in SEQ ID NO: 11 or a sequence exhibiting 95% or greater identity thereto.
  • the isolated or recombinant protein comprises a VH comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 12, 13 and 14 respectively, and a VL comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 15, 16 and 17 respectively.
  • the isolated or recombinant protein comprises three CDRs comprised within the sequence set forth in SEQ ID NO: 10 and three CDRs comprised within the sequence set forth in SEQ ID NO: 11.
  • an isolated or recombinant protein useful in the method of the disclosure comprises:
  • VH heavy chain variable domain
  • VL light chain variable domain
  • VH comprising the sequence set forth in SEQ ID NO: 18 or a sequence exhibiting 95% or greater identity thereto and a VL comprising the sequence set forth in SEQ ID NO: 19 or 20 or a sequence exhibiting 95% or greater identity thereto;
  • VH comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 21, 22 and 23 respectively
  • VL comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 24, 25 and 26 respectively
  • the isolated or recombinant protein comprises a VH comprising the sequence set forth in SEQ ID NO: 18 or a sequence exhibiting 95% or greater identity thereto. In one example, the isolated or recombinant protein comprises a VL comprising the sequence set forth in SEQ ID NO: 19 or 20 or a sequence exhibiting 95% or greater identity thereto. In one example, the isolated or recombinant protein comprises a VH comprising the sequence set forth in SEQ ID NO: 18 or a sequence exhibiting 95% or greater identity thereto and a VL comprising the sequence set forth in SEQ ID NO: 19 or 20 or a sequence exhibiting 95% or greater identity thereto.
  • the isolated or recombinant protein comprises a VH comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 21, 22 and 23 respectively, and a VL comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 24, 25 and 26 respectively.
  • the isolated or recombinant protein comprises three CDRs comprised within the sequence set forth in SEQ ID NO: 18 and three CDRs comprised within the sequence set forth in SEQ ID NO: 19 or 20.
  • the isolated or recombinant protein comprises a heavy chain variable domain (VH) and a light chain variable domain (VL).
  • VH heavy chain variable domain
  • VL light chain variable domain
  • the VH and the VL are in a single polypeptide chain.
  • the isolated or recombinant protein may be:
  • the VL and VH are provided in separate polypeptide chains.
  • the isolated or recombinant protein may be:
  • the isolated or recombinant protein is a chimeric, de-immunized, humanized or human antibody.
  • the isolated or recombinant protein may comprise a human or non human primate heavy chain immunoglobulin constant region selected from a group consisting of IgGl, IgG2, IgG3, IgG4, IgM, IgE and IgA.
  • the isolated or recombinant protein may be conjugated to a compound.
  • the isolated or recombinant protein may conjugated to a compound is selected from the group consisting of a radioisotope, a detectable label, a therapeutic compound, a colloid, a toxin, a nucleic acid, a peptide, a protein, a compound that increases the half-life of the protein in a subject and mixtures thereof.
  • the subject has received or will receive treatment with one or more other agents for treatment of myocarditis or cardiomyopathy.
  • the method comprises administering the isolated or recombinant protein in combination with one or more other agents for treatment of myocarditis or cardiomyopathy.
  • the present disclosure also provides for use of an isolated or recombinant protein comprising an antigen binding domain of an antibody which binds specifically to an epitope located within the N-domain of midkine (MK) protein and which inhibits or reduces a function of MK in the preparation of a medicament for treating and/or preventing myocarditis or cardiomyopathy in a subject in need thereof.
  • MK midkine
  • Suitable isolated or recombinant proteins are as described in any example hereof.
  • treatment with the medicament prevents cardiac hypertrophy and/or prevents cardiac fibrosis and/or improves heart function in the subject.
  • the subject is suffering from myocarditis and treatment with the medicament prevents development of cardiomyopathy in the subject.
  • the myocarditis is the result of injury to the myocardium caused by toxins and/or drugs e.g., including therapeutic drugs.
  • the myocarditis is the result of injury to the myocardium caused by infectious agents, such as e.g., viral, bacterial, rickettsial, fungal, and parasitic agents.
  • the myocarditis is the result of injury to the myocardium caused by an autoimmune disease.
  • the myocarditis is giant cell myocarditis.
  • the myocarditis is the result of a hypersensitivity reaction to a drug, such as an antibiotics, sulfonamides, anticonvulsants, and anti inflammatories.
  • the myocarditis is acute. In one example, the myocarditis is subacute. In one example, the myocarditis is chronic.
  • the cardiomyopathy may be selected from the group consisting of hypertrophic cardiomyopathy, dilated cardiomyopathy, restrictive cardiomyopathy, arrhythmogenic right ventricular dysplasia, metabolic cardiomyopathy and unclassified cardiomyopathy.
  • the cardiomyopathy is hypertrophic cardiomyopathy.
  • the cardiomyopathy is dilated cardiomyopathy.
  • the cardiomyopathy is restrictive cardiomyopathy.
  • the cardiomyopathy is arrhythmogenic right ventricular dysplasia.
  • the cardiomyopathy is metabolic cardiomyopathy (e.g., diabetic cardiomyopathy, alcoholic cardiomyopathy, or drug-induced cardiomyopathy).
  • the cardiomyopathy is unclassified cardiomyopathy.
  • Figure 1 shows that blocking midkine with an antibody targeting the N-domain of midkine (i.e., IP 10) reduced heart/body weight ratio in EAM mice at day 21 post treatment.
  • Figure 2 shows that blocking midkine with an antibody targeting the N-domain of midkine (i.e., IP 10) reduces cardiac fibrosis in an EAM mouse model of myocarditis
  • a Representative cross sections of cardiac tissue of non-immunized mice (left panel), vehicle- treated immunized mice (middle panel) as well as immunized mice after treatment with the anti-N-MK antibody IP 10 (right panel) on day 63 using Masson's trichrome staining. Fibrotic tissue appears blue. Scale bar (overview), 1 mm; (magnification), 100 pm.
  • Figure 3 shows that blocking midkine with an antibody targeting the N-domain of midkine (i.e., IP 10) preserves cardiac function in an EAM mouse model of myocarditis
  • Parameters describing systolic function using (b) fractional shortening and (c) left ventricular ejection fraction (LVEF).
  • n 25 per group.
  • Figure 4 shows that administration of an Isotype IgG control antibody does not affect myocarditis
  • a Degree of fibrosis in the cardiac tissue as analyzed histologically using semi- quantitative score on day 63 after induction of myocarditis.
  • Mice were treated with IgGl isotype antibody from day 0 until day 21 of EAM myocarditis (immunized + isotype Ctrl or left untreated for control (immunized + vehicle)
  • N 25. n.s. not significant.
  • SEQ ID NO: 1 Human midkine protein sequence.
  • SEQ ID NO: 10 IP-10 variable heavy chain protein sequence.
  • SEQ ID NO: 12 IP-10 variable heavy chain CDR1 protein sequence.
  • SEQ ID NO: 13 IP-10 variable heavy chain CDR2 protein sequence.
  • SEQ ID NO: 14 IP-10 variable heavy chain CDR3 protein sequence.
  • SEQ ID NO: 15 IP-10 variable light chain CDR1 protein sequence.
  • SEQ ID NO: 16 IP-10 variable light chain CDR2 protein sequence.
  • SEQ ID NO: 17 IP- 10 variable light chain CDR3 protein sequence.
  • SEQ ID NO: 18 IP- 13 variable heavy chain protein sequence.
  • SEQ ID NO: 19 IP-13 variable light chain vl protein sequence.
  • SEQ ID NO:28 IP- 14 variable heavy chain CDR2 protein sequence.
  • SEQ ID NO:30 IP- 14 variable light chain CDR1 protein sequence.
  • SEQ ID NO:33 Amino acid sequence for synthetic cardiac peptide aMyHC.
  • composition of matter, group of steps or group of compositions of matter shall be taken to encompass one and a plurality ( i.e . one or more) of those steps, compositions of matter, groups of steps or groups of compositions of matter.
  • variable regions and parts thereof, immunoglobulins, antibodies and fragments thereof herein may be further clarified by the discussion in Kabat Sequences of Proteins of Immunological Interest , National Institutes of Health, Bethesda, Md., 1987 and 1991, Bork et al, (1994) J. Mol. Biol. 242:309-320, Chothia and Lesk (1987) J. Mol. Biol. 196:901 -917, Chothia et al. (1989) Nature 342:877-883, and/or Al-Lazikani et al., (1997) J. Mol. Biol. 273:927-948.
  • an“antibody” is generally considered to be a protein that comprises a variable region made up of a plurality of immunoglobulin chains, e.g., a polypeptide comprising a VL and a polypeptide comprising a VH.
  • An antibody also generally comprises constant domains, some of which can be arranged into a constant region or constant fragment or fragment crystallisable (Fc).
  • a VH and a VL interact to form an Fv comprising an antigen binding region that is capable of specifically binding to one or a few closely related antigens.
  • a light chain from mammals is either a k light chain or a l light chain and a heavy chain from mammals is a, d, e, g, or m.
  • Antibodies can be of any type (e.g., IgG, IgE, IgM, IgD, IgA, and IgY), class (e.g., IgGi, IgG 2 , IgGs, IgG 4 , IgAi and IgA 2 ) or subclass.
  • the term“antibody” also encompasses humanized antibodies, de-immunized antibodies, non-depleting antibodies, non-activating antibodies, primatized antibodies, human antibodies and chimeric antibodies.
  • the term“antibody” is also intended to include formats other than full-length, intact or whole antibody molecules, such as Fab, F(ab')2, and Fv which are capable of binding the epitopic determinant. These formats may be referred to as antibody“fragments”. These antibody formats retain some ability to selectively bind to human midkine, examples of which include, but are not limited to, the following:
  • Fab the fragment which contains a monovalent binding fragment of an antibody molecule and which can be produced by digestion of whole antibody with the enzyme papain to yield an intact light chain and a portion of one heavy chain;
  • Fab' the fragment of an antibody molecule which can be obtained by treating whole antibody with pepsin, followed by reduction, to yield an intact light chain and a portion of the heavy chain; two Fab' fragments are obtained per antibody molecule;
  • (Fab')2 the fragment of the antibody that can be obtained by treating whole antibody with the enzyme pepsin without subsequent reduction
  • F(ab)2 is a dimer of two Fab' fragments held together by two disulfide bonds;
  • Fv defined as a genetically engineered fragment containing the variable region of the light chain and the variable region of the heavy chain expressed as two chains;
  • Single chain antibody defined as a genetically engineered molecule containing the variable region of the light chain, the variable region of the heavy chain, linked by a suitable polypeptide linker as a genetically fused single chain molecule; such single chain antibodies may be in the form of multimers such as diabodies, triabodies, and tetrabodies etc which may or may not be polyspecific (see, for example, WO1994/007921 and WO 1998/044001); and
  • Single domain antibody typically a variable heavy domain devoid of a light chain.
  • an antibody in accordance with the present disclosure includes separate heavy chains, light chains, Fab, Fab', F(ab')2, Fc, a variable light domain devoid of any heavy chain, a variable heavy domain devoid of a light chain and Fv.
  • Such fragments can be produced by recombinant DNA techniques, or by enzymatic or chemical separation of intact immunoglobulins.
  • full-length antibody “intact antibody” or “whole antibody” are used interchangeably to refer to an antibody in its substantially intact form, as opposed to an antigen binding fragment of an antibody.
  • whole antibodies include those with heavy and light chains including an Fc region.
  • the constant domains may be wild-type sequence constant domains (e.g ., human wild-type sequence constant domains) or amino acid sequence variants thereof.
  • the intact antibody may have one or more effector functions.
  • the antibody disclosed herein may be a humanized antibody.
  • humanized antibody refers to an antibody derived from a non-human antibody, typically murine, that retains or substantially retains the antigen-binding properties of the parent antibody but which is less immunogenic in humans.
  • the antibody disclosed herein may be a non-depleting antibody.
