CA2624976A1 - Anti-myostatin antibodies - Google Patents

Anti-myostatin antibodies Download PDF

Info

Publication number
CA2624976A1
CA2624976A1 CA002624976A CA2624976A CA2624976A1 CA 2624976 A1 CA2624976 A1 CA 2624976A1 CA 002624976 A CA002624976 A CA 002624976A CA 2624976 A CA2624976 A CA 2624976A CA 2624976 A1 CA2624976 A1 CA 2624976A1
Authority
CA
Canada
Prior art keywords
antibody
myostatin
seq
monoclonal antibody
antibodies
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002624976A
Other languages
French (fr)
Inventor
Bomie Han
Andrew Korytko
Pamela Jean Mitchell
Rosamund Carol Smith
O'bryan (Nee Tobias), Linda
Rong Wang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Eli Lilly and Co
Original Assignee
Eli Lilly And Company
Bomie Han
Andrew Korytko
Pamela Jean Mitchell
Rosamund Carol Smith
O'bryan (Nee Tobias), Linda
Rong Wang
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Eli Lilly And Company, Bomie Han, Andrew Korytko, Pamela Jean Mitchell, Rosamund Carol Smith, O'bryan (Nee Tobias), Linda, Rong Wang filed Critical Eli Lilly And Company
Publication of CA2624976A1 publication Critical patent/CA2624976A1/en
Abandoned legal-status Critical Current

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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/90Immunoglobulins specific features characterized by (pharmaco)kinetic aspects or by stability of the immunoglobulin
    • C07K2317/92Affinity (KD), association rate (Ka), dissociation rate (Kd) or EC50 value
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/582Recycling of unreacted starting or intermediate materials

Abstract

A neutralizing epitope is identified within amino acids 40-64 of the mature form of human myostatin. Antibodies that bind this epitope with high affinity preferentially bind GDF-8 over GDF-11 and may be chimeric, humanized or fully human antibodies, immunoconjugates of the antibodies or antigen-binding fragments thereof. The antibodies of the invention are useful for increasing muscle mass, increasing bone density, or for the treatment of various disorders in mammalian and avian species.

Description

DEMANDE OU BREVET VOLUMINEUX

LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVET COMPREND
PLUS D'UN TOME.

NOTE : Pour les tomes additionels, veuillez contacter le Bureau canadien des brevets JUMBO APPLICATIONS/PATENTS

THIS SECTION OF THE APPLICATION/PATENT CONTAINS MORE THAN ONE
VOLUME

NOTE: For additional volumes, please contact the Canadian Patent Office NOM DU FICHIER / FILE NAME:

NOTE POUR LE TOME / VOLUME NOTE:

ANTI-MYOSTATIN ANTIBODIES
FIELD OF THE INVENTION

The invention is in the field of medicine, particularly in the field of monoclonal antibodies against myostatin. More specifically the invention relates to high affinity chimeric, humanized or human anti-myostatin antibodies that preferentially bind myostatin over GDF-1-1 and use of the antibodies for therapy, prophylaxis or diagnosis of various disorders or conditions in mammalian and avian species.
BACKGROUND OF THE INVENTION

Members of the transforming growth factor beta (TGF-(3) superfamily of proteins are involved in embryonic development and adult tissue homeostasis. The TGF-(3 superfamily members share a common structure including a peptide signal sequence required for secretion of the protein and an amino-terminal fragment that is proteolytically cleaved about 105-140 amino acids from the carboxy-terminus of the large precursor protein to produce the mature protein. The mature protein is characterized by highly .conserved cysteine residues, while the active form of the mature protein is a disulfide-linked homodimer of the proteolytically-cleaved proprotein (Gray, A., and Maston, A., Science, 247:1328, 1990).
Myostatin, also referred to as growth differentiation factor-8 (GDF-8), is a member of the TGF-(3 superfamily of proteins. Myostatin shares structural similarities with other TGF-(3 family members. It contains a hydrophobic amino-terminus that acts as a secretory signal and a conserved RSRR domain that is important for proteolytic processing. Cleavage of the protein gives rise to an amino-terminal latency associated peptide and a carboxy-terminal mature signaling peptide which forms the biologically active homodimer. Myostatin is expressed largely in developing and adult skeletal muscle and functions as a negative regulator of skeletal muscle. Systemic over-expression of myostatin in adult mice leads to muscle wasting (Zimmers, et al., Science, 296:1486-1488, 2002) while conversely, a myostatin knock-out mouse is characterized by hypertrophy and hyperplasia of the skeletal muscle resulting in two- to threefold greater muscle mass than their wild type littermates and a decrease in fat accumulation (McPherron, et al. Nature, 387:83-90, 1997). A human with a myostatin knock-out mutation was reported to be associated with gross muscle hypertrophy (Scheulke, et al., New Eizg. J. Med. 350:2682, 2004).
There are presently limited treatments available for muscle wasting or for disorders or conditions which would benefit from an increase in muscle mass and/or muscle strength including, for example, rimuscular dystrophy, frailty, disuse atrophy and, cachexia, as well as disorders which are associated with muscle wasting, for exainple, renal disease, cardiac failure or disease, and liver disease. Due to its role as a negative regulator of skeletal muscle growth, myostatin is a desirable target for therapeutic or prophylactic intervention for such disorders or conditions or for monitoring progression of such disorders or conditions. Apart from its direct role in skeletal muscle regulation, myostatin may also be involved in other physiological processes including preadipocyte differentiation to adipocytes (Kim et al. BBRC, 281:902-906, 2001), and, indirectly, with glucose homeostasis (McPherron, A and Lee S-J. JCI 109:595, 2002) and inhibition of bone formation (Hamrick, M. Mol. Cell Evol. Biol. 272 388-91, 2003; Hamrick et al.
Calcif Tissue Int. 71:63, 2002). Therefore, myostatin-specific antagonists, e.g., myostatin-specific antibodies, may also prove useful for treating, preventing or monitoring disorders or conditions such as those which benefit from increasing bone density (e.g., osteoporosis), Type II diabetes, metabolic syndrome, obesity and osteoarthritis.
Myostatin is highly conserved across species; the amino acid sequence of the mature form of myostatin in human, mouse, rat, chicken, turkey and cow are 100%
identical (See Figs. 2 and 3). There are naturally occurring myostatin mutations in cattle, which have been linked to a double-muscled phenotype (McPherron, et al. PNAS, 94:12457-61, 1997). Since myostatin is highly conserved in sequence and in function across species, not only does an anti-myostatin antibody provide a promising means of increasing muscle mass, or treatment or prevention of such disorders or conditions listed above in humans, but also in other mammals including, e.g., domestic animals (e.g., canine and feline), sports animals (e.g., equine), food-source animals (e.g., bovine, porcine and ovine) and in avian species (e.g., chicken, turkey, duck and other game birds and poultry).
Growth differentiation factor-11, also referred to as GDF-11 or BMP-1:1, is the member of the TGF-(3 superfamily of proteins that is most homologous to myostatin. The amino acid sequence of the mature forms of human myostatin and GDF- 11 are about 90%
identical; however, GDF-11 is expressed in a wider range of tissues than is including dental pulp, brain, heart, kidney and lung as well as muscle and adipose tissue (Nakashima, et al. Mech. of Developfnent 80:185, 1999). GDF-11 knock-out mice die within 24 hours of birth with multiple abnormalities. In particular, the mice exhibit extra pairs of ribs, lack kidneys and show defects in the stomach, spleen and pancreas (McPherron et al., Nature Genetics 22:260, 1999; Esquela and Lee, Dev. Biol.
257:356, 2003; Harmon et al., Devpt. 131:6163, 2004). Human GDF-11 has recently been found to govern the temporal windows-during-which mulfipotent-progenitors retain competence to produce distinct neural progeny (Kim, J. et al. Science 308:1927-1930, 2005).
There is a therapeutic need to specifically inhibit a myostatin activity while not inhibiting, or minimally inhibiting, an activity of other TGF-P superfamily proteins, particularly GDF-11,-in order to minimize the possibility of undesirable side effects resulting from binding of the myostatin antagonist to another TGF-(3 superfamily protein.
Furthermore, there is a diagnostic need for an anti-myostatin antibody that does not cross-react, or minimally cross-reacts, with another TGF-(3 superfamily protein, particularly GDF- 11, in order to more accurately monitor or determine myostatin levels in a sample.
Furthermore, there is a need for myostatin-specific antibodies which specifically and preferentially bind myostatin with a high affinity and thereby allow the dosage level that patients receive to be minimized which may thereby result in less frequent dosing with such an antibody than with an antibody that binds myostatin with a lesser affinity (i.e., a higher KD). A high affinity antibody is also desirable in that it may allow for more flexibility in the route of administration of the antibody to a patient since it is less -desirable for a drug to be administered intravenously than subcutaneously for example.
There is also a need for myostatin-specific antibodies with a low or otherwise favorable IC50 value in a myostatin bioactivity assay in order to generate a therapeutic anti-myostatin antibody with a minimum effective therapeutic dose. It is also desirable to provide antibodies specific to myostatin where any immune response to the antibody evoked by a patient receiving the antibody is reduced to a minimum. The present invention satisfies these needs and provides related advantages.
SUMMARY OF THE INVENTION