  • the immune system’s effector functions are dependent on interactions of the Fc-domain with Clq, the first component of the complement cascade, and/or receptors (FcR).
  • FcR Factor receptors
  • Complement-dependent cytotoxicity (CDC) is initiated by multiple Fc-domains interacting with Clq, which can ultimately result in lysis of target cells through the formation of the membrane attack complex (MAC).
  • MAC membrane attack complex
  • cells of the immune system such as granulocytes, macrophages, and NR cells, may interact via FcRs with mAbs bound to target cells.
  • Non depleting antibodies include antibody fragments without an Fc domain, including for example, monovalent (e.g., Fab, scFv, nanobodies and dAbs), bivalent (e.g., F(ab’)2 and diabodies) and multivalent (e.g., triabodies and pentabodies) formats.
  • monovalent e.g., Fab, scFv, nanobodies and dAbs
  • bivalent e.g., F(ab’)2 and diabodies
  • multivalent e.g., triabodies and pentabodies
  • non depleting antibodies include antibodies that have been modified to remove effector functions without impacting pharmokinetics, for example, amino acid residues in the Fc-domain that play a dominant role in interaction with Clq and FcRs could be modified, or the N-linked glycosylation site in the CH2 domain could be removed.
  • the chances of engineering a non-depleting antibody are linked to the constant region used to produce the antibody.
  • An IgG3 constant region is more likely to produce a depleting antibody than an IgGl constant region which in turn is more likely to produce a depleting antibody than an IgG2 constant region, whereas an IgG4 constant region will generally mean that the antibody is non-depleting.
  • modifications to a constant region could convert a depleting antibody into a non-depleting antibody and vice versa.
  • the antibody disclosed herein may be a non-activating antibody.
  • a “non-activating antibody” refers to antibodies that bind cell surface receptors and negate or block the action of endogenous ligands.
  • EU numbering system of Rabat will be understood to mean the numbering of an immunoglobulin heavy chain is according to the EU index as taught in Rabat et al, 1991, Sequences of Proteins of Immunological Interest, 5th Ed., United States Public Health Service, National Institutes of Health, Bethesda.
  • the EU index is based on the residue numbering of the human IgGl EU antibody.
  • variable region refers to the portions of the light and/or heavy chains of an antibody as defined herein that is capable of specifically binding to an antigen and, for example, includes amino acid sequences of CDRs; i.e., CDR1, CDR2, and CDR3, and framework regions (FRs).
  • the variable region comprises three or four FRs (e.g ., FR1, FR2, FR3 and optionally FR4) together with three CDRs.
  • VH refers to the variable region of the heavy chain.
  • VL refers to the variable region of the light chain.
  • amino acid positions assigned to CDRs and FRs can be defined according to Rabat (1987 and 1991, supra ) or other numbering systems in the performance of methods according to the present disclosure, e.g., the hypervariable loop numbering system of Clothia and Lesk (1987 and/or 1989, supra and/or Al-Lazikani et al, 1997, supra).
  • CDRs complementarity determining regions
  • CDR1, CDR2, and CDR3 refers to the amino acid residues of an antibody variable domain that form loops between the FRs the sequence of which vary between antibodies. Some or all of the CDRs confer the ability to bind antigen on the antibody.
  • Each variable domain typically has three CDR regions identified as CDR1, CDR2 and CDR3.
  • Each complementarity determining region may comprise amino acid residues from a "complementarity determining region" as defined by Rabat et al, (1991) and/or those residues from a "hypervariable loop” Chothia and Lesk (1987), or any other known numbering technique or combination thereof, including the IMGT numbering system (Lefranc et al, (2003) Dev. Comp. Immunol., 27(1)55-77).
  • Framework regions are those variable domain residues other than the CDR residues.
  • constant region or“fragment crystalizable” or“Fc” or“Fc region” or“Fc portion” (which can be used interchangeably herein) as used herein, refers to a portion of an antibody comprising at least one constant domain and which is generally (though not necessarily) glycosylated and which is capable of binding to one or more Fc receptors and/or components of the complement cascade.
  • the heavy chain constant region can be selected from any of the five isotypes: a, d, e, g, or m.
  • heavy chains of various subclasses are responsible for different effector functions and thus, by choosing the desired heavy chain constant region, proteins with desired effector function can be produced.
  • the constant regions of the antibodies of the disclosure are derived from human immunoglobulins.
  • Exemplary heavy chain constant regions are gamma 1 (IgGl), gamma 2 (IgG2), gamma 3 (IgG3), gamma 4 (IgG4), or hybrids thereof.
  • the light chain constant region can be of the kappa or lambda type, preferably of the kappa type.
  • A“constant domain” is a domain in an antibody the sequence of which is highly similar in antibodies/antibodies of the same type, e.g., IgG or IgM or IgE.
  • a constant region of an antibody generally comprises a plurality of constant domains, e.g., the constant region of g, a and d heavy chains comprises two constant domains.
  • the term“residue” as used herein refers to an amino acid residue.
  • the word“residue” may be used interchangeably with the term“amino acid”.
  • the term "recombinant" in the context of an antibody refers to the antibody when produced by a cell, or in a cell-free expression system, in an altered amount or at an altered rate compared to its native state.
  • the cell is a cell that does not naturally produce the antibody or immunoglobulin chain.
  • the cell may be a cell which comprises a non-endogenous gene that causes an altered, preferably increased, amount of the polypeptide to be produced.
  • a recombinant antibody of the disclosure includes polypeptides which have not been separated from other components of the transgenic (recombinant) cell, or cell-free expression system, in which it is produced, and an antibody produced in such cells or cell-free systems which are subsequently purified away from at least some other components.
  • the antibody disclosed herein may specifically bind to midkine protein (such as human midkine protein).
  • midkine protein such as human midkine protein
  • the term“specifically binds” shall be taken to mean a protein reacts or associates more frequently, more rapidly, with greater duration and/or with greater affinity with midkine or a specified epitope thereof than it does with alternative antigens or epitopes. As such, “specific binding” does not necessarily require exclusive binding or non-detectable binding of another antigen.
  • the term specifically binds” is used interchangeably with“selectively binds” herein.
  • overlapping in the context of two epitopes shall be taken to mean that two epitopes share a sufficient number of amino acid residues to permit an antibody that binds to one epitope to competitively inhibit the binding of an antibody that binds to the other epitope.
  • the two epitopes share at least 1 or 2 or 3 or 4 or 5 or 6 or more amino acids.
  • references herein to“monoclonal antibody IP-9” or to“IP-9” is a reference to the monoclonal antibody which has a variable heavy chain sequence as shown in SEQ ID NO:2 and a variable light chain sequence as shown in SEQ ID NO:3.
  • references herein to“monoclonal antibody IP- 10” or to“IP- 10” is a reference to the monoclonal antibody which has a variable heavy chain sequence as shown in SEQ ID NO: 10 and a variable light chain sequence as shown in SEQ ID NO: 11.
  • Reference herein to“monoclonal antibody IP-13” or to“IP- 13” is a reference to the monoclonal antibody which has a variable heavy chain sequence as shown in SEQ ID NO: 18 and a variable light chain sequence as shown in SEQ ID NO: 19 or 20.
  • Reference herein to“monoclonal antibody IP- 14”,“IP- 14” or“murine IP- 14” is a reference to the monoclonal antibody which has a variable heavy chain sequence comprising CDR1, CDR2 and CDR3 as shown in SEQ ID NOs: 27, 28 and 29, respectively, and a variable light chain sequence comprising CDR1, CDR2 and CDR3 as shown in SEQ ID NOs: 30, 31 and 32, respectively.
  • mAh IP14 is the same antibody as designated CSM-4 in W02008/059616.
  • the terms “treating”, “treat” or “treatment” and variations thereof, refer to clinical intervention designed to alter the natural course of the individual or cell being treated during the course of clinical pathology. Desirable effects of treatment include decreasing the rate of disease progression, ameliorating or palliating the disease state, and remission or improved prognosis.
  • An individual is successfully "treated", for example, if one or more symptoms associated with a disease/condition (e.g ., myocarditis or cardiomyopathy) and/or injury (e.g., cardiac injury associated with myocarditis or cardiomyopathy) are mitigated or eliminated or the clinical outcome or prognosis of the disease/condition or injury is improved.
  • a disease/condition e.g ., myocarditis or cardiomyopathy
  • injury e.g., cardiac injury associated with myocarditis or cardiomyopathy
  • treatment may result in a reduction in cardiac inflammation and a return or approach to normal cardiac function or rhythm.
  • treatment may prevent cardiac hypertrophy and/or prevent cardiac fibrosis and/or improve heart function in the subject.
  • prevention refers to the provision of prophylaxis with respect to occurrence or recurrence of a disease or condition in an individual.
  • An individual may be predisposed to or at risk of developing the disease/condition or disease/condition relapse but has not yet been diagnosed with the disease/condition or the relapse.
  • the term prevention does not require absolute prevention but includes inhibiting the progression of the disease or condition to some extent or delaying the onset on the disease or condition or a symptom/injury associated with the disease/condition.
  • prevention of myocarditis and/or cardiomyopathy may include reducing cardiac inflammation and/or preventing cardiac hypertrophy and/or preventing cardiac fibrosis and/or preventing injury to the myocardium and/or improving heart function in the subject.
  • an “effective amount” refers to at least an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic or prophylactic result.
  • An effective amount can be provided in one or more administrations.
  • the term "effective amount” is meant an amount necessary to effect treatment of a disease or condition as hereinbefore described.
  • the effective amount may vary according to the disease or condition to be treated and also according to the weight, age, racial background, sex, health and/or physical condition and other factors relevant to the mammal being treated.
  • the effective amount will fall within a relatively broad range (e.g. a "dosage" range) that can be determined through routine trial and experimentation by a medical practitioner.
  • the effective amount can be administered in a single dose or in a dose repeated once or several times over a treatment period.
  • a “therapeutically effective amount” is at least the minimum concentration required to effect a measurable improvement of a particular disease or condition (e.g., myocarditis or cardiomyopathy).
  • a therapeutically effective amount herein may vary according to factors such as the disease state, age, sex, and weight of the patient, and the ability of the protein to elicit a desired response in the individual.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the protein are outweighed by the therapeutically beneficial effects.
  • ECso effective concentration 50%
  • concentration 50% represents the concentration of an antibody of the disclosure that is required for 50% of a given effect of the molecule the antibody targets (e.g. inhibiting/displacing binding of human midkine to a target thereof). It will be understood by one in the art that a lower EC so value corresponds to a more potent antibody.
  • The“mammal” treated according to the present disclosure may be a human, primate, livestock (e.g. sheep, horses, cattle, pigs, donkeys), companion animal (e.g. pets such as dogs and cats), laboratory test animal (e.g. mice, rabbits, rats, guinea pigs), performance animal (e.g. racehorses, camels, greyhounds) or captive wild animal.
  • livestock e.g. sheep, horses, cattle, pigs, donkeys
  • companion animal e.g. pets such as dogs and cats