Antibodies of the invention are chimeric, humanized, or fully human anti-myostatin monoclonal antibodies, and antigen-binding portions thereof, that antagonize or neutralize at least one in vitro or in vivo biological activity or property associated with myostatin or a portion thereof. , In one embodiment, antibodies of the invention that specifically and/or preferentially bind myostatin are significantly less reactive with GDF-11 than with myostatin, i.e., a monoclonal antibody of the invention binds myostatin at least about 2, 3, 5, 10, 20, 22, 24, or 25-times greater than it binds GDF-11 as measured by a technique in the art e.g., by competition ELISA, or by BIACORE or KINEXA assay to demonstrate higher affinity (i.e., lower KD) of the antibody to GDF-8 than GDF-11. Most preferably the antibodies of the invention, when expressed as Fabs, do not bind GDF-11 above background levels in any binding assay available in the art. Preferably antibodies of the invention specifically bind myostatin, within the domain spanning amino acids [ANYCSGECEFVFLQKYPHTHLVHQA for human], 43-57 [CSGECEFVFLQKYPH
OR CSGESEFVFLQKYPH for human] and/or 45-59 [GECEFVFLQKYPHTH for human] of mature myostatin or they specifically bind a polypeptide consisting of amino acids 40-64, 43-57 and/or amino acids 45-59 of mature myostatin.
In one embodiment, antibodies of the invention have an IC50 of less than, or equal to, about 25 nM, 20 nM, 16 nM, 14 nM, 10 nM, 9 nM, 6 nM or 5.2 nM in an in vitro myostatin/SBE reporter assay (see Example 6). Preferably, such antibodies of the invention are further characterized in that they are significantly less reactive with GDF- 11 than with GDF-8, i.e., binds myostatin at least about 2, 3, 5, 10, 20, 22, 24, or 25-times greater than it binds GDF-11 as measured by a technique in the art e.g., by competition ELISA, or by BIACORE or KINEXA assay to demonstrate higher affinity (i.e., lower KD) of the antibody to GDF-8 than GDF-11. and even more preferably they bind myostatin within the domain spanning amino acids 40-64, 43-57 and/or 45-59 of mature myostatin or they bind a polypeptide consisting of amino acids 40-64, 43-57 and/or 45-59 of mature myostatin.

In one embodiment, antibodies of the invention are characterized by a strong binding affinity (KD) for myostatin, i.e., less than about 3 x 10-$ M, 1 x 10-8 M or 1 x 10-9 M, preferably less than about 9 x 10-1 M, 8.7 x 10-1 M or most. preferably, less than about 8 x 10-11 M. Alternatively, the antibodies of the invention are characterized by a KD for 5 myostatin of no greater than about 3 x 10"8 M, 1 x 10-8 M, 1 x 10"9 M or 9 x 10-10 M, more preferably no greater than about 8.7 x 10-10 M and most preferably no greater than about 8 x 10"1 i M. Preferably, antibodies of the invention characterized by a strong binding affinity as described above also have an IC50 of less than 25 nM, 20 nM, 16 nM, 14 nM, nM, 9 nM, 6 nM, or 5.2 nM in an in vitro myostatin/SBE reporter assay and/or are 10 significantly less reactive with GDF-11 than with GDF-8. Even more preferably they bind myostatin within the domain spanning amino acids 40-64, 43-57 and/or 45-59 of mature myostatin and/or they bind a polypeptide consisting of amino acids 40-64, 43-57 and/or 45-59 of mature myostatin.
In another embodiment, an anti-myostatin monoclonal antibody of the invention comprises a light chain variable 'region ("LCVR") polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NOs: 5-12. In another embodiment, an anti-myostatin monoclonal antibody of the invention comprises a heavy chain variable region ("HCVR") polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NOs: 13-26, 55 and 56. The sequences associated with each SEQ ID Number are shown in Figures 5-9 herein.
In another embodiment, a monoclonal antibody of the invention is one which can compete for binding to human myostatin with a competing antibody as demonstrated by an available assay in the art (e.g., a competition ELISA), wherein the competing antibody comprises two polypeptides with the sequences selected from the group consisting of: (i) SEQ ID NOs: 5 and 15, (ii) SEQ ID NOs: 5 and 16, and (iii) SEQ ID NOs: 10 and 26.
Preferably, an antibody of the invention which competes with the competing antibody defined above is further characterized by preferentially binding GDF-8 over GDF-1 1;
even more preferably they bind myostatin within the domain spanning amino acids 40-64, 43-57 and/or 45-59 of mature myostatin and/or they bind a polypeptide consisting of amino acids 40-64, 43-57 and/or 45-59 of mature myostatin. Preferably, an antibody of the invention which competes with the competing antibody defined above is further characterized by having a KD for myostatin of no greater than about 3 x 10-8 M, 1 x 10-8 M, 1 x 10"9 M or 9 x 10"10 M, 8.7 x 10-10 M or 8 x 10'11 M. 'Preferably, an antibody of the invention which competes with the competing antibody defined above is further ciiaracterized by having an IC50 of less than 25 nM, 20 nM, 16 nM, 14 nM, 10 nM, 9 nM, 6 nM, or 5.2 nM in an in vitro myostatin/SBE reporter assay.
In one embodiment,'an anti-myostatin antibody of the invention has a heavy and a light chain variable region, wherein the heavy chain variable region comprises CDR
regions with the following amino acid sequences: CDRH1 (SEQ ID NO: 59), CDRH2 (SEQ ID NO: 60) and CDRH3 (SEQ ID NO: '61); and/or wherein the light chain variable region comprises CDR regions with the following amino acid sequences: CDRLl (SEQ' ID NO: 57), CDRL2 (SEQ ID NO: 30) and CDRL3 (SEQ ID NO: 58).
An anti-myostatin monoclonal antibody of the invention may further comprise a heavy chain constant region selected from the group consisting of human (or substantially of human origin) IgGI, IgG2, IgG3, IgG4, IgA, IgE, IgM and IgD, preferably IgGI or IgG4.
An anti-myostatin monoclonal antibody of the invention may further comprise a human kappa or lambda light chain constant region. When the antibody is to be used as a therapeutic in a human, the constant region is preferably substantially of human origin.
When the antibody is to be used as a therapeutic in a non-human animal, or egg of a non-human animal, the constant region preferably substantially originates from the animal in which the antibody is to be used as a therapeutic. (see, e.g., Clarkson, C. et al., Mol. Imm.
30:1195-1204, 1993; U.S. application number 2002/01651350; and Genbank accession numbers X69797, U03778, X16701, X07174, AB016711).
Various forms of the antibody are contemplated herein. For example, an anti-myostatin monoclonal antibody of the invention may conlprise or consist of an intact antibody (i.e., full-length, having an intact Fc region), a substantially intact antibody, an antigen-binding portion thereof (e.g., a Fab, Fab', F(ab')2) or a single chain Fv fragment.
It is understood that all such forms of the antibodies are encompassed herein and throughout within the term "antibody." Furthermore, an antibody of the invention may be labeled with a detectable label, immobilized on a solid phase and/or conjugated with a heterologous compound, e.g., an enzyme or polyethylene glycol molecule.
Furthermore, antibodies of the invention are contemplated to be "monoclonal" even though they may differ in glycosylation pattern.
Diagnostic, therapeutic and prophylactic uses for the monoclonal antibodies of the invention are contemplated herein. In one diagnostic application, the invention provides a method for determining the presence and/or quantity of myostatin protein comprising exposing a test sample suspected of containing the myostatin protein to an anti-myostatin antibody of the invention and determining specific binding of the antibody to the sample.
An anti-myostatin antibody of the invention may be used to determine the levels of myostatin in test samples by comparing test sample values to a standard curve generated by binding said antibody to samples with known amounts of myostatin using any method available in the art, e.g., an ELISA assay. The invention further provides.a kit comprising an antibody of the invention and, preferably, instructions for using the antibody to detect myostatin protein in e.g., a test sarnple.
In another embodiment, the invention provides a pharmaceutical composition comprising an anti-myostatin monoclonal antibody of the invention. The pharmaceutical con7position of the invention may further comprise a pharmaceutically acceptable carrier.
In said pharmaceutical composition, the anti-myostatin monoclonal antibody of the invention is the active ingredient. Preferably the pharmaceutical composition comprises a homogeneous or substantially homogeneous population of an anti-myostatin monoclonal antibody of the invention. The composition for therapeutic use is sterile and may be lyophilized, preferably supplied with an appropriate diluent.
The invention provides a method of inhibiting at least one myostatin biological activity in an animal, preferably a mammalian or avian species, preferably a human, in need thereof, comprising administering a therapeutically effective amount, or prophylactically effective amount, or myostatin-neutralizing or myostatin-inhibiting amount of an anti-myostatin monoclonal antibody of the invention to said mammalian or avian species. The invention further provides a method of enhancing muscle mass or treating or preventing a disease or disorder or condition ameliorated by neutralizing or antagonizing a myostatin bioactivity that comprises administering to a patient (e.g., a human) in need of such treatment or prevention a therapeutically or prophylactically effective amount of a monoclonal antibody of the invention.
The invention embodies an anti-myostatin monoclonal antibody of the invention for use in the manufacture of a medicament for administration to a mammal, preferably a human, for the treatment of e.g., frailty, cachexia, age-related sarcopenia, muscle wasting or weakness, myopathy, muscular dystrophy, osteoporosis, obesity, COPD, renal failure or disease, liver failure or disease, cardiac failure or disease, metabolic syndrome and Type II diabetes in a mammal, preferably a human, in need thereof by administering to said mammal a therapeutically effective or prophylactically effective amount of an anti-myostatin monoclonal antibody of the invention.
The invention embodies an article of manufacture comprising a packaging material and an antibody of the invention'contained within said packaging material and wherein the packaging material comprises a package insert which preferably indicates that the antibody specifically neutralizes or antagonizes a myostatin activity or decreases the ' level of myostatin. Optionally, the package insert further indicates that the antibody preferentially-neutralizes or antagonizes a myostatin activity with respect to a GDF- 11 activity or preferentially decreases the level of myostatin with respect to decreasing the level of GDF- 11 by preferentially binding myostatin with respect to binding GDF- 11.
The invention further provides isolated nucleic acid encoding an antibody of the invention; a vector (or vectors) comprising that nucleic acid, optionally operably linked to control sequences recognized by a host cell transformed with the vector; a host cell comprising that vector; a process for producing an antibody of the invention comprising culturing the host cell so that the nucleic acid is expressed and, optionally, recovering the antibody from the host cell medium.
BRIEF DESRIPTION OF THE DRAWINGS
FIG. 1 shows the amino acid sequence of human promyostatin with the signal sequence underlined and the portion of the protein at the carboxy-terminus that makes up a monomer of the mature form of myostatin in bold letters.
FIG. 2 shows the amino acid sequence of the monomeric human mature myostatin.
Active human myostatin is a homodimer of this polypeptide associated by disulfide bonds. The antigenic epitope of the present invention is underlined. This sequence is identical to that of the mature myostatin in mouse, rat, chicken, turkey, dog, horse, and pig.
FIG. 3 shows an alignment of the amino acid sequence of mature myostatin of various mammalian and avian species.
FIG. 4 shows the alignment of the amino acid sequence of the mature form of human myostatin and human GDF- 11 with the antigenic epitope of the present invention underlined and the residues within the antigenic epitope that differ between myostatin and GDF-11 in bold print.
FIG: 5 shows the alignment of the amino acid sequence of HCVRs with a VH2-70 framework. The CDR domains are in bold print.
FIG. 6 shows the alignment of the amino acid sequence of HCVRs with a VH4-39 framework. The CDR domains are in bold print.
FIGS. 7 and 8 show the alignment of the amino acid sequence of LCVRs with an 02 framework. The CDR domains are in bold print.
----- . FIG: 9 shows the amino acid sequence of CDRs of the LCVR and HCVR of various antibodies of the invention.
FIG. 10 shows the amino acid sequence of the LCVR of various murine anti-myostatin antibodies, any of which may be. an exemplary parent molecule for antibodies of the invention. See U.S. patent applications 60/559,621 and 60/555,456 incorporated herein.
FIG. 11 shows the amino acid sequence of the HCVR of various murine anti-myostatin antibodies, any of which may be an exemplary parent molecule for antibodies of the invention. See U.S. patent applications 60/559,621 and 60/555,456.
DETAILED DESCRIPTION OF THE INVENTION

The invention presents anti-myostatin monoclonal antibodies able to specifically bind to manunalian or avian myostatin (proprotein or mature protein or a portion thereof;
monomeric or homodimeric), wherein the antibodies are further characterized as being chimeric, humanized or fully human antibodies or antigen-binding portions thereof able to neutralize or antagonize at least one myostatin activity in vitro and/or in vivo; preferably demonstrating an IC501ess than about 25 nM, 20 nM, 16 nM, 14 nM, 10 nM, 9 nM, nM, or 5.2 nM in a myostatin/SBE reporter assay and/or preferably demonstrating a high binding affinity with myostatin. The antibodies of the invention are further characterized in that they bind myostatin within the domain spanning the amino acids at residues 40-64 of the mature form of myostatin (e.g., SEQ ID NO: 53) and are significantly less reactive with myostatin's nearest homologue, GDF-11.

Definitions 5 When used herein, the term "mature myostatin" (see SEQ ID NO: 2 for human, murine, rat, chicken, turkey, canine, equine and porcine species) refers to the monomeric or the homodimeric form of the protein resulting after proteolytic cleavage at Arg 266 of the 375 amino acid proprotein form of myostatin. When used herein, the term "myostatin" refers to mature myostatin. When used herein, the term "promyostatin" or 10 "proprotein form of myostatin" when used with reference to the human protein refers to a protein comprising-the sequence shown in SEQ ID NO: 1 either as a monomer or homodimer.
A full-length antibody as it exists naturally is an immunoglobulin molecule comprised of four peptide chains, two heavy (H) chains (about 50-70 kDa when full length) and two light (L) chains (about 25 kDa when full length) interconnected by disulfide bonds. The amino terminal portion of each chain includes a variable region of about 100-110 or more amino acids primarily responsible for antigen recognition. The carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function.
Light chains are classified as kappa or lambda and characterized by a particular constant region as known in the art. Heavy chains are classified as gamma, mu, alpha, delta, or epsilon, and define the antibody's isotype as IgG, IgM, IgA, IgD, and IgE, respectively and several of these may be further divided into subclasses (isotypes) e.g., IgGI, IgG2, IgG3, IgG4, IgAj, and IgA2. Each heavy chain type is characterized by a particular constant region known in the art. The subunit structures and three-dimensional configurations of different classes of antibodies are well known in the art.
Each heavy chain is comprised of an N-terminal heavy chain variable region (herein "HCVR") and a heavy chain constant region. The heavy chain constant region is comprised of three domains (CH1, CH2, and CH3) for IgG, IgD, and IgA; and 4 domains (CH1, CH2, CH3, and CH4) for IgM and IgE. Each light chain is comprised of a light chain variable region (herein "LCVR") and a light chain constant region, CL. The HCVR and LCVR
regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDRs), interspersed with regions that are more conserved, termed framework regions (FR). Each HCVR and LCVR is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order:
FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. Herein the 3 CDRs of the heavy chain are referred to as "CDRH1, CDRH2, and CDRH3" and the 3 CDRs of the light chain are referred to as "CDRLl, CDRL2 and CDRL3." The CDRs contain most of the residues which form specific interactions with the antigen. Assignment of amino acids to each domain is in accordance with well-known conventions [e.g., Kabat, "Sequences of Proteins of Immunological Interest," National Institutes of Health, Bethesda, Md. (1991) or Chothia numbering scheme as described in Al-Lazikani et al., J. Mol. Biol.
.273:927-945, 1997, see also the internet site http:www.rubic.rdg.ac.uk/-andrew/bioinf:org/abs.
The functional ability of an antibody to bind a particular antigen is largely influenced by the six CDRs.

The term "antibody," in reference to an anti-myostatin monoclonal antibody of the invention (or simply, "monoclonal antibody of the invention"), as used herein, refers to a monoclonal antibody. A "monoclonal antibody" as used herein refers to a chimeric antibody, a humanized antibody or a fully human antibody, unless otherwise indicated herein. Preferably a monoclonal antibody of the invention exists in a homogeneous or substantially homogeneous population. Monoclonal antibodies of the invention can be produced using e.g., hybridoma techniques well known in the art, as well as recombinant technologies, phage display technologies, synthetic technologies or combinations of such technologies or other technologies readily known in the art. "Monoclonal antibody"
refers to an antibody that is derived from a single copy or clone, including e.g., any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced. A
"monoclonal antibody" can be an intact antibody (comprising a complete or full length Fc region), a substantially intact antibody, or a portion or fragment of an antibody comprising an antigen-binding portion, e.g., a Fab fragment, Fab' fragment or F(ab')2 fragment of a chimeric, humanized or human antibody.