  • laboratory test animal e.g. mice, rabbits, rats, guinea pigs
  • performance animal e.g. racehorses, camels, greyhounds
  • the mammal is a human.
  • isolated or recombinant proteins comprising an antigen binding domain of an antibody which binds selectively to the N-domain of MK are contemplated for use in the method of the present disclosure.
  • Antibodies which bind selectively to the N- domain of MK are known in the art, including, but not limited to, those anti-MK antibodies as described in W02008/059616, WO2012/122590 and W02014/070642. Further anti-MK antibodies are described in Sun X. Z, et ah, (1997) J. Neuropathol. Exp. Neurol. 56(12): 1339- 48 and Muramatsu EL, et ah, (2004) J. Biochem., 119: 1171-77).
  • other isolated or recombinant proteins suitable for use in methods of the disclosure including antibodies which bind selectively to the N-domain of MK and proteins comprising antigen binding domains thereof, may be produced by methods known in the art.
  • Methods for producing isolated or recombinant proteins suitable for use in a method of the disclosure including antibodies which bind selectively to the N-domain of MK and binding fragments thereof, are described herein. Furthermore, functional assays for determining MK binding activity of a protein and its suitability for use in a method of the disclosure are also described herein.
  • An isolated or recombinant protein contemplated for use in a method of the disclosure is protein comprising an antigen binding domain of an antibody which binds specifically to an epitope located within the N-domain of MK e.g., as defined by amino acid residues 1-61 of the sequence set forth in SEQ ID NO: l, and thereby inhibits or reduces a function of MK.
  • an isolated or recombinant protein contemplated for use in a method of the disclosure recognizes at least a portion of a high electrostatic potential cluster located at amino acid residues 1-61 of the sequence set forth in SEQ ID NO: 1.
  • Suitable proteins for use in a method of the disclosure may bind specifically to a conformational epitope located within the N-domain of MK formed by the amino acid sequence set forth in SEQ ID NO: l, wherein the epitope includes at least two residues selected from the group consisting of 18W, 20W, 34F, 35R, 36E, 38T, 43T, 45R, 47R and 49R.
  • the conformational epitope is defined by residues 18W, 20W, 35R and 49R.
  • the conformational epitope is defined by residues 18W, 20W, 36E, 38T, 43T and 45R.
  • the conformational epitope is defined by residues 18W, 20W, 34F, 36E, 45R and 47R.
  • an isolated or recombinant protein contemplated for use in a method of the disclosure comprises an antigen binding domain of an antibody designated IP-9 as described in WO2012/122590 (the contents of which are incorporated by reference herein).
  • the isolated or recombinant protein may comprise:
  • VH comprising the sequence set forth in SEQ ID NO: 2 or a sequence exhibiting 95% or greater identity thereto;
  • VL comprising the sequence set forth in SEQ ID NO: 3 or a sequence exhibiting 95% or greater identity thereto;
  • VH comprising the sequence set forth in SEQ ID NO: 2 or a sequence exhibiting 95% or greater identity thereto and a VL comprising the sequence set forth in SEQ ID NO: 3 or a sequence exhibiting 95% or greater identity thereto;
  • VH comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 4, 5 and 6 respectively
  • VL comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 7, 8 and 9 respectively
  • a particulartly preferred protein for use in the method of the disclosure in accordance with this example is the antibody designated IP-9 having a VH comprising the sequence set forth in SEQ ID NO: 2 and a VL comprising the sequence set forth in SEQ ID NO: 3.
  • an isolated or recombinant protein contemplated for use in a method of the disclosure comprises an antigen binding domain of an antibody designated PM0 as described in WO2012/122590 (the contents of which are incorporated by reference herein).
  • the isolated or recombinant protein may comprise:
  • VH comprising the sequence set forth in SEQ ID NO: 10 or a sequence exhibiting 95% or greater identity thereto;
  • VL comprising the sequence set forth in SEQ ID NO: 11 or a sequence exhibiting 95% or greater identity thereto;
  • VH comprising the sequence set forth in SEQ ID NO: 10 or a sequence exhibiting 95% or greater identity thereto and a VL comprising the sequence set forth in SEQ ID NO: 11 or a sequence exhibiting 95% or greater identity thereto;
  • VH comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 12, 13 and 14 respectively
  • VL comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 15, 16 and 17 respectively
  • a particulartly preferred protein for use in the method of the disclosure in accordance with this example is the antibody designated IP- 10 having a VH comprising the sequence set forth in SEQ ID NO: 10 and a VL comprising the sequence set forth in SEQ ID NO: 11.
  • an isolated or recombinant protein contemplated for use in a method of the disclosure comprises an antigen binding domain of an antibody designated PM3 as described in WO2012/122590 (the contents of which are incorporated by reference herein).
  • the isolated or recombinant protein may comprise:
  • VH comprising the sequence set forth in SEQ ID NO: 18 or a sequence exhibiting 95% or greater identity thereto;
  • VL comprising the sequence set forth in SEQ ID NO: 19 or 20 or a sequence exhibiting 95% or greater identity thereto;
  • VH comprising the sequence set forth in SEQ ID NO: 18 or a sequence exhibiting 95% or greater identity thereto and a VL comprising the sequence set forth in SEQ ID NO: 19 or 20 or a sequence exhibiting 95% or greater identity thereto;
  • VH comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 21, 22 and 23 respectively
  • VL comprising CDR1, CDR2 and CDR3 comprising the sequences set forth in SEQ ID NO: 24, 25 and 26 respectively
  • a particularly preferred protein for use in the method of the disclosure in accordance with this example is the antibody designated IP- 13 having a VH comprising the sequence set forth in SEQ ID NO: 18 and a VL comprising the sequence set forth in SEQ ID NO: 19.
  • Another particulartly preferred protein for use in the method of the disclosure in accordance with this example is the antibody designated IP- 13 having a VH comprising the sequence set forth in SEQ ID NO: 18 and a VL comprising the sequence set forth in SEQ ID NO: 20.
  • the query sequence is at least 50 amino acids in length, and the GAP analysis aligns the two sequences over a region of at least 50 amino acids. Even more preferably, the query sequence is at least 100 amino acids in length and the GAP analysis aligns the two sequences over a region of at least 100 amino acids. Most preferably, the two sequences are aligned over their entire length.
  • the isolated or recombinant protein comprises an amino acid sequence which is at least 95%, more preferably at least 96%, more preferably at least 97%, more preferably at least 98%, more preferably at least 99%, more preferably at least 99.1%, more preferably at least 99.2%, more preferably at least 99.3%, more preferably at least 99.4%, more preferably at least 99.5%, more preferably at least 99.6%, more preferably at least 99.7%, more preferably at least 99.8%, and even more preferably at least 99.9% identical to the relevant nominated SEQ ID NO.
  • one residue is added to the nominated SEQ ID NO, one residue is deleted from the nominated SEQ ID NO, one residue is added and one residue is deleted compared to the nominated SEQ ID NO, two residues are added to the nominated SEQ ID NO, two residues are deleted from the nominated SEQ ID NO, one residue is changed from the nominated SEQ ID NO, two residues are changed from the nominated SEQ ID NO, one residue is changed and one residue is deleted from the nominated SEQ ID NO, or one residue is changed and one residue is added to the nominated SEQ ID NO, or any combination thereof.
  • the algorithm does not need to create a gap in a contiguous stretch of amino acids to obtain an optimal (highest % identity) alignment.
  • Amino acid sequence mutants of the isolated or recombinant protein contemplated for use in the method of the present disclosure can be prepared by introducing appropriate nucleotide changes into a nucleic acid of the present disclosure, or by in vitro synthesis of the desired polypeptide.
  • Such mutants include, for example, deletions, insertions or substitutions of residues within the amino acid sequence.
  • a combination of deletion, insertion and substitution can be made to arrive at the final construct, provided that the final polypeptide product possesses the desired characteristics.
  • Mutant (altered) polypeptides can be prepared using any technique known in the art.
  • a polynucleotide of the disclosure can be subjected to in vitro mutagenesis.
  • in vitro mutagenesis techniques include sub-cloning the polynucleotide into a suitable vector, transforming the vector into a "mutator" strain such as the E. coli XL-l red (Stratagene) and propagating the transformed bacteria for a suitable number of generations.
  • Products derived from mutated/altered DNA can readily be screened using techniques described herein to determine if they have receptor-binding and/or -inhibitory activity.
  • the location of the mutation site and the nature of the mutation will depend on characteristic(s) to be modified.
  • the sites for mutation can be modified individually or in series, e.g., by (1) substituting first with conservative amino acid choices and then with more radical selections depending upon the results achieved, (2) deleting the target residue, or (3) inserting other residues adjacent to the located site.
  • Amino acid sequence deletions generally range from about 1 to 15 residues, more preferably about 1 to 10 residues and typically about 1 to 5 contiguous residues.
  • Substitution mutants have at least one amino acid residue in the antibody and/or immunoglobulin chain molecule removed and a different residue inserted in its place.
  • the sites of greatest interest for substitutional mutagenesis include sites identified as important for antigen binding. These sites, especially those falling within a sequence of at least three other identically conserved sites of human antibodies and/or immunoglobulin chains, are preferably substituted in a relatively conservative manner. Such conservative substitutions are shown in Table 1 under the heading of "exemplary substitutions".
  • unnatural amino acids or chemical amino acid analogues can be introduced as a substitution or addition into the antibody and/or immunoglobulin chain of the present disclosure.
  • amino acids include, but are not limited to, the D-isomers of the common amino acids, 2,4-diaminobutyric acid, a-amino isobutyric acid, 4-aminobutyric acid, 2-aminobutyric acid, 6-amino hexanoic acid, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, b-alanine, fluoro-amino acids, designer amino acids such as a-methyl amino acids, Ca-methyl amino acids,
  • the isolated or recombinant protein contemplated for use in a method of the disclosure may comprise a heavy chain variable domain (VH) and a light chain variable domain (VL) of an anti-MK antibody described herein.
  • VH and the VL are provided in a single polypeptide chain.
  • the VH and the VL may be provided in separate polypeptide chains.
  • the isolated or recombinant protein of the disclosure may be provided in the form of:
  • the isolated or recombinant protein of the disclosure may be provided in the form of:
  • the antibody may be a chimeric, de-immunized, humanized or human antibody.
  • the isolated or recombinant protein may also comprise a human or non-human primate heavy chain immunoglobulin constant region selected from a group consisting of IgGl, IgG2, IgG3, IgG4, IgM, IgE and IgA.
  • an isolated or recombinant protein described herein is an immunoglobulin light chain variable region joined directly to an immunoglobulin light chain contant region described herein.
  • an immunoglobulin heavy chain variable region described herein is joined directly to an immunoglobulin heavy chain contant region described herein.
  • variable and constant regions of an immunoglobulin heavy or light chain can be joined as described by using standard recombinant DNA technology to create a polynucleotide (encoding the joined variable and constant domains) that can be expressed in a suitable host (to produce the said immunoglobulin chain(s)) or by using peptide chemistry to synthesise the joined variable and constant domains.
  • the isolated or recombinant protein is a humanised anti-MK antibody or binding fragment thereof