The variable regions of each light/heavy chain pair form the antigen-binding sites of the antibody. Thus, an intact IgG antibody has two binding sites. Except in bifunctional or bispecific antibodies, the two binding sites are the same. As used herein, the "antigen-binding portion" or "antigen-binding region" or "antigen-binding fragment"
refers interchangeably herein to that portion of an antibody molecule, within the variable region, which contains the amino acid residues that interact with an antigen and confer on the antibody its specificity and affinity for the antigen. The antigen-binding portion of the antibody includes framework amino acid residues necessary to maintain the proper conformation of the antigen-binding residues. Preferably, the CDRs of the antigen-binding region, or the entire antigen-binding region, of the antibodies of the invention will be of murine origin or substantially of murine origin with certain amino acids residues altered to improve a particular activity (see e.g., Fig. 9). Preferably, the framework regions of antibodies of the invention are of human origin or substantially of human origin (at least 85%, 90%, 95%, 97% or 99% of human origin). In other embodiments, the antigen-binding-r-egion; or the CDRs of the antigen-binding region, can be derived from other non-human species including, but not limited to, rabbit, rat or hamster. In other embodiments, the antigen-binding region can be entirely of human origin or substantially of human origin with certain amino acids residues altered to improve a particular activity, e.g., affinity or specificity (see e.g., the amino acid positions of Fig. 9 which are in bold print and underlined).
Furthermore, a "monoclonal antibody" as used herein can be a single chain Fv fragment that may be produced by joining the DNA encoding the LCVR and HCVR
with a linker sequence. (See, Pluckthun, The Phannacology of Monoclonal Antibodies, vol.
113, Rosenburg and Moore eds., Springer-Verlag, New York, pp 269-315, 1994).
It is understood that regardless of whether fragments or portions are specified, the term "antibody" as used herein includes such fragments or portions as well as single chain forms. As long as the protein retains the ability to specifically or preferentially bind its intended target (i.e., epitope or antigen), it is included within the term "antibody."
-- -A "population-of monoclonal antibodies," refers to a homogeneous or substantially homogeneous antibody population (i.e., at least about 85%, 87%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, more preferably at least about 97% or 98% or most preferably at least 99% of the antibodies in the population would compete in an ELISA
assay for the same antigen or epitope). Antibodies may or may not be glycosylated and still fall within the bounds of the invention. Monoclonal antibodies may be homogeneous if they have identical amino acid sequence although they may differ in a post-translational modification, e.g., glycosylation pattern.
A "variant"anti-myostatin antibody, refers herein to a molecule which differs in amino acid sequence from a "parent" anti-myostatin antibody amino acid sequence by virtue of addition, deletion and/or substitution of one or more amino acid residue(s) in the parent antibody sequence. Iii the preferred embodiment, the variant comprises one or more amino acid substitution(s) in one or more CDR region(s) of the parent antibody. For example, the variant may comprise at least one (e.g., from about one to about ten, and preferably from about two to about five) substitution in one or more CDR
regions of the parent antibody. Identity or homology with respect to the variant sequence is defined herein as the percentage of amino acid residues in the variant sequence that are identical with the parent antibody residues, after aligning the sequences and introducing gaps, if necessary, to achieve-the maximum-percent sequence-identity. None of N-terminal, C-terminal, or internal extensions, deletions or insertions in the antibody sequence shall be construed as affecting sequence identity or homology. The variant retains the ability to bind myostatin and preferably has properties which are superior to those of the parent antibody. For example, the variant may have stronger binding affinity, lower IC50 in a SBE/reporter assay or enhanced ability to inhibit a myostatin bioactivity. The variant antibody of particular interest herein is one which displays at least about 2 fold, 5 fold, preferably at least about 10 fold, and more preferably at least about 20, 30, or 50 fold enhancement in a biological activity when compared to the parent antibody.
The "parent" antibody herein is one which is encoded by an amino acid sequence used for the preparation of the variant. The parent antibody may have a murine framework, but preferably has a human framework region. The parent antibody may be a murine (see e.g., Figs 10 and 11 herein), chimeric, humanized or human antibody.
The term "specifically binds" as used herein refers to the situation in which one member of a specific binding pair does not significantly bind to molecules other than its specific binding partner(s) (i.e., cross-reactivity of less than about 35%, 33%, 30%, 20%, or 10%) as measured by a technique in the art e.g., by competition ELISA or by measurement of KD with BIACORE or KINEXA assay. The term is also applicable where e.g., an antigen-binding domain of an antibody of the invention is specific for a particular epitope that is carried by a number of antigens, in which case the specific antibody carrying the antigen-binding domain will be able to specifically bind to the various antigens carrying the epitope. Accordingly a monoclonal antibody of the present invention specifically binds an epitope localized within amino acids 40-64, 43-57 and/or 45-59 of the mature form of myostatin.
The term "preferentially binds" as used herein refers to the situation in which an antibody binds a specific antigen at least about 2, 3, 5, 10, 20, 22, 24, or 25-times greater than it binds a different antigen as measured by a technique in the art e.g., by competition ELISA or by measurement of KD with BIACORE or KINEXA assay. Accordingly, a monoclonal antibody of the present inven'tion preferentially binds GDF-8 over GDF-11.
. Similarly, an antibody may preferentially bind one epitope within an antigen over a different epitope within the same antigen.
The term "epitope" refers to that portion of a molecule capable of being recognized by and bound-by an antibody at one or more of the antibody's antigen-binding regions. Epitopes often consist of a chemically active surface grouping of molecules such as amino acids or sugar side chains and have specific three-dimensional structural characteristics as well as specific charge characteristics. By "inhibiting epitope" and/or "neutralizing epitope" is intended an epitope, which when in the context of the intact molecule (in this case, myostatin) and when bound by antibody specific to the epitope, results in loss or diminution of a biological activity of the molecule or organism containing the molecule, in vivo or in vitro.
The term "epitope," as used herein, further refers to a portion of a polypeptide having antigenic and/or immunogenic activity in an animal, preferably a mammal, e.g., a mouse or a human. The term "antigenic epitope," as used herein, is defined as a portion of a polypeptide to which an antibody can specifically bind as determined by any method well known in the art, for example, by conventional immunoassays. Antigenic epitopes need not necessarily be immunogenic, but may be immunogenic. An "immunogenic epitope," as-used herein, is defined as a portion of a polypeptide that elicits an antibody response in an animal, as determined by any method known in the art. (See, e.g., Geysen et al., Proc. Natl. Acad. Sci. USA 81:3998-4002 (1983)).
The phrases "biological property" or "bioactivity," "activity" or "biological activity," in reference to an antibody of the present invention, are used interchangeably herein and include, but are not limited to, epitope/antigen affinity and specificity, ability to neutralize or antagonize an activity of myostatin in vivo or in vitro, IC50 in a myostatin/SBE reporter assay or other in vitro activity assay, the in vivo stability of the antibody and the immunogenic properties of the antibody. Other identifiable biological properties of an antibody include, for example, cross-reactivity, (i.e., with non-human homologs of the targeted peptide, or with other proteins or tissues, generally), and ability to preserve high expression levels of protein in mammalian cells. The aforementioned 5 properties or characteristics can be observed or measured or assessed using art-recognized techniques including, but not limited to, ELISA, competitive ELISA, surface plasma resonance analysis, in vitro and in vivo neutralization assays without limit, receptor binding, cytokine or growth factor production and/or secretion, Xenopus animal cap development, signal transduction and immunohistochemistry with tissue sections from 10 different sources including human, primate, or any other source as the need may be.
The term "myostatin-activity" as used herein refers to one or more of physiologically growth-regulatory or morphogenetic activities associated with active myostatin protein. For example, active myostatin is a negative regulator of skeletal muscle mass. Active myostatin can also modulate the production of muscle-specific 15 enzymes (e.g., creatine kinase), stimulate myoblast proliferation, and modulate preadipocyte differentiation to adipocytes.
The term "inhibit" or "neutralize" as used herein with respect to an activity of an antibody of the invention means the ability to substantially antagonize, prohibit, prevent, restrain, slow, disrupt, eliminate, stop, reduce or reverse e.g., progression or severity of that which is being inhibited- including; but not limited to, a biological activity or property, a disease or a condition. The inhibition or neutralization is preferably at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or higher.
The term "isolated" when used in relation to a nucleic acid or protein (e.g., an antibody) refers to a nucleic acid sequence or protein that is identified and separated from at least one contaminant with which it is ordinarily associated in its natural source.
Preferably, an "isolated antibody" is an antibody that is substantially free of other antibodies having different antigenic specificities (e.g., pharmaceutical compositions of the invention comprise an isolated antibody that specifically binds myostatin and is substantially free of antibodies that specifically bind antigens other than myostatin).
The terms "Kabat numbering" and "Kabat labeling" are used interchangeably herein. These terms, which are recognized in the art, refer to a system of numbering amino acid residues which are more variable (i.e., hypervariable) than other amino acid residues in the heavy and light chain variable regions of an antibody (Kabat, et al., Ann.
NYAcad. Sci. 190:382-93 (1971); Kabat, et al., Sequences of Proteins of Immunological Interest, Fifth Edition, U.S. Department of Health and Human Services, NIH
Publication No. 91-3242 (1991)).
A polynucleotide is 'operably linked" when it is placed into a functional relationship with another polynucleotide. For example, a promoter or enhancer is operably linked to a coding sequence if it' affects the transcription of the sequence. A
peptide is "operably linked" to another peptide when the polynucleotides encodirig them are operably linked; preferably they are in the same open reading frame.
The terms "individual," "subject," and "patient," used interchangeably herein, refer to an-animal, preferably a mammalian (including a nonprimate and a primate) or avian species, including, but not limited to, murines, simians, humans, mammalian farm animals (e.g., bovine, porcine, ovine), mammalian sport animals (e.g., equine), and mammalian pets (e.g., canine and feline); preferably the term refers to humans. The term also refers to avian species, including, but not limited to, chickens and turkeys. In a certain embodiment, the subject, preferably a mammal, preferably a human, is further characterized with a disease or disorder or condition that would benefit from a decreased level or decreased bioactivity of myostatin. In another embodiment the subject, preferably a mammal, preferably a human, is further characterized as being at risk of developing a --dis-order; disease-or condition that would benefit from a decreased level of myostatin or a decreased bioactivity of myostatin.
The term "vector" includes a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked including, but not limited to, plasmids and viral vectors. Certain vectors are capable of autonomous replication in a host cell into which they-are intr-oduced while other vectors can be integrated into the genome of a host cell upon introduction into the host cell, and thereby, are replicated along with the host genome. Moreover, certain vectors are capable of directing the expression of genes to which they are operably linked. Such vectors are referred to herein as "recombinant expression vectors" (or simply "expression vectors") and exemplary vectors are well known in the art.
As used herein, the expressions "cell," "host cell," "cell line," and "cell culture"
are used interchangeably and include an individual cell or cell culture that is a recipient of any isolated polynucleotide of the invention or any recombinant vector(s) comprisirig a sequence encoding a HCVR, LCVR or monoclonal antibody of the invention. Host cells include'progeny of a single host cell, and the progeny may not necessarily be completely identical (in morphology or in total DNA complement) to the original parent cell due to natural, accidental, or deliberate mutation and/or change. A host cell includes cells transformed, transduced or infected in vivo or in vitro with one or more a recombinant vectors or a polynucleotide expressing a monoclonal antibody of the invention or a light chain or heavy chain thereof. A host cell which comprises a recombinant vector of the invention (either stably incorporated into the host chromosome or not) may also be referred to as a"recombinant host cell". Preferred host cells for use in the invention are CHO cells (e.g., ATCC CRL-9096), NSO cells, SP2/0 cells and COS cells (ATCC
e.g., CRL-1650, CRL-1651), HeLa (ATCC CCL-2). Additional host cells for use in the invention include plant cells, yeast cells, other mammalian cells and prokaryotic cells.
Antibody Characterization The present invention relates to isolated, monoclonal antibodies that specifically bind myostatin with high affinity. The antibodies of the inventiori are preferably chimeric, humanized or human antibodies or antigen-binding portions thereof and preferably bind to myostatin within the region of the mature form of myostatin spanning -amino acids 40=64-or more preferably within the region of the mature form of myostatin spanning amino acids 43-57 and/or 45-59. Furthermore, antibodies of the invention neutralize a myostatin biological activity in vivo or in vitro. Specific binding of anti-myostatin monoclonal antibodies of the invention to myostatin allows the antibodies of the invention to be used as therapeutics or prophylactics for myostatin-associated -conditions, diseases or disorders, i.e., conditions, diseases or disorders which benefit from lowering myostatin levels or antagonizing or inhibiting a myostatin biological activity.
Further, antibodies of the invention may be used to diagnose or monitor conditions, diseases or disorders which benefit from an altered level or bioactivity of myostatin or to determine the level of myostatin in a sample.
In a preferred embodiment, the invention provides an anti-myostatin monoclonal antibody that binds myostatin or a portion thereof with a binding affinity (KD) for myostatin of less than or equal to about 3 x 10-$ M, 1 x 10-8 M or 1 x 10-9 M, preferably less than about 9 x 10"10 M or 8.7 x 10-10 M or 8 x 10-11 M. 'Preferably the antibodies of the invention are characterized by a KD for myostatin of no greater than about 3 x 10'8 M, 1 x 10"8 M, 1 x 10-9 M or 9 x 10'10 M and most preferably by a KD for myostatin of no greater than about 8.7 x 10-10M or 8 x 10"li M. Antibody affinities may be determined as described in the examples he'reinbelow or by use of any suitable method available in the art.