  • the humanised antibody or binding fragment will retain a significant proportion of the binding properties of the parent or precursor antibody or fragment.
  • Suitable humanised antibodies will retain the ability to specifically bind MK protein e.g., human MK and/or mouse MK, recognized by the parent or precursor antibody used to produce such antibodies.
  • a humanised antibody for use in a method of the disclosure exhibits substantially the same or improved binding affinity and avidity as the parent or precursor antibody.
  • the affinity (KD) of the antibody for midkine will be greater than the parent antibody affinity for midkine.
  • Binding affinity can be determined by association (Ka) and dissociation (Kd) rate.
  • Equilibrium affinity constant, K is the ratio of Ka/Kd.
  • Association (Ka) and dissociation (Kd) rates can be measured using surface plasmon resonance (SPR) (Rich and Myszka, (2000) Curr. Opin. Biotechnol. 11 :54; Englebienne P, (1998) Analyst. 123(7): 1599-1603). Instrumentation and methods for real time detection and monitoring of binding rates are known and are commercially available (BiaCore 2000, Biacore AB, ETpsala, Sweden; and Malmqvist M (1999), Biochem. Soc. Trans. 27:335-340). Methods for assaying binding affinity are well known in the art and include half-maximal binding assays, competition assays, and Scatchard analysis.
  • “avidity” relates to the overall strength of interaction between two molecules, such as an antibody and antigen. Avidity depends on both the affinity and the valency of interactions. Furthermore,“affinity” relates to the strength of the binding between a single binding site of a molecule (e.g., an antibody) and a ligand (e.g., an antigen).
  • the affinity of a molecule X for a ligand Y is represented by the dissociation constant (Kd), which is the concentration of Y that is required to occupy the combining sites of half the X molecules present in a solution. A smaller Kd indicates a stronger or higher affinity interaction, and a lower concentration of ligand is needed to occupy the sites.
  • An anti-MK antibody suitable for use in a method of the disclosure may also be a heteroconjugate antibody.
  • Heteroconjugate antibodies are composed of two covalently joined antibodies. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (ETS 4,676,980), and for treatment of HIV infection (W01991/000360; WO1992/200373; EP 586505). It is contemplated that the antibodies may be prepared in vitro using known methods in synthetic protein chemistry, including those involving crosslinking agents.
  • an antibody of the disclosure may be desirable to modify an antibody of the disclosure with respect to effector function, so as to enhance, e.g., the effectiveness of the antibody in treating a disorder described herein, such as cardiomyopathy or myocarditis.
  • cysteine residue(s) may be introduced into the Fc region, thereby allowing interchain disulfide bond formation in this region.
  • the homodimeric antibody thus generated may have improved internalization capability and/or increased complement-mediated cell killing and antibody-dependent cellular cytotoxicity (ADCC) (Caron et al, (1992) J. Exp. Med., 176(4): 1191-1195; Shopes B. (1992) J. Immunol., 148(9):2918-2922).
  • ADCC complement-mediated cell killing and antibody-dependent cellular cytotoxicity
  • Homodimeric antibodies with enhanced activity may also be prepared using heterobifunctional cross-linkers as described in Wolff et al. (1993).
  • an antibody can be engineered that has dual Fc regions and may thereby have enhanced complement lysis and ADCC capabilities (Stevenson et al, (1989) JAMA, 261 :884- 888).
  • Isolated or recombinant proteins for use in the method of the disclosure may be produced by the intervention of man e.g., as described herein.
  • the isolated or recombinant protein of the disclosure is “substantially purified” or “purified”.
  • substantially purified or purified we mean an isolated or recombinant protein e.g., an antibody or binding fragment thereof, that has been separated from one or more lipids, nucleic acids, other polypeptides, or other contaminating molecules with which it is associated in its native state. It is preferred that the substantially purified polypeptide is at least 60% free, more preferably at least 75% free, and more preferably at least 90% free from other components with which it is naturally associated.
  • substantially purified or “purified” means that the molecule that is the predominant species in the composition wherein it is found with respect to the class of molecules to which it belongs ii.e., it makes up at least about 50% of the type of molecule in the composition and typically will make up at least about 70%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, or more of the species of molecule, e. g., peptide, in the composition).
  • Isolated or recombinant proteins useful in a method of the present disclosure can be produced using methods available in the art, examples of which are described herein.
  • an isolated or recombinant protein useful in a method of the present disclosure is an antibody that binds the N-domain of MK.
  • Methods for generating antibodies are known in the art and/or described in Harlow and Lane (editors) Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, (1988).
  • a MK protein or immunogenic fragment or epitope thereof or a cell expressing and displaying same ii.e., an immunogen optionally formulated with any suitable or desired carrier, adjuvant, or pharmaceutically acceptable excipient, is administered to a non-human animal, for example, a mouse, chicken, rat, rabbit, guinea pig, dog, horse, cow, goat or pig.
  • the immunogen may be administered intranasally, intramuscularly, subcutaneously, intravenously, intradermally, intraperitoneally, or by other known route.
  • polyclonal antibodies may be monitored by sampling blood of the immunized animal at various points following immunization. One or more further immunizations may be given, if required to achieve a desired antibody titer. The process of boosting and titering is repeated until a suitable titer is achieved. When a desired level of immunogenicity is obtained, the immunized animal is bled and the serum isolated and stored, and/or the animal is used to generate monoclonal antibodies (Mabs).
  • Mabs monoclonal antibodies
  • Monoclonal antibodies are one exemplary form of MK-binding protein contemplated for use in a method of the present disclosure.
  • the term“monoclonal antibody” or“mAb” refers to a homogeneous antibody population capable of binding to the same antigen(s), for example, to the same epitope within the antigen. This term is not intended to be limited as regards to the source of the antibody or the manner in which it is made.
  • mAbs For the production of mAbs any one of a number of known techniques may be used, such as, for example, the procedure exemplified in US Patent No. 4,196,265 or Harlow and Lane (1988), supra.
  • a suitable animal is immunized with an immunogen under conditions sufficient to stimulate antibody producing cells.
  • Rodents such as rabbits, mice and rats are exemplary animals.
  • Mice genetically-engineered to express human immunoglobulin proteins and, for example, do not express murine immunoglobulin proteins, can also be used to generate an antibody which is suitable for use in a method of the present disclosure ( e.g ., as described in W02002/066630).
  • somatic cells with the potential for producing antibodies, specifically B lymphocytes (B cells), are selected for use in the mAb generating protocol. These cells may be obtained from biopsies of spleens, tonsils or lymph nodes, or from a peripheral blood sample. The B cells from the immunized animal are then fused with cells of an immortal myeloma cell, generally derived from the same species as the animal that was immunized with the immunogen.
  • Hybrids are amplified by culture in a selective medium comprising an agent that blocks the de novo synthesis of nucleotides in the tissue culture media.
  • agents are aminopterin, methotrexate and azaserine.
  • the amplified hybridomas are subjected to a functional selection for antibody specificity and/or titer, such as, for example, by flow cytometry and/or immunohistochemistry and/or immunoassay (e.g. radioimmunoassay, enzyme immunoassay, cytotoxicity assay, plaque assay, dot immunoassay, and the like).
  • immunoassay e.g. radioimmunoassay, enzyme immunoassay, cytotoxicity assay, plaque assay, dot immunoassay, and the like.
  • ABL-MYC technology NeoClone, Madison WI 53713, USA
  • MAbs e.g ., as described in Largaespada et al, (1996) J. Immunol. Methods. 197:85-95).
  • Antibodies can also be produced or isolated by screening a display library, e.g., a phage display library, e.g., as described in US Patent No. 6,300,064 and/or US Patent No. 5,885,793.
  • a display library e.g., a phage display library, e.g., as described in US Patent No. 6,300,064 and/or US Patent No. 5,885,793.
  • an isolated or recombinant protein useful in a method of the present disclosure is a chimeric antibody which binds the N-domain of MK.
  • the term“chimeric antibody” refers to antibodies in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequences in antibodies derived from a particular species (e.g., murine, such as mouse) or belonging to a particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequences in antibodies derived from another species (e.g., primate, such as human) or belonging to another antibody class or subclass.
  • chimeric antibodies utilize rodent or rabbit variable regions and human constant regions, in order to produce an antibody with predominantly human domains. Methods for producing chimeric antibodies are described in, e.g., US Patent No. 4,816,567; and US Patent No. 5,807,715.
  • the present disclosure also contemplates the use of a chimeric immunoglobulin, e.g., in which a variable region from one species is fused to a region of a protein from another species.
  • a chimeric immunoglobulin e.g., in which a variable region from one species is fused to a region of a protein from another species.
  • the disclosure contemplates the use of an immunoglobulin comprising a variable region from a T cell receptor of one species fused to a T cell receptor constant domain from a separate species.
  • an isolated or recombinant protein useful in a method of the present disclosure may be humanized or human antibody or protein that binds the N-domain of MK.
  • humanized antibody shall be understood to refer to a subclass of chimeric antibodies having an antigen binding site or variable region derived from an antibody from a non-human species and the remaining antibody structure of the molecule based upon the structure and/or sequence of a human antibody.
  • the antigen-binding site comprises the complementarity determining regions (CDRs) from the non-human antibody grafted onto appropriate FRs in the variable domains of a human antibody and the remaining regions from a human antibody.
  • Antigen binding sites may be wild type or modified by one or more amino acid substitutions. In some instances, FR residues of the human immunoglobulin are replaced by corresponding non-human residues.
  • human antibody refers to antibodies having variable regions (e.g. VH, VL) and, optionally constant regions derived from or corresponding to sequences found in humans, e.g. in the human germline or somatic cells.
  • the "human” antibodies can include amino acid residues not encoded by human sequences, e.g. mutations introduced by random or site directed mutations in vitro (in particular mutations which involve conservative substitutions or mutations in a small number of residues of the antibody, e.g., in 1, 2, 3, 4 or 5 of the residues of the antibody, e.g., in 1, 2, 3, 4 or 5 of the residues making up one or more of the CDRs of the antibody).
  • Human antibodies do not actually need to be produced by a human, rather, they can be produced using recombinant means and/or isolated from a transgenic animal (e.g., mouse) comprising nucleic acid encoding human antibody constant and/or variable regions (e.g., as described above).
  • Human antibodies can be produced using various techniques known in the art, including phage display libraries (e.g., as described in US Patent No. 5,885,793).
  • Human antibodies which recognize a selected epitope can also be generated using a technique referred to as "guided selection.”
  • a selected non-human monoclonal antibody e.g., a mouse antibody
  • is used to guide the selection of a completely human antibody recognizing the same epitope e.g., as described in US Patent No. 5,565,332.
  • an isolated or recombinant protein useful in a method of the present disclosure may be a de-immunized antibody or protein which binds the N-domain of MK.
  • De-immunized antibodies and proteins have one or more epitopes, e.g., B cell epitopes or T cell epitopes removed (i.e., mutated) to thereby reduce the likelihood that a mammal will raise an immune response against the antibody or protein.
  • epitopes e.g., B cell epitopes or T cell epitopes removed (i.e., mutated) to thereby reduce the likelihood that a mammal will raise an immune response against the antibody or protein.
  • Methods for producing de- immunized antibodies and proteins are known in the art and described, for example, in W02000/034317, W02004/108158 and W02004/064724.
  • an isolated or recombinant protein useful in a method of the present disclosure may be a heavy chain of an antibody which binds the N-domain of MK.
  • Heavy chain antibodies differ structurally from many other forms of antibodies, in so far as they comprise a heavy chain, but do not comprise a light chain. Accordingly, these immunoglobulins are also referred to as “heavy chain only antibodies”. Heavy chain immunoglobulins are found in, for example, camelids and cartilaginous fish (also called IgNAR).
  • variable regions present in naturally occurring heavy chain antibodies are generally referred to as "VHH domains" in camelid antibodies and V-NAR in IgNAR, in order to distinguish them from the heavy chain variable regions that are present in conventional 4- chain antibodies (which are referred to as "VH domains”) and from the light chain variable regions that are present in conventional 4-chain antibodies (which are referred to as "VL domains").
  • an isolated or recombinant protein useful in a method of the present disclosure is or comprises a single-domain antibody (which is used interchangeably with the term“domain antibody” or“dAb”) which binds the N-domain of MK.
  • a single-domain antibody is a single polypeptide chain comprising all or a portion of the heavy chain variable domain of an antibody.
  • a single-domain antibody is a human single- domain antibody (Domantis, Inc., Waltham, MA; see, e.g., US Patent No. 6,248,516).
  • an isolated or recombinant protein which binds the N-domain of MK useful in a method of the present disclosure is or comprises a diabody, triabody, tetrabody or higher order protein complex such as those described in WO 1998/044001 and/or WO 1994/007921.
  • a diabody is a protein comprising two associated polypeptide chains, each polypeptide chain comprising the structure VL-X-VH or VH-X-VL, wherein VL is an antibody light chain variable region, VH is an antibody heavy chain variable region, X is a linker comprising insufficient residues to permit the VH and VL in a single polypeptide chain to associate (or form an Fv) or is absent, and wherein the VH of one polypeptide chain binds to a VL of the other polypeptide chain to form an antigen binding site, i.e., to form a Fv molecule capable of specifically binding to one or more antigens.
  • the VL and VH can be the same in each polypeptide chain or the VL and VH can be different in each polypeptide chain so as to form a bispecific diabody (i.e., comprising two Fvs having different specificity).
  • a diabody, triabody, tetrabody, etc capable of inducing effector activity can be produced using an antigen binding domain capable of binding to IL-3Ra and an antigen binding domain capable of binding to a cell surface molecule on an immune cell, e.g., a T cell (e.g., CD3).
  • an immune cell e.g., a T cell (e.g., CD3).
  • an isolated or recombinant protein useful in a method of the present disclosure is or comprises a scFvs fragment which binds the N-domain of MK.
  • scFvs comprise VH and VL regions in a single polypeptide chain and a polypeptide linker between the VH and VL which enables the scFv to form the desired structure for antigen binding (i.e., for the VH and VL of the single polypeptide chain to associate with one another to form a Fv).
  • the linker comprises in excess of 12 amino acid residues with (Gly 4 Ser)3 being one of the more favoured linkers for a scFv.
  • the present disclosure also contemplates the use of a disulfide stabilized Fv (or diFv or dsFv), in which a single cysteine residue is introduced into a FR of VH and a FR of VL and the cysteine residues linked by a disulfide bond to yield a stable Fv.
  • a disulfide stabilized Fv or diFv or dsFv
  • the present disclosure encompasses the use of a dimeric scFv, i.e., a protein comprising two scFv molecules linked by a non-covalent or covalent linkage, e.g., by a leucine zipper domain (e.g., derived from Fos or Jun).
  • a leucine zipper domain e.g., derived from Fos or Jun.
  • two scFvs are linked by a peptide linker of sufficient length to permit both scFvs to form and to bind to an antigen, e.g., as described in US20060263367.
  • the present disclosure also contemplates the use of a dimeric scFv capable of inducing effector activity.
  • the dimeric protein is a combination of a dAb and a scFv.
  • bispecific antibody fragments capable of inducing effector function are described, for example, in US Patent No. 7,235,641.
  • an isolated or recombinant protein useful in a method of the present disclosure is produced by recombinant techniques.
  • a nucleic acid encoding same can be cloned into expression vectors, which are then transfected into host cells, such as E. coli cells, yeast cells, insect cells, or mammalian cells, such as simian COS cells, Chinese Hamster Ovary (CHO) cells, human embryonic kidney (HEK) cells, or myeloma cells that do not otherwise produce immunoglobulin protein.
  • host cells such as E. coli cells, yeast cells, insect cells, or mammalian cells, such as simian COS cells, Chinese Hamster Ovary (CHO) cells, human embryonic kidney (HEK) cells, or myeloma cells that do not otherwise produce immunoglobulin protein.
  • CHO Chinese Hamster Ovary
  • HEK human embryonic kidney
  • myeloma cells that do not otherwise produce immunoglobulin protein.
  • Exemplary cells used for expressing an immunoglobulin are CHO cells, myeloma cells or HEK cells.
  • nucleic acid is inserted operably-linked to a promoter in an expression construct or expression vector for further cloning (amplification of the DNA) or for expression in a cell-free system or in cells.
  • the term“promoter” is to be taken in its broadest context and includes the transcriptional regulatory sequences of a genomic gene, including the TATA box or initiator element, which is required for accurate transcription initiation, with or without additional regulatory elements (e.g., upstream activating sequences, transcription factor binding sites, enhancers and silencers) that alter expression of a nucleic acid, e.g., in response to a developmental and/or external stimulus, or in a tissue specific manner.
  • the term“promoter” is also used to describe a recombinant, synthetic or fusion nucleic acid, or derivative which confers, activates or enhances the expression of a nucleic acid to which it is operably-linked.
  • Exemplary promoters can contain additional copies of one or more specific regulatory elements to further enhance expression and/or alter the spatial expression and/or temporal expression of said nucleic acid.
  • operably-linked to means positioning a promoter relative to a nucleic acid such that expression of the nucleic acid is controlled by the promoter.
  • the vector components generally include, but are not limited to, one or more of the following: a signal sequence, a sequence encoding an immunoglobulin (e.g., derived from the information provided herein), an enhancer element, a promoter, and a transcription termination sequence.
  • a signal sequence e.g., a sequence encoding an immunoglobulin (e.g., derived from the information provided herein)
  • an enhancer element e.g., derived from the information provided herein
  • a promoter e.g., derived from the information provided herein
  • a transcription termination sequence e.g., a sequence encoding an immunoglobulin (e.g., derived from the information provided herein)
  • the skilled artisan will be aware of suitable sequences for expression of an immunoglobulin.
  • Exemplary signal sequences include prokaryotic secretion signals (e.g., pelB, alkaline phosphatase, penicillinase, Ipp, or heat-stable enterotoxin II), yeast secretion signals (e.g., invertase leader, a factor leader, or acid phosphatase leader) or mammalian secretion signals (e.g., herpes simplex gD signal).
  • prokaryotic secretion signals e.g., pelB, alkaline phosphatase, penicillinase, Ipp, or heat-stable enterotoxin II
  • yeast secretion signals e.g., invertase leader, a factor leader, or acid phosphatase leader
  • mammalian secretion signals e.g., herpes simplex gD signal.
  • Exemplary promoters active in mammalian cells include cytomegalovirus immediate early promoter (CMV-IE), human elongation factor l-a promoter (EF1), small nuclear RNA promoters (Lila and Ulb), a-myosin heavy chain promoter, Simian virus 40 promoter (SV40), Rous sarcoma virus promoter (RSV), Adenovirus major late promoter, b-actin promoter; hybrid regulatory element comprising a CMV enhancer/ b-actin promoter or an immunoglobulin promoter or active fragment thereof.
  • CMV-IE cytomegalovirus immediate early promoter
  • EF1 human elongation factor l-a promoter
  • EF1 small nuclear RNA promoters
  • SV40 Simian virus 40 promoter
  • RSV Rous sarcoma virus promoter
  • Adenovirus major late promoter b-actin promoter
  • hybrid regulatory element comprising a CMV enhancer/ b-actin promote
  • Examples of useful mammalian host cell lines are monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture; baby hamster kidney cells (BEK, ATCC CCL 10); or Chinese hamster ovary cells (CHO).
  • COS-7 monkey kidney CV1 line transformed by SV40
  • human embryonic kidney line (293 or 293 cells subcloned for growth in suspension culture
  • BEK baby hamster kidney cells
  • ATCC CCL 10 Chinese hamster ovary cells
  • Typical promoters suitable for expression in yeast cells such as for example a yeast cell selected from the group comprising Pichia pastoris, Saccharomyces cerevisiae and S. pombe , include, but are not limited to, the ADH1 promoter, the GAL1 promoter, the GAL4 promoter, the CUP1 promoter, the PH05 promoter, the nmt promoter, the RPR I promoter, or the TEF1 promoter.
  • Means for introducing the isolated nucleic acid or expression construct comprising same into a cell for expression are known to those skilled in the art. The technique used for a given cell depends on the known successful techniques. Means for introducing recombinant DNA into cells include microinjection, transfection mediated by DEAE-dextran, transfection mediated by liposomes such as by using lipofectamine (Gibco, MD, ETSA) transduction mediated by a viral delivery system, and/or cellfectin (Gibco, MD, ETSA), PEG-mediated DNA uptake, electroporation and microparticle bombardment such as by using DNA-coated tungsten or gold particles (Agracetus Inc., WI, ETSA) amongst others.
  • lipofectamine Gibco, MD, ETSA
  • PEG-mediated DNA uptake electroporation and microparticle bombardment
  • electroporation and microparticle bombardment such as by using DNA-coated tungsten or gold particles (Agracetus Inc., WI, ETSA)
  • the host cells used to produce the immunoglobulin may be cultured in a variety of media, depending on the cell type used.
  • Commercially available media such as Ham's F10 (Sigma), Minimal Essential Medium ((MEM), (Sigma), RPM1-1640 (Sigma), and Dulbecco's Modified Eagle's Medium ((DMEM), Sigma) are suitable for culturing mammalian cells.
  • Media for culturing other cell types discussed herein are known in the art.
  • MK-binding proteins used in accordance with the method of the present disclosure are preferably isolated, and, more preferably, provided in a substantially purified form. Methods for isolating and purifying antibodies and proteins are known in the art and/or described herein.
  • supernatants from such expression systems are generally first concentrated using a commercially available protein concentration filter, for example, an Amicon or Millipore Pellicon ultrafiltration unit.
  • a protease inhibitor such as PMSF may be included in any of the foregoing steps to inhibit proteolysis and antibiotics may be included to prevent the growth of adventitious contaminants.
  • the antibodies and antibody fragments prepared from the cells can be purified using, for example, ion exchange, hydroxyapatite chromatography, hydrophobic interaction chromatography, gel electrophoresis, dialysis, affinity chromatography ( e.g ., protein A affinity chromatography or protein G chromatography), or any combination of the foregoing. These methods are known in the art and described, for example in WO 1999/057134 or Ed Harlow and David Lane (editors) Antibodies: A Laboratory Manual, Cold Spring Harbour Laboratory, (1988).