In another preferred embodiment,'the invention provides an anti-myostatin monoclonal antibody that has an IC50 of less than or equal to about 25 nM, 20 nM, 16 nM, 14 nM, 10 nM, 9 nM, 6 nM, 5.2 nM or less in an in vitro myostatin/SBE reporter assay as described in the examples hereinbelow. Such antibodies may be further characterized by a KD for myostatin-of no greater, than about 3 x 10"8 M, 1 x-10-8 M, 1 x 10"9 M or 9 x 10-10 M and most preferably by a KD for myostatin of no greater than about 8.7 x 10"10 M or 8 x 10"11 M. Such antibodies may be further characterized by preferentially binding myostatin when compared to their ability to bind GDF-1 1 using any art-available method, e.g., ELISA assay or competitive ELISA assay or a KD value measured e.g., with a BIACORE
or KINEXA assay.
In one embodiment, a monoclonal antibody of the invention has less than about 35, 33, 30, 28, 25, 23, or 20% cross-reactivity (more preferably, less than about 19, 18, 17, 16, 15, 14, 13, 12, 11,10, 9, 8, 7 or 6, even more preferably, less than about 5 or 4 per cent-cross-reactivity) with a non-myostatin protein (such as, e.g., GDF-1 1) or with a non-myostatin peptide consisting of at least 15, 14, 13, 12, 11, 10 or 9 contiguous amino acids, as measured by a standard technique in the art such as an ELISA assay, a competitive ELISA assay or KD values as measured, e.g., with a BIACORE or KINEXA assay.
Preferably an antibody of the invention binds myostatin at least 2, 3, 5, 10, 20, 22, 24 or ?5-fold-greater than with which it binds GDF-11, as determined e.g., by competition ELISA or BIACORE or KINEXA assay. Most preferably, the antibodies of the invention do not bind GDF-11 at levels greater than background levels when using any binding assay available to one or ordinary skill in the art. A monoclonal antibody of the invention binds a monomeric or dimeric form of myostatin or a portion thereof comprising amino acids spanning residues 40-64, 43-57 and/or 45-59 or mature myostatin.
Preferably the myostatin and GDF- 11 polypeptides tested for preferential binding of an antibody of the invention are both homodimeric forms of the mature protein, preferably of mammalian or avian origin, even more preferably of human origin.
However, the myostatin and GDF-11 polypeptides tested for preferential binding of an antibody of the invention may be the monomeric form of the mature protein or proprotein form or a polypeptide comprising a portion of the mature protein spanning amino acids 40-64, 43-57 and/or 45-59 of the mature form of the protein.
The anti-myostatin monoclonal antibodies of the invention bind an antigenic epitope discovered to be localized within amino acids 40-64 (SEQ ID NO: 53 for human) of mature myostatin preferably within amino acids 43-57 and/or 45-59 of mature myostatin. Furthermore, a myostatin immunogenic epitope of the invention is localized within amino acids 40-64 of mature myostatin (SEQ ID NO: 53 for human), preferably within amino acids 43-57 -and/or 45-59 of mature myostatin of any mammalian or avian species. An immunogenic epitope of the invention is also contemplated to be an antigenic epitope. The antigenic epitope may possess additional myostatin residues outside of amino acids 40-64 of mature myostatin, but the monoclonal antibodies of the invention do _not-require these additional residues to specifically bind myostatin.
Additionally, residues of myostatin outside of the amino acids 40-64 may affect the conformational structure of the antigenic domain and thereby alter binding of an antibody of the invention to the antigenic epitope. Preferably the monoclonal antibodies of the invention do not significantly react (i.e., bind) with GDF-11 compared to the level with which they bind --GDF-S:- More-preferably; the monoclonal antibodies of the invention bind myostatin at least 2, 3, 5, 10, 20, 22, 24 or 25-fold greater (e.g., greater affinity or greater specificity) than with which it binds GDF-11 as determined e.g., by ELISA assay, competition ELISA
assay or KD values in a BIACORE or KINEXA assay.
A peptide consisting of amino acids 40-64 (inclusive), 43-57 or 45-59 of mature myostatin or any peptide consisting of an immunogenic epitope as described herein may be used as an immunogenic peptide, preferably conjugated to a carrier protein e.g., KLH, to generate monoclonal antibodies of the invention using methods available to one of ordinary skill in the art. It is contemplated that cysteine residues within these peptides may be changed to serine residues and still function as an immunogenic epitope. The immunogenic peptide may be used to immunize a rion-human animal, preferably a mammal, more preferably a mouse. For a fully human antibody, the immunogenic peptide may be used to immunize a transgenic strain of mouse in which mouse antibody gene expression is suppressed and effectively replaced with human antibody gene expression. Then anti-myostatin antibodies are isolated from the immunized animal and screened by methods well known in the art to isolate those antibodies that specifically bind within the domain spanning amino acids 40-64 of mature myostatin, preferably 5 within the domain spanning'amino acids 43-57 and/or 45-59 preferably screened for a binding affinity less than about 3 x 10"8 M, 1 x 10-8 M or 1 x 10"9 M, preferably less than about 9 x 10-10M, 8.7 x 10"10M or 8 x 10'11 M.and/or screened for an IC50 of less than or .. equal to about 25 nM, 20 nM, 16 nM, 14 nM; 10 nM, 9 nM, 6 nM, 5.2 nM or less in an in vitro myostatin/SBE assay (see the assay described in Example 6 herein).
10 Monoclonal antibodies may be made using the hybridoma method widely known in the art (see e.g., Kohler et al., Nature, 256:495, 1975) or may be made by recombinant DNA methods (e.g., as in U.S. Patent No. 4,816,567) or other methods available in the art. Generally, a hybridoma can be produced by fusing a suitable immortal cell line (e.g., a myeloma cell line such as SP2/0) with antibody producing cells of the immunized 15 animal The antibody producing cell, preferably those of the spleen or lymph nodes, are obtained from animals immunized with the antigen of interest. The fused cells (hybridomas) can be isolated using selective culture conditions, and cloned by limiting dilution. Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen. Preferably, the binding specificity 20 of mabs produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or ELISA. Cells which produce antibodies with the desired binding properties can be selected by a suitable screening assay. Methods for such isolation and screening are well known in the art.
Other suitable methods of producing or isolating antibodies which bind within the domain spanning-amino acids 40-64, 43-57 and/or 45-59 of mature myostatin, including human or artificial antibodies, can be used, including, for example, methods which select a recombinant antibody (e.g., single chain Fv or Fab) from a library, or which rely upon immunization of transgenic animals (e.g., mice) capable of producing a repertoire of human antibodies (see e.g., Jakobovits et al., Proc. Natl. Acad. Sci. USA, 90:2551-2555, 1993; Jakobovits et al., Nature, 362:255-258, 1993; Lonberg et al., U.S.
Patent Number 5,545,806; Surani et al., U.S. Patent Number 5,545,807).
Single chain antibodies, and chimeric, humanized or primatized (CDR-grafted) antibodies, as well as chimeric or CDR-grafted single chain antibodies, and the like, comprising portions derived from different species, are also encompassed by the present invention and the term "antibody". The various portions of these antibodies can be joined together chemically by conventional techniques, syntlietically, or can be prepared as a contiguous protein using genetic engineering techniques. For example, nucleic acids encoding a chimeric or humanized chain can be expressed to produce a contiguous protein. See e.g., U.S. Patent No. 4,816,567; European Patent No. 0,125,023 Bl; U.S.
Patent No. 4,816,397; European Patent No. 0,120,694 B1; WO 86/01533; European Patent No. 0,194,276 B 1; U.S. Patent No. 5,225, 539; European Patent No.
0,239,400 B 1 - and U.S. Patent Nos. 5,585,089 and 5,698,762. See also, Newman; R. et al.
BioTechnology, 10:1455-1460, 1993, regarding primatized antibody, and Ladner et al., U.S. Patent No. 4,946,778 and Bird, R.E. et al., Science, 242:423-426, 1988, regarding single chain antibodies.
In addition, functional portions of antibodies, including antigen-binding portions of chimeric, humanized, human or single chain antibodies, can also be produced.
Functional portions of the foregoing antibodies retain at least one antigen-binding function and/or biological function or bioactivity of the full-length antibody from which they are derived. Preferred functional portions retain an antigen-binding function of a corresponding full-length antibody (e.g., the ability to bind a mammalian mature form of myostatin). Particularly preferred functional portions or fragments retain the ability to inhibit one or more functions or bioactivities characteristic of a mammalian mature myostatin, such as a binding activity, a signaling activity, and/or stimulation of a cellular response. For example, in one embodiment, a functional portion or fragment can inhibit the interaction of mature myostatin with one or more of its-ligands-and/or can inhibit one or more receptor-mediated functions.
Antibody portions or fragments capable of binding to mature myostatin or a portion thereof (preferably within amino acids 40-64, 43-57 and/or 45-59 of mature myostatin), include, but are not limited to, Fv, Fab, Fab' and F(ab')2 fragments and are encompassed by the invention. Such fragments can be produced by enzymatic cleavage or by recombinant techniques. For instance, papain or pepsin cleavage can generate Fab or F(ab')2 fragments, respectively. The smallest antigen-binding fragment is the Fv, which consist of the HCVR and the LCVR domains. The Fab fragment consists of the HCVR-CHl and LCVR-CL domains covalently linked by a disulfide bond between the constant regions. To overcome the tendency of non-covalently linked HCVR and LCVR
domains in the Fv to dissociate when co-expressed in a host cell, a so-called single chain (sc) Fv fragment (scFv) can'be constructed, in which a flexible and adequately long polypeptide links either the C-terminus of the HCVR to the N-terminus of the LCVR or the C-terminus of the LCVR to the N-terminus of the HCVR. The most commonly used linker has been a 15-residue (Gly4Ser)3 peptide, but other linkers are also known in the art.
Antibodies can also be produced in a variety of truncated forms using antibody genes in ' which one or more stop codons has been introduced upstream of the natural stop site. For example, a chimeric gene encoding a F(ab')2 heavy chain portion can be designed to include DNA sequences encoding the CHl domain and hinge region of the heavy chain.
Selection of antibody fragments from libraries using enrichment technologies such as phage-display (Matthews DJ and Wells JA. Science. 260:1113-7, 1993), ribosome display (Hanes, et al., Proc. Natl. Acad. Sci. (USA) 95:14130-5, 1998), bacterial display (Samuelson P., et al., Journal of Biotechnology. 96:129-54, 2002) or yeast display (Kieke MC, et al., Protein Engineerirag, 10:1303-10, 1997) has proven to be successful alternatives to classical hybridoma technology (recent reviews: Little M. et al., Iinniunology Today, 21:364-70, 2000;).
Variant Antibodies A murine monoclonal antibody or a human antibody (produced e.g., in a transgenic mouse) raised against an immunogenic epitope (amino acids 40-64, 43-57 or 45-59 of mature myostatin) of the invention or against a protein comprising an - immunogenic epitope of the invention is a parent antibody. A murine parent antibody may be further altered to create a chimeric or humanized form of the antibody using methods well known in the art. Such chimeric or humanized antibodies, may serve as parent antibodies for further variation or mutagenesis. Parent antibodies of the invention may be further mutagenized e.g., within the CDR domain(s) (see, e.g., Fig. 9) to create a variant antibody with an optimized property of interest, e.g., binding affinity, IC50, specificity, etc. An amino acid substitution variant antibody is preferred and has at least one amino acid residue of the parent antibody molecule removed and a different residue inserted in its place. The sites of greatest interest for substitutional mutagenesis include the CDR regions, but FR alterations are also contemplated. Conservative amino acid substitutions are preferred. If such substitutions result in a change in a biological activity of the antibody; then more substantial changes, i.e., non-conservative amino acid changes, may be introduced and the products screened.
A convenient way for generating substitutional variants is affinity maturation using phage display. Briefly, several CDR region sites are mutated to generate all possible amino acid substitutions at each site. The antibody variants thus generated are displayed in a monovalent fashion from filamentous phage particles as fusions to the gene III product of M 13 packaged within each particle. The phage-displayed variants are then screened for their biological activity-(e.g., binding affinity, specificity, IC$o) as herein disclosed. In order to identify candidate CDR region sites for modification, alanine scanning mutagens can be performed to identify CDR region residues contributing significantly to antigen binding. Alternatively, or in addition, it may be beneficial to analyze a crystal structure of the antigen-antibody coinplex to identify contact points between the antibody and myostatin. Such contact residues and neighboring residues are candidates for substitution according to the techniques elaborated herein or known in the art. Alternatively, or in addition, random mutagenesis may be performed on one or more - CDR sequences at one or more residue positions, either while the CDR is operably linked to the variable region or while the CDR is independent of other variable region sequence and then the altered CDR returned to a variable region using recombinant DNA
technology. Once such variant antibodies are generated and expressed, the panel of variants is subjected to screening as described herein and antibodies with superior properties in one or more relevant assays may be selected for further development.
- Any cysteine residue not involved in maintaining the proper conformation of an anti-myostatin antibody of the invention may be substituted, generally with serine, to improve the oxidative stability of the molecule and prevent aberrant crosslinking.
Conversely, cysteine bond(s) may be added to the antibody to improve its stability (particularly where the antibody is an antibody fragment such as an Fv fragment).
Another type of amino acid variant of the antibody alters the original glycosylation pattern of the antibody. By altering is meant deleting one or more carbohydrate moieties found in the antibody, and/or adding one or more glycosylation sites that are not present in the parent antibody.
Glycosylation of antibodies is typically either N-linked or 0-linked. N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagines residue. The tripeptide sequence asparagines-X-serine and asparagines-X-threonine, where X is any amino acid except praline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to 'the asparagines side chain. Thus the presence of either of these tripeptide sequences in a polypeptide creates a potential glycosylation site.
0-linked glycosylation refers to the attachment of one of the sugars N-aceylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, -although 5-hydroxyproline or 5-hydroxylysine may also be used.
Addition of glycosylation sites to the antibody is conveniently accomplished by altering the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites). The alteration may also be made by the addition of, or substitution by, one or more serine or threonine residues to the sequence of the original antibody (for 0-linked glycosylation sites).

Sequence A preferred monoclonal antibody of the invention has a LCVR comprising a peptide with a sequence selected from the group consisting of SEQ ID NOs: 5-12 and/or a HCVR comprising a peptide with a sequence selected from the group consisting of SEQ
ID NOs: 13-26, 55 and 56 (See Figs. 5-9 herein). Furthermore, a monoclonal antibody of the invention is one that is competitively inhibited from binding mature human myostatin (or a portion thereof) by a monoclonal antibody comprising two polypeptides with the sequences shown-in the group consisting of (i) SEQ ID NOs: 5 and 15; (ii) SEQ
ID NOs:
5 and 16; and (iii) SEQ ID NOs: 10 and 26. Such competitive inhibition between antibodies may be measured by assays readily known to one of skill in the art, e.g., a competition ELISA assay.
In another embodiment, an anti-myostatin monoclonal antibody of the invention comprises (i) a LCVR polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NOs: 5-12 and (ii) a HCVR polypeptide with an amino acid sequence selected from the group consisting of SEQ IID NOs: 13-26, 55 and 56.

Preferably, an antibody comprising an LCVR polypeptide with an amino acid sequence of SEQ ID NO: 5 further comprises a HCVR polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NOs: 13-20. Preferably, an antibody comprising an LCVR polypeptide with an amino acid sequence of SEQ ID NO: 6 further 5 comprises a HCVR polypeptide with an amino acid sequence of SEQ ID NO: 14.
Preferably, an antibody comprising an LCVR polypeptide with an amino acid sequence of SEQ ID NO: 7 further comprises a HCVR polypeptide with an amino acid sequence selected from the group consisting of SEQ ID'NOs: 21-25 and 56. Preferably, an antibody comprising an LCVR polypeptide with an an-uno acid sequence of SEQ ID
NO:
10 10 further comprises a HCVR polypeptide with an amino acid sequence of SEQ
ID NO:
26 or 55. Preferably, an antibody comprising an HCVR polypeptide with an amino acid sequence of SEQ ID NO: 55 further comprises a LCVR polypeptide with an amino acid sequence selected from the group consisting of SEQ ID NOs: 8-12.
The skilled artisan will appreciate that the antibodies of the invention are not 15 limited to the specific sequences of HCVR and LCVR as stated in Figs 5-9 herein, but also include variants of these sequences that retain antigen binding ability.
Such variants may be derived from the provided sequences using techniques known in the art as described above.
In another embodiment, an anti-myostatin antibody of the invention has a heavy 20 and a light chain variable-region, wherein the heavy chain variable region comprises CDR
regions with the following amino acid sequences: CDRH1 (SEQ ID NO: 59), CDRH2 (SEQ ID NO: 60) and CDRH3 (SEQ ID NO: 61); and/or wherein the light chain variable region comprises CDR regions with the following amino acid sequences: CDRLl (SEQ
ID NO: 57), CDRL2 (SEQ ID NO: 30 or 52) and CDRL3 (SEQ ID NO: 58). Preferably, 25 --the heavy chain CDRs of an antibody of the invention are as shown in Figs. 5, 6 or 9 and the light chain CDRs of an antibody of the invention are as shown in Figs. 7, 8 or 9 and the CDRs exist in the antibody at the CDR location defined in the Figures. For example, one antibody of the invention has CDRL1 with SEQ ID NO: 27, CDRL2 with SEQ ID
NO: 30, CDRL3 with SEQ ID NO: 31, CDRH1 with SEQ ID NO: 36, CDRH2 with SEQ
ID NO: 43 and CDRH3 with SEQ ID NO: 47 (as does antibody IC7.1 in Figure 9).
It is further contemplated that an anti-myostatin antibody of the invention comprises a heavy chain variable region comprising a CDRHl region with a sequence selected from the group consisting of that as shown in SEQ ID NOs: 36-42 and/or a CDRH2 region with a sequence with a sequence selected from the group consisting of that as shown in SEQ ID NOs: 43-46 and/or a CDRH3 region with a sequence selected from the group consisting of that as shown in SEQ ID NOs: 47-51. In another embodiment, an anti-myostatin antibody of the invention comprises a light chain variable region comprising a CDRL1 region with a sequence selected from the group consisting of that as shown in SEQ ID NOs: 27-29 and/or a CDRL2 region with a sequence selected from the group consisting of that as shown in SEQ ID NOs 30 or 52 and/or a region with a sequence selected from the group consisting of that as shown in SEQ ID
NOs: 31-35. In a preferred embodiment, an anti-myostatin antibody of the invention comprises a heavy chain variable region comprising a CDRH1 region with a sequence selected from the group consisting of that as shown in SEQ ID NOs: 36-42 and/or a CDRH2 region with a sequence selected from the group consisting of that as shown in SEQ ID NOs: 43-46 and/or a CDRH3 region with a sequence selected from the group consisting of that as shown in SEQ ID NOs: 47-51 and further comprises a light chain variable region comprising a CDRLl region with a sequence selected from the group consisting of that as shown in SEQ ID NOs: 27-29 and/or a CDRL2 region with a sequence selected from the group consisting of that as shown in SEQ ID NOs 30 or 52 and/or a CDRL3 region with a sequence selected from the group consisting of that as shown in SEQ ID NOs: 31-35.
The structure for carrying a CDR of the invention will generally be an antibody heavy or light chain sequence or a substantial portion thereof, in which the CDR is located at a location corresponding to the CDR of naturally occurring HCVR and LCVR
(Kabat et al, Sequences of Proteins of Immunological Interest, US Dept of HHS, 1991).
Preferably, the three hypervariable regions (CDRs) for each chain, light and heavy, are provided in a human framework region, e.g., as a contiguous sequence represented by the following formula: FRl-CDRl-FR2-CDR2-FR3-CDR3-FR4. The heavy chain or light chain FR1, FR2, FR3 and FR4 combine to form the complete framework when arranged as a contiguous sequence with the CDRs in the order stated.
In addition to CDR sequences, the LCVR and HCVR further comprise framework sequence. In a humanized antibody for therapeutic use in humans, the framework sequence is preferably entirely or substantially of human origin (i.e., at least 85 Io, 87%, 90%, 92%, 95%, 96%, 97%, 98% or 99% of human origin). Preferably the light chain framework region of a humanized, human or chimeric antibody of the invention is 02 as shown in Fig. 7, comprised of FR1 with SEQ ID NO: 65, FR2 with SEQ ID NO: 66, with SEQ ID NO: 67 and FR4 with SEQ ID NO: 68. Preferably the heavy chain framework region of a humainized, human or chimeric antibody of the invention is VH2-70 or VH4-39 as shown in Figs. 5 and 6 respectively. VH2-70 framework is comprised of FR1 with SEQ ID NO: 69, FR2 with SEQ ID NO: 70, FR3 with SEQ ID NO: 71, and . with SEQ ID NO: 72. VH4-39 is comprised of FR1 with SEQ ID NO: 73, FR2 with SEQ
ID NO: 74, FR3 with SEQ ID NO: 75 and FR4 with SEQ ID NO: 76. In an antibody for use in a non-human animal, the framework region sequence may substantially originate from the human genome (preferably-used in a non-human animal when it is an embryo or newly born) or from the genome of the animal in which it is to be used therapeutically.
In one embodiment, an anti-myostatin antibody of the invention wherein all or a portion of the variable region is limited by a particular sequence as shown by a SEQ ID
NO herein is further characterized by being a chimeric, humanized, or fully human antibody or antigen-binding portion thereof that antagonizes or neutralizes at least one myostatin activity in vivo or in vitro. An anti-myostatin antibody of the invention wherein all or a portion of the variable region is limited by a particular sequence as shown by a SEQ ID NO herein is preferably further characterized by preferentially binding GDF-8- over GDF-11.- More preferably such antibodies bind myostatin at least about 2, 3, 5, 10, 20, 22, 24, or 25-times greater than it binds GDF-11. Most preferably such antibodies, when expressed as Fabs, do not bind GDF-11 above background levels.
An anti-myostatin- antibody of the invention wherein all or a portion of the variable region is limited by a particular sequence as shown by a SEQ ID NO
herein are preferably further characterized by (i) binding a peptide consisting of an amino acid sequence as shown in SEQ ID NO: 53, preferably a peptide consisting of an amino acid sequence as shown in SEQ ID NO: 62; and/or (ii) having an IC50 less than or equal to about 25 nM, 20 nM, 16 nM, 10 nM, 9 nM, 6 nM, 5.7 nM in an in vitro myostatin/SBE
reporter assay and/or (iii) having a binding affinity (KD) for myostatin less than about 3 x 10"8 M, 1 x 10"8 M or 1 x 10-9 M, preferably less than about 9 x 10-10 M or 8.7 x 10-10 M or 8 x 10-11 M; alternatively characterized as a KD for myostatin of no greater than about 3 x 10-8 M, 1 x 10-8 M, 1 x 10"9 M or 9 x 10-10 M; more preferably a KD no greater than about 8.7 x 10"10M and most preferably a KD no greater than about 8 x 10-11 M.