  • a protein of the disclosure e.g., such as an antibody or fragment thereof, can be modified to include a tag to facilitate purification or detection, e.g., a poly-histidine tag, e.g., a hexa-histidine tag, or an influenza virus hemagglutinin (HA) tag, or a Simian Virus 5 (V5) tag, or a FLAG tag, or a glutathione S- transferase (GST) tag.
  • a tag to facilitate purification or detection e.g., a poly-histidine tag, e.g., a hexa-histidine tag, or an influenza virus hemagglutinin (HA) tag, or a Simian Virus 5 (V5) tag, or a FLAG tag, or a glutathione S- transferase (GST) tag.
  • HA hemagglutinin
  • V5 Simian Virus 5
  • FLAG tag e.g
  • an immunoglobulin comprising a hexa-his tag is purified by contacting a sample comprising the immunoglobulin with nickel-nitrilotriacetic acid (Ni-NTA) that specifically binds a hexa-his tag immobilized on a solid or semi-solid support, washing the sample to remove unbound antibodies, and subsequently eluting the bound antibodies.
  • Ni-NTA nickel-nitrilotriacetic acid
  • a ligand or antibody that binds to a tag may be used in an affinity purification method.
  • Isolated or recombinant proteins used in accordance with the present disclosure may be provided as conjugates of proteins e.g., antibodies or binding fragments thereof, as described herein according to any embodiment.
  • an isolate or recombinant protein of the disclosure can be modified to contain additional non-proteinaceous moieties that are known in the art and readily available.
  • the moieties suitable for derivatization of the protein are physiologically acceptable polymer, preferably a water soluble polymer.
  • Such polymers are useful for increasing stability and/or reducing clearance (e.g., by the kidney) and/or for reducing immunogenicity of a protein of the disclosure.
  • Non limiting examples of water soluble polymers include, but are not limited to, polyethylene glycol (PEG), polyvinyl alcohol (PVA), or propropylene glycol (PPG).
  • an isolated or recombinant protein as described herein according to any embodiment is conjugated or linked to another protein, including another protein of the disclosure or a protein comprising an antibody variable region, such as an antibody or a protein derived therefrom, e.g., as described herein.
  • Other proteins are not excluded. Additional proteins will be apparent to the skilled artisan and include, for example, an immunomodulator or a half-life extending protein or a peptide or other protein that binds to serum albumin amongst others. Exemplary serum albumin binding peptides or protein are described in US20060228364 or US20080260757.
  • an isolated or recombinant protein used in accordance with the present disclosure comprises one or more detectable markers to facilitate detection and/or isolation.
  • the compound comprises a fluorescent label such as, for example, fluorescein (FITC), 5,6-carboxymethyl fluorescein, Texas red, nitrobenz-2-oxa-l,3- diazol-4- yl (NBD), coumarin, dansyl chloride, rhodamine, 4'-6-diamidino-2- phenylinodole (DAPI), and the cyanine dyes Cy3, Cy3.5, Cy5, Cy5.5 and Cy7, fluorescein (5-carboxyfluorescein-N- hydroxysuccinimide ester), rhodamine (5,6- tetramethyl rhodamine).
  • FITC fluorescein
  • NBD nitrobenz-2-oxa-l,3- diazol-4- yl
  • DAPI nitrobenz-2-oxa-l,
  • the absorption and emission maxima, respectively, for these fluors are: FITC (490 nm; 520 nm), Cy3 (554 nm; 568 nm), Cy3.5 (581 nm; 588 nm), Cy5 (652 nm: 672 nm), Cy5.5 (682 nm; 703 nm) and Cy7 (755 nm; 778 nm).
  • the isolated or recombinant protein as described herein according to any embodiment is labelled with, for example, a fluorescent semiconductor nanocrystal (as described, for example, in US Patent No. 6,306,610).
  • the isolated or recombinant protein is labelled with, for example, a magnetic or paramagnetic compound, such as, iron, steel, nickel, cobalt, rare earth materials, neodymium-iron-boron, ferrous-chromium-cobalt, nickel-ferrous, cobalt- platinum, or strontium ferrite.
  • a magnetic or paramagnetic compound such as, iron, steel, nickel, cobalt, rare earth materials, neodymium-iron-boron, ferrous-chromium-cobalt, nickel-ferrous, cobalt- platinum, or strontium ferrite.
  • the isolated or recombinant protein must bind to the N-domain of MK and thereby inhibit or reduce MK activity and/or block transmission of a biological signal of MK. Accordingly, one or more functional assays may be performed to determine binding specificity and affinity of a candidate protein e.g., an antibody or fragment thereof, to the N-domain of human MK to thereby determine suitability of that protein for use in a method of the disclosure.
  • a candidate protein e.g., an antibody or fragment thereof
  • binding specificity and affinity of an antibody or binding protein to human MK may be assessed by ELISA.
  • the dissociation constant (Kd) of a candidate antibody may be determined.
  • the "Kd" or "Kd value" for a candidate protein or antibody may be measured by a radiolabelled MK binding assay (RIA) to determine its suitability for use in a method of the disclosure.
  • RIA radiolabelled MK binding assay
  • This assay equilibrates the test protein or antibody with a minimal concentration of radioactive MK protein in the presence of a titration series of unlabelled MK protein. Following washing to remove unbound MK protein, the amount of radioactivity is determined, which is indicative of the Kd of the test protein or antibody.
  • the“Kd” or“Kd value” is measured by using surface plasmon resonance assays, e.g., using BIAcore surface plasmon resonance (BIAcore, Inc., Piscataway, NJ) with immobilized IL-3Ra.
  • surface plasmon resonance assays e.g., using BIAcore surface plasmon resonance (BIAcore, Inc., Piscataway, NJ) with immobilized IL-3Ra.
  • a chemotaxis assay can be used to assess the ability of an isolated or recombinant protein of the disclosure to block binding of MK protein to a receptor thereof and/or inhibit function associated with binding of the MK to its receptor. These assays are based on the functional migration of cells in vitro or in vivo induced by a compound (chemoattractant). Chemotaxis can be assessed by any suitable means, for example, in an assay utilizing a 96-well chemotaxis plate, or using other art-recognized methods for assessing chemotaxis.
  • chemotaxis assays monitor the directional movement or migration of a suitable cell into or through a barrier (e.g., endothelium, a filter), toward increased levels of a compound, from a first surface of the barrier toward an opposite second surface.
  • a barrier e.g., endothelium, a filter
  • Membranes or filters provide convenient barriers, such that the directional movement or migration of a suitable cell into or through a filter, toward increased levels of a compound, from a first surface of the filter toward an opposite second surface of the filter, is monitored.
  • the membrane is coated with a substance to facilitate adhesion, such as ICAM-l, fibronectin or collagen.
  • Such assays provide an in vitro approximation of cell "homing”.
  • a suitable membrane having a suitable pore size for monitoring specific migration in response to compound, including, for example, nitrocellulose, polycarbonate, is selected.
  • pore sizes of about 3-8 microns, and preferably about 5-8 microns can be used. Pore size can be uniform on a filter or within a range of suitable pore sizes.
  • the distance of migration into the filter, the number of cells crossing the filter that remain adherent to the second surface of the filter, and/or the number of cells that accumulate in the second chamber can be determined using standard techniques (e.g., microscopy and flow cytometry).
  • the cells are labelled with a detectable label (e.g., radioisotope, fluorescent label, antigen or epitope label), and migration can be assessed in the presence and absence of a candidate antibody by determining the presence of the label adherent to the membrane and/or present in the second chamber using an appropriate method (e.g ., by detecting radioactivity, fluorescence, immunoassay).
  • a detectable label e.g., radioisotope, fluorescent label, antigen or epitope label
  • the extent of migration induced or inhibited can be determined relative to a suitable control (e.g., compared to background migration determined in the absence of the antibody, compared to the extent of migration induced by a second compound (i.e., a standard), compared with migration of untransfected cells induced by the antibody).
  • a suitable control e.g., compared to background migration determined in the absence of the antibody, compared to the extent of migration induced by a second compound (i.e., a standard), compared with migration of untransfected cells induced by the antibody.
  • a population of cells to which MK protein binds or which is capable if migrating to MK protein is placed in a chamber of a cell culture device that is in liquid communication with another chamber comprising MK protein (chemoattractant).
  • the two chambers are separated by a suitable membrane, e.g., a membrane that mimics the extracellular matrix found in a subject.
  • the amount of cell migration from one chamber to the other through the membrane is assessed in the presence or absence of candidate proteins or antibodies.
  • a protein or antibody that prevents or reduces the amount of MK-mediated cell migration compared to a control sample (containing no protein or antibody) is considered to have MK inhibitory activity.
  • in vivo assays may be employed to assess the usefulness of isolated or recombinant protein which binds the N-domain of MK in a method of the disclosure.
  • a candidate protein may be administered to a non-human mammal suffering from myocarditis e.g., such as the experimental autoimmune myocarditis (EAM) mouse model described in Example 1 or a viral infection-induced myocarditis mouse model as described in Fairweather and Rose (2007) Methods 41(1): 118-122.
  • screening may involve evaluating the effects of the candidate protein or antibody on cardiac muscle structure and/or function in the animal model of myocarditis e.g., as described in Example 1 or using other methods known in the art.
  • compositions or methods for administration of the isolated or recombinant proteins or conjugates of the disclosure to a mammal the isolated or recombinant protein or conjugate is combined with a pharmaceutically acceptable carrier, diluent and/or excipient, as is understood in the art.
  • a pharmaceutical composition comprising the isolated or recombinant protein or conjugate thereof combined with a pharmaceutically acceptable carrier, diluent and/or excipient.
  • the isolated or recombinant proteins or conjugates of this disclosure can be lyophilised for storage and reconstituted in a suitable carrier prior to use according to art- known lyophilisation and reconstitution techniques.
  • the disclosure provides a kit comprising a pharmaceutically acceptable carrier, diluent and/or excipient suitable for combining or mixing with the isolated or recombinant protein or conjugate prior to administration to the mammal.
  • a pharmaceutically acceptable carrier, diluent and/or excipient suitable for combining or mixing with the isolated or recombinant protein or conjugate prior to administration to the mammal.
  • the isolated or recombinant proteins or conjugates of this disclosure can be provided in a lyophilized form for combining or mixing with a pharmaceutically acceptable carrier, diluent and/or excipient prior to administration to the mammal.
  • the kit may further comprise instructions for use e.g., in accordance with a method of the disclosure.
  • carrier diluent or excipient
  • carrier diluent or excipient
  • carrier diluent or excipient
  • a solid or liquid filler binder, diluent, encapsulating substance, emulsifier, wetting agent, solvent, suspending agent, coating or lubricant that may be safely administered to any mammal, e.g., a human.
  • a variety of acceptable carriers, diluents or excipients known in the art may be used, as for example described in Remington's Pharmaceutical Sciences (Mack Publishing Co. N.J. USA, 1991).
  • the carriers, diluents or excipients may be selected from a group including sugars (e.g. sucrose, maltose, trehalose, glucose), starches, cellulose and its derivatives, malt, gelatine, talc, calcium sulphate, oils inclusive of vegetable oils, synthetic oils and synthetic mono- or di-glycerides, lower alcohols, polyols, alginic acid, phosphate buffered solutions, lubricants such as sodium or magnesium stearate, isotonic saline and pyrogen-free water.