Antibody Expression The present invention is also directed to cell lines that express an anti-myostatin monoclonal antibody of the invention or portion thereof. Creation and isolation of cell lines producing a monoclonal antibody of the invention can be accomplished using standard techniques known in the art. Preferred cell lines include COS, CHO, SP2/0, NSO and yeast (available from public repositories such as ATCC, American Type Culture Collection, Manassas, VA).
--A wide variety of host expression systems can be used to express an antibody of the present invention including prokaryotic (bacterial) and eukaryotic expression systems (such as yeast, baculovirus, plant, mammalian and other animal cells, transgenic animals, and hybridoma cells), as well as phage display expression systems. An example of a suitable bacterial expression vector is pUC119 and a suitable eukaryotic expression vector is a modified pcDNA3.1 vector with a weakened DHFR selection system. Other antibody expression systems are also known in the art and are contemplated herein.
An antibody of the invention can be prepared by recombinant expression of immunoglobulin light and heavy chain genes in a host cell. To express an antibody recombinantly, a host cell is transformed, transduced, infected or the like with one or more recombinant expression vectors carrying DNA fragments encoding the immunoglobulin light and/or heavy chains of the antibody such that the light and/or heavy chains are expressed in the host cell. The heavy chain and the light chain may be expressed independently from different promoters to which they are operably linked in one vector or, alternatively, the heavy chain and the light chain may be expressed independently from different promoters to which they are operably linked in two vectors -one expressing the heavy chain and one expressing the light chain. Optionally the heavy chain and light chain may be expressed in different host cells. Preferably, the recombinant antibodies are secreted into the medium in which the host cells are cultured, from which the antibodies can be recovered or purified. Standard recombinant DNA
methodologies are used to obtain antibody heavy and light chain genes, incorporate these genes into recombinant expression vectors, and introduce the vectors into host cells. Such standard recombinant DNA technologies are described, for example, in Sambrook, Fritsch; and Maniatis (Eds.), Molecular Cloning;, A Labor-atory Manual, Second Edition, Cold Spring Harbor, N.Y., 1989; Ausubel, et al (Eds.) Current Protocols in Molecular Biology, Greene Publishing Associates, 1989.
An isolated DNA encoding a HCVR region can be converted to a full-length heavy chain gene by operably linking the HCVR-encoding DNA to another DNA
molecule encoding heavy chain constant iegions (CH1, CH2,' and CH3). The sequences of human heavy chain constant region genes are known in the art. See, e.g., Kabat, et al., Sequences of Proteins of.Inimunological Interest, Fifth Edition, U.S.
Department of Health and Human Services, NIH Publication No. 91-3242 (1991). DNA fragments encompassing these regions can be obtained e.g., by- standard PCR
amplification. The heavy chain constant region can be of any type, (e.g., IgG, IgA, IgE, IgM or IgD), class (e.g., IgGI, IgG2, IgG3 and IgG4) or subclass constant region and any allotypic variant thereof as described in Kabat (supra). Alternatively, the antigen binding portion can be a Fab fragment, Fab' fragment, F(ab')2 fragment, Fd, or a single chain Fv fragment (scFv).
For a Fab fragment heavy chain gene, the HCVR-encoding DNA may be operably linked to another DNA molecule encoding only a heavy chain CH1 constant region.
An isolated DNA encoding a LCVR region may be converted to a full-length light chain gene (as well as to a Fab light chain gene) by operably linking the LCVR-encoding DNA to another DNA molecule encoding a light chain constant region, CL. The sequences of human light chain constant region genes are known in the art.
See, e.g., Kabat, supra. DNA fragments encompassing these regions can be obtained by standard PCR amplification. The light chain constant region can be a kappa or lambda constant region.
To create an scFv gene, the HCVR- and LCVR-encoding DNA fragments are operably linked to another fragment encoding a flexible linker, e.g., encoding the amino acid sequence (Gly4-Ser)3, such that the HCVR and LCVR sequences can be expressed as a contiguous single-chain protein, with the LCVR and HCVR regions joined by the flexible linker. See, e.g., Bird, et al.., Science 242:423-6, 1988; Huston, et al., Proc.lVatl.
Acad. Sci. USA 85:5879-83, 1988; McCafferty, et al., Nature 348:552-4, 1990.
To express an antibody of the invention, a DNA encoding a partial or full-length light and/or heavy chain, obtained as described above, are inserted into an expression vector such that the gene is operably linlced to transcriptional and translational control sequences. The expression vector and expression control sequences are chosen to be compatible with the expression host cell used. The antibody light chain gene and, the antibody heavy chain gene can be inserted into separate vectors or, more typically, both 5 genes are inserted into the same expression vector. The antibody genes are inserted into the expression vector by standard methods. Additionally, the recombinant expression vector can encode a signal peptide that facilitates secretion of the anti-myostatin monoclonal antibody light and/or heavy chain from a host cell. The anti-myostatin monoclonal antibody light and/or heavy chain gene can be cloned into the, vector such that 10 the signal peptide is operably linked in-frame to the amino terminus of the antibody chain gene. T-he signal peptide can be an immunoglobulin signal peptide -or a heterologous signal peptide.
In addition to the antibody heavy and/or light chain gene(s), a recombinant expression vector of the invention carries regulatory sequences that control the expression 15 of the antibody chain gene(s) in a host cell. The term "regulatory sequence" is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals), as needed, that control the transcription or translation of the antibody chain gene(s). The design of the expression vector, including the selection of regulatory sequences may depend on such factors as the choice of the host cell to be transformed, the 20 -le,vel of expression of protein desired. Preferred regulatory sequences for mammalian host cell expression include viral elements that direct high levels of protein expression in mammalian cells, such as promoters and/or enhancers derived from cytomegalovirus (CMV), Simian Virus 40 (SV40), adenovirus, (e.g., the adenovirus major late promoter (AdMLP)) and polyoma virus.
25 In addition to the antibody heavy and/or light chain genes and regulatory sequences, the recombinant expression vectors of the invention may carry additional sequences, such as sequences that regulate replication of the vector in host cells (e.g., origins of replication) and one or more selectable marker genes. The selectable marker gene facilitates selection of host cells into which the vector has been introduced. For 30 example, typically the selectable marker gene confers resistance to drugs, such as G418, hygromycin, or methotrexate, on a host cell into which the vector has been introduced.
Preferred selectable marker genes include the dihydrofolate reductase (DHFR) gene (for use in DHFR-minus host cells with methotrexate selection%amplification), the neo gene (for G418 selection), and glutamine synthetase (GS) in a GS-negative cell line (such as NSO) for selection/amplification.
For expression of the light and/or heavy chains, the expression vector(s) encoding the heavy and/or light chains is introduced into a host cell by standard techniques e.g., electroporation, calcium phosphate precipitation, DEAE-dextran transfection, transduction, infection and the like. Although it is theoretically possible to express the antibodies of the invention in either prokaryotic or eukaryotic host cells, eukaryotic cells are preferred, and most preferably mammalian host cells, because such cells, are more likely to assemble and secrete a properly folded'and immunologically active antibody.
Preferred mammalian host cells for expressing the recombinant antibodies of the invention include Chinese Hamster Ovary (CHO cells) (including DHFR-CHO cells, described in Urlaub and Chasin, Proc. Natl. Acad. Sci. USA 77:4216-20, 1980, used with a DHFR selectable marker, e.g., as described in Kaufman and Sharp, J. Mol.
Biol.
159:601-21, 1982, NSO myeloma cells, COS cells, and SP2/0 cells. When recombinant expression vectors encoding antibody genes are introduced into mammalian host cells, the antibodies are produced by culturing the host cells for a period of time sufficient to allow for expression of the antibody in the host cells or, more preferably, secretion of the -antibody into the culture medium in which the host cells are grown. Antibodies can be recovered from the host cell and/or the culture medium using standard purification methods.
Host cells can also be used to produce portions, or fragments, of intact antibodies, e.g., Fab fragments or scFv molecules by techniques that are conventional. It will be understood by a skilled artisan that variations on the above procedure are within the scope of the present -invention. For example, it may be desirable to transfect a host cell with DNA encoding either the light chain or the heavy chain of an antibody of this invention.
Recombinant DNA technology may also be used to remove some or all the DNA
encoding either or both of the light and heavy chains that is not necessary for binding to myostatin. The molecules expressed from such truncated DNA molecules are also encompassed by the antibodies of the invention.
In a preferred system for recombinant expression of an antibody of the invention, a recombinant expression vector encoding both the antibody heavy chain and the antibody light chain is introduced into DHFR-CHO cells by e.g., calcium phosphate-mediated transfection. Within the recombinant expression vector, the antibody heavy and light chain genes are each operably linked to enhancer/promoter regulatory elements (e.g., derived from SV40, CMV, adenovirus and the like, such as a CMV enhancer/AdMLP
promoter regulatory element or an SV40 enhancer/AdMLP promoter regulatory element) to drive high levels of transcription of the genes. The recombinant expression vector also carries a DHFR gene, which allows for selection of CHO cells that have been transfected with the vector using methotrexate selection/amplification. The selected transformant host cells are cultured to allow for expression of the antibody heavy and light chains and intact antibody is recovered from the culture medium. Standard molecular biology -techniques are-used -to-prepare the recombinant expression vector, transfect the host cells, select for transformants, culture the host cells and recover the antibody from the culture medium. Antibodies, or antigen-binding portions thereof, of the invention can be expressed in an animal (e.g., a mouse) that is transgenic for human immunoglobulin genes (see, e.g., Taylor, et al., Nucleic Acids Res. 20:6287-95, 1992).
Once expressed, the intact antibodies, their dimers, individual light and heavy chains, or other immunoglobulin forms of the present invention can be purified according to standard procedures of the art, including ammonium sulfate precipitation, ion -exchange, affinity, reverse phase, hydrophobic interaction column chromatography, gel electrophoresis and the like. Substantially pure immunoglobulins of at least about 90%, 92%, 94% or 96% homogeneity are preferred, and 98 to 99% or more homogeneity most preferred, for pharmaceutical uses. Once purified, partially or to homogeneity as desired, the peptides may then be used therapeutically or prophylactically, as directed herein.