  • the carrier, diluent or excipient is compatible with, or suitable for, parenteral administration.
  • Parenteral administration includes any route of administration that is not through the alimentary canal.
  • Non-limiting examples of parenteral administration include injection, infusion and the like.
  • administration by injection includes intravenous, intra-arterial, intramuscular and subcutaneous injection.
  • delivery by a depot or slow-release formulation which may be delivered intradermally, intramuscularly and subcutaneously, for example.
  • the present disclosure provides a method of treating and/or preventing myocarditis or cardiomyopathy in a subject, comprising administering to the subject an isolated or recombinant protein comprising an antigen binding domain of an antibody which binds specifically to an epitope located within the N-domain of MK protein and which inhibits or reduces a function of MK.
  • the aetiology of myocarditis can be due to a wide variety of injuries to the myocardium, including injury caused by toxins and drugs (e.g., cocaine and interleukin 2) or infectious agents, most commonly including viral (e.g., coxsackievirus, adenovirus, HIV and hepatitis C virus), bacterial (e.g., diphtheria, meningococcus, psittacosis and streptococcus), rickettsial (e.g., typhus and Rocky Mountain spotted fever), fungal (e.g., aspergillosis and candidiasis), and parasitic (Chagas disease, toxoplasmosis), as well as giant cell myocarditis, and hypersensitivity reactions to drugs such as antibiotics, sulfonamides, anticonvulsants, and anti-inflammatories. It is contemplated that the method of the disclosure will be suitable for treatment or prevention of myocardium, including
  • the myocarditis may be acute, subacute, or chronic, and there may be either focal or diffuse involvement of the myocardium, involving any or all cardiac chambers. Severe diffuse myocarditis can result in dilatation of all cardiac chambers and there may be mural thrombus formation in any chamber. With subacute and chronic myocarditis, interstitial fibrosis may replace fiber loss, and myofiber hypertrophy may be seen.
  • administering the isolated or recombinant protein to the subject prevents cardiac hypertrophy and/or prevents cardiac fibrosis and/or improves heart function in the subject.
  • administering the isolated or recombinant protein to the subject prevents development of cardiomyopathy.
  • the cardiomyopathy may be selected from hypertrophic cardiomyopathy, dilated cardiomyopathy, restrictive cardiomyopathy, arrhythmogenic right ventricular dysplasia, metabolic cardiomyopathy and unclassified cardiomyopathy.
  • the cardiomyopathy is hypertrophic cardiomyopathy.
  • the cardiomyopathy is dilated cardiomyopathy.
  • the cardiomyopathy is restrictive cardiomyopathy.
  • the cardiomyopathy is arrhythmogenic right ventricular dysplasia.
  • the cardiomyopathy is metabolic cardiomyopathy (e.g., diabetic cardiomyopathy, alcoholic cardiomyopathy, or drug-induced cardiomyopathy).
  • the cardiomyopathy is unclassified cardiomyopathy.
  • the method of the present disclosure may further comprise the step of identifying a subject in need of treatment e.g., a subject suffering from, or who is likely to suffer from, myocarditis or cardiomyopathy.
  • a subject in need of treatment e.g., a subject suffering from, or who is likely to suffer from, myocarditis or cardiomyopathy.
  • Methods for diagnosing myocarditis and cardiomyopathy are known in the art and contemplated for use herein.
  • an active agent e.g., a protein or conjugate of the disclosure
  • the appropriate dosage of an active agent will depend on the underlying cause of the myocarditis and/or cardiomyopathy to be treated, the severity and course of the myocarditis and/or cardiomyopathy, whether the active agent (e.g., a protein or conjugate of the disclosure) is administered for preventive or therapeutic purposes, previous therapy received by the patient (such as those described below under the heading“combination therapy”), the patient's clinical history and response to the active agent, and the discretion of the attending physician.
  • Some patients may have chronic and irreversible damage to the heart muscle requiring lifelong treatment, while other patients may require treatment for just a few months and then recover completely.
  • a therapeutically effective amount of the MK-binding protein or conjugate will be administered.
  • the phrase "a therapeutically effective amount” refers to an amount sufficient to promote, induce, and/or enhance treatment or other therapeutic effect in the subject being treated.
  • the therapeutically effective amount should be large enough to produce the desired effect but should not be so large as to cause adverse side effects.
  • the particular dosage regimen i.e., dose, timing, and repetition, will depend on the particular individual and that individual's medical history as assessed by a physician.
  • a clinician will administer an active agent (e.g ., MK-binding protein or conjugate comprising same) until a dosage is reached that achieves the desired result.
  • the dosage will vary with the age, condition, sex and extent of the disease, disorder and/or injury in the patient and can be determined by one of skill in the art. The dosage can be adjusted by the individual physician in the event of any complication.
  • normal dosage amounts may vary from about lOng/kg up to about lOOmg/kg of an individual's body weight or more per day. Exemplary dosages and ranges thereof are described herein.
  • the treatment can be sustained until a desired suppression of symptoms or treatment is achieved.
  • a MK-binding protein or conjugate as described herein is administered at an initial (or loading) dose of between about lmg/kg to about 30mg/kg, such as from about lmg/kg to about lOmg/kg, or about 2mg/kg or about 3mg/kg or 4mg/kg or 5mg/kg.
  • the MK-binding protein or conjugate can then be administered at a maintenance dose of between about O.OOOlmg/kg to about lmg/kg, such as from about 0.0005mg/kg to about lmg/kg, for example, from about O.OOlmg/kg to about lmg/kg, such as about 0.005mg/kg to about lmg/kg, for example from about 0. lmg/kg to about lmg/kg, such as about 0.2mg/kg or 0.3mg/kg or 0.4mg/kg or 0.5mg/kg.
  • the maintenance doses may be administered every 7-30 days, such as, every 10-15 days, for example, every 10 or 11 or 12 or 13 or 14 or 15 days.
  • Dosages for a particular MK-binding protein or conjugate may be determined empirically in mammals that have been given one or more administrations of the respective MK-binding protein or conjugate.
  • a clinical symptom of a disease, condition or injury being treated e.g., myocarditis and/or cardiomyopathy, can be monitored following administration.
  • efficacy of a dosage of MK-binding protein or conjugate of the disclosure in treatment of myocarditis and/or cardiomyopathy may be assessed based on one or more of the following criteria: the level of reduction in cardiac inflammation achieved; the level of improvement in heart function achieved; a return or approach to normal cardiac function or rhythm; prevention of or reduction in cardiac hypertrophy; and/or prevention of or reduction in cardiac fibrosis in the subject.
  • Administration of a MK-binding protein or conjugate or composition according to the methods of the present disclosure can be continuous or intermittent, depending, for example, on the recipient's physiological condition, whether the purpose of the administration is therapeutic or prophylactic, and other factors known to skilled practitioners.
  • the administration of a MK-binding protein or conjugate or composition may be essentially continuous over a preselected period of time or may be in a series of spaced doses.
  • routes of administration are possible including, but not necessarily limited to, oral, dietary, topical, parenteral (e.g ., intravenous, intraarterial, intramuscular, subcutaneous injection), inhalation (e.g., intrabronchial, intraocular, intranasal or oral inhalation, intranasal drops), depending on the underlying cause of the myocarditis and/or cardiomyopathy, the severity of the myocarditis and/or cardiomyopathy, and the particular patient.
  • parenteral e.g ., intravenous, intraarterial, intramuscular, subcutaneous injection
  • inhalation e.g., intrabronchial, intraocular, intranasal or oral inhalation, intranasal drops
  • Other suitable methods of administration can also include rechargeable or biodegradable devices and slow release polymeric devices.
  • the isolated or recombinant protein or conjugate or composition as described herein is administered in combination with one or more other compounds or agents useful for treating or preventing myocarditis and/or cardiomyopathy.
  • the specific combination will depend upon the underlying cause of the myocarditis and/or cardiomyopathy and upon the individual patient to be treated.
  • the isolated or recombinant protein or conjugate or composition as described may be conveniently administered in combination with one or more existing standard of care treatments for myocarditis and/or cardiomyopathy.
  • the isolated or recombinant protein or conjugate or composition as described herein may be administered in combination with antibiotics, heart protective agents, and antioxidant (such as high dose vitamin C, vitamin E and coenzyme Q10).
  • the isolated or recombinant protein or conjugate or composition as described herein may be administered in combination with drugs to reduce the heart’s workload (e.g., ACE inhibitors, Angiotensin III receptor blockers (ARBs) or beta blockers), drugs to eliminate excess fluid (e.g., diuretics) and/or intravenous medication to improve the heart pumping function.
  • drugs to reduce the heart’s workload e.g., ACE inhibitors, Angiotensin III receptor blockers (ARBs) or beta blockers
  • drugs to eliminate excess fluid e.g., diuretics
  • intravenous medication e.g., diuretics
  • the isolated or recombinant protein or conjugate or composition as described herein may be administered to a subject who has received or will receive a pump in the aorta (intra-aortic balloon pump) or a temporary artificial heart (e.g ., ventricular assist device (VAD)), and/or who is in need of urgent heart transplantation.
  • a pump in the aorta intra-aortic balloon pump
  • a temporary artificial heart e.g ., ventricular assist device (VAD)
  • VAD ventricular assist device
  • Other suitable treatment for myocarditis and/or cardiomyopathy which are contemplated are described in e.g., Maisch and Pankuweit (2013) Heart Fail. Rev. 18:761-795 and Fung et al, (2016) Circ. Res. 118:496-514, the full contents of which are incorporated herein.
  • the inventors evaluated the effects of intraperitoneally administered anti-midkine antibody on cardiac muscle structure and function in mice undergoing heart failure due to myocarditis.
  • the EAM mice then received one of the following treatments twice a week until day
  • mice were removed, rinsed with saline and fixed in paraformaldehyde 4% for at least 24 hours.
  • the pericardium, connective tissue and vascular remains were excised carefully before hearts were weighed using a microbalance (CP64-0CE, Sartorius, Gottingen, Germany). The hearts were then dehydrated in a graded series of ethanol concentrations, after which the tissues were embedded in paraffin. Cardiac sections were stained with Masson’s trichrome staining (day 63) to assess cardiac fibrosis.
  • fibrosis was divided into scores ranging from 0 to 5 using a semi -quantitative score (0: 0-1%; 1 : 1-2%; 2: 2- 3%; 3: 3-4% and 5: >4% fibrosis). All analyses were performed in a blinded manner.
  • Transthoracic echocardiography was conducted using a Vevo2l00 imaging system (VisualSonics, Toronto, Canada) with a 40 MHz transducer at day 63. Anesthesia was performed with 2% isoflurane. To ensure a stable body temperature in the range of 35.5— 36.5°C, mice were transferred to a heated platform (40°C). Parasternal long-axis M-mode was used to standardize mid-ventricular transducer positioning for short-axis M-mode imaging as previously described (Grabmaier et al (2014). PloS one , 9:e946l5). For evaluation of cardiac function, three consecutive cycles were measured and averaged.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Genetics & Genomics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Vascular Medicine (AREA)
  • Urology & Nephrology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Cardiology (AREA)
  • Molecular Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Endocrinology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

La présente invention concerne des procédés et des réactifs pour le traitement ou la prévention d'une myocardite et/ou d'une cardiomyopathie. Par exemple, le procédé et les réactifs de l'invention peuvent être utiles pour le traitement ou la prévention de l'hypertrophie cardiaque et/ou pour la prévention de la fibrose cardiaque et/ou pour l'amélioration de la fonction cardiaque chez un sujet souffrant d'une cardiomyopathie. En particulier, la présente invention concerne l'utilisation d'un anticorps isolé ou recombinant ou d'un fragment de liaison à l'antigène correspondant, qui se lie au domaine N-terminal de la protéine midkine (MK) et qui inhibe ou réduit la fonction de MK dans le traitement ou la prévention d'une myocardite et/ou d'une cardiomyopathie.
PCT/AU2019/050006 2018-01-09 2019-01-09 Procédés de traitement d'une myocardite et/ou d'une cardiomyopathie et réactifs correspondants WO2019136516A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AU2018900052 2018-01-09
AU2018900052A AU2018900052A0 (en) 2018-01-09 Methods of treating myocarditis and/or cardiomyopathy and reagents therefor

Publications (1)

Publication Number Publication Date
WO2019136516A1 true WO2019136516A1 (fr) 2019-07-18

Family

ID=67218151

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2019/050006 WO2019136516A1 (fr) 2018-01-09 2019-01-09 Procédés de traitement d'une myocardite et/ou d'une cardiomyopathie et réactifs correspondants

Country Status (1)

Country Link
WO (1) WO2019136516A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2020027089A (ja) * 2018-08-17 2020-02-20 国立大学法人埼玉大学 癌の診断用モノクローナル抗体及び癌診断キット
WO2021000004A1 (fr) * 2019-07-04 2021-01-07 Cellmid Limited Methodes de traitement d'une myocardite et/ou d'une cardiomyopathie et réactifs correspondants

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040219614A1 (en) * 2000-10-30 2004-11-04 Tomohiro Mitsumoto Monoclonal antibody specific to truncated midkine (tMK) protein and uses thereof
WO2012122590A1 (fr) * 2011-03-14 2012-09-20 Cellmid Limited Anticorps de reconnaissance de domaine n-terminal de midkine
WO2016058047A1 (fr) * 2014-10-14 2016-04-21 Cellmid Limited Anticorps anti-midkine améliorés
WO2017147653A1 (fr) * 2016-03-01 2017-09-08 Cellmid Limited Méthodes de traitement de maladies osseuses, de troubles et/ou de lésions et réactifs correspondants

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040219614A1 (en) * 2000-10-30 2004-11-04 Tomohiro Mitsumoto Monoclonal antibody specific to truncated midkine (tMK) protein and uses thereof
WO2012122590A1 (fr) * 2011-03-14 2012-09-20 Cellmid Limited Anticorps de reconnaissance de domaine n-terminal de midkine
WO2016058047A1 (fr) * 2014-10-14 2016-04-21 Cellmid Limited Anticorps anti-midkine améliorés
WO2017147653A1 (fr) * 2016-03-01 2017-09-08 Cellmid Limited Méthodes de traitement de maladies osseuses, de troubles et/ou de lésions et réactifs correspondants

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BRUNNER, S. ET AL.: "Blocking the N-terminal domain of midkine attenuates acute inflammation and prevents pathological remodelling and heart failure in experimental autoimmune myocarditis", EUROPEAN JOURNAL OF HEART FAILURE, vol. 18, no. 1, 2016, DOI: 10.1002/ejhf.539 *
HONDA, Y. ET AL.: "Midkine Deteriorates Cardiac Remodeling via Epidermal Growth Factor Receptor Signaling in Chronic Kidney Disease", HYPERTENSION, vol. 67, 2016, pages 857 - 865 *
JONES, D.R.: "Measuring midkine: the utility of midkine as a biomarker in cancer and other diseases", BRITISH JOURNAL OF PHARMACOLOGY, vol. 171, no. 12, 2014, pages 2925 - 2939, XP055626622 *
NETSU, S. ET AL.: "Midkine exacerbates pressure overload-induced cardiac remodeling", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 443, 2014, pages 205 - 210, XP028805830, DOI: 10.1016/j.bbrc.2013.11.083 *
WECKBACH, L.T. ET AL.: "Midkine In Inflammation", THE SCIENTIFIC WORLD JOURNAL, vol. 11, 2011, pages 2491 - 2505, XP055626618, DOI: 10.1100/2011/517152 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2020027089A (ja) * 2018-08-17 2020-02-20 国立大学法人埼玉大学 癌の診断用モノクローナル抗体及び癌診断キット
WO2021000004A1 (fr) * 2019-07-04 2021-01-07 Cellmid Limited Methodes de traitement d'une myocardite et/ou d'une cardiomyopathie et réactifs correspondants

Similar Documents

Publication Publication Date Title
JP4754219B2 (ja) 腫瘍壊死因子を対象とする抗体、およびそれらの使用
KR101954906B1 (ko) FcRn 특이적 인간 항체 및 이를 포함하는 자가면역질환 치료용 조성물
JP2021531826A (ja) 栄養膜細胞表面抗原2(trop2)に対する特異的な抗体
JP2020127430A (ja) 抗プロ/潜在型−ミオスタチン抗体およびその使用
CN114929278A (zh) 抗ccr8抗体及其用途
JP2021527110A (ja) CD47とSIRPaの相互作用を遮断できる抗体及びその応用
CN103153331B (zh) 靶向于钙粘着蛋白‑11的ec1结构域的人源化抗体及相关组合物和方法
US9884915B2 (en) Antibodies against CCR9 and methods of use thereof
US20190382483A1 (en) NEW USES OF ANTI-SIRPg ANTIBODIES
JP2018535948A (ja) レプチン受容体を活性化させる抗原結合タンパク質
CN110494445A (zh) 抗α-SYN抗体及其用途
CN114685666B (zh) 抗间皮素纳米抗体及其应用
JP2021531825A (ja) 葉酸受容体アルファに特異的な抗体
CN115109156A (zh) 一种靶向bcma的纳米抗体及其应用
CN114853890B (zh) 一种prlr抗原结合蛋白及其制备方法和应用
WO2022143550A1 (fr) Molécule de liaison à la mésothéline et son utilisation
JP2022514786A (ja) Muc18に特異的な抗体
WO2019136516A1 (fr) Procédés de traitement d'une myocardite et/ou d'une cardiomyopathie et réactifs correspondants
WO2021000004A1 (fr) Methodes de traitement d'une myocardite et/ou d'une cardiomyopathie et réactifs correspondants
WO2017122666A1 (fr) Anticorps anti-myl9
AU2022210221A1 (en) Immunomodulatory antibodies and uses thereof
WO2016143702A1 (fr) Anticorps contre l'adam28 humain de type membranaire
KR20220050182A (ko) 항-cd22 항체 및 그의 용도
CN118027202B (zh) 结合slc13a5膜蛋白的抗体
WO2022143552A1 (fr) Molécule de liaison pd-1 et son application

Legal Events

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

Ref document number: 19738503

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19738503

Country of ref document: EP

Kind code of ref document: A1