Chimeric antibody -As used herein, the term "chimeric antibody" includes monovalent, divalent or polyvalent immunoglobulins. A monovalent chimeric antibody is a dimer formed by a chimeric heavy chain associated through disulfide bridges with a chimeric light chain. A
divalent chimeric antibody is a tetramer formed by two heavy chain-light chain dimers associated through at least one disulfide bridge.
A chimeric heavy chain of an antibody comprises an antigen-binding region derived from the heavy chain of a non-human antibody specific for myostatin, which is operably linked to at least a portion of a human or substantially human (or species different from that from which the antigen-binding region was derived) heavy chain constant region. A chimeric light chain of an antibody comprises an antigen binding region derived entirely or substantially from the light chain of a non-human antibody operably linked to at least a'portion of a human or substantially human (or species different from that from which the antigen-binding region was derived) light chain constant region (CL). Antibodies, fragments or derivatives having chimeric heavy chains and light chains of the same or different variable region binding specificity, can also be prepared by appropriate association of the individual polypeptide chains, according to known method steps.
With- thi-s-approach, hosts expressing chimeric heavy chains are separately cultured from hosts expressing chimeric light chains, and the immunoglobulin chains are separately recovered and then associated. Alternatively, the hosts can be co-cultured and the chains allowed to associate spontaneously in the culture medium, followed by recovery of the assembled immunoglobulin or fragment. Methods for producing chimeric antibodies are known in the art (see, e.g., U.S. Patent Nos.: 6,284,471;
5,807,715;
4,816,567; and 4,816,397).

Humanized antibodies Preferably an- antibody of the invention to be used for therapeutic purposes, would have the sequence of the framework and constant region (to the extent it exists in the antibody) derived from the mammal in which it would be used as a therapeutic so as to decrease the possibility that the mammal would illicit an immune response against the therapeutic antibody. Humanized antibodies are of particular interest since they are considered-to be valuable for therapeutic application and avoid the human anti-mouse antibody response frequently observed with rodent antibodies. Additionally, in humanized antibodies the effector portion is human so it may interact better with the other parts of the human immune system (e.g., destroy the target cells more efficiently by complement-dependent cytotoxicity or antibody-dependent cellular cytotoxicity). Also, injected humanized antibodies may have a half-life more like that of naturally occurring human antibodies than do e.g., murine antibodies, thereby allowing smaller and less frequent doses to be given. The term "humanized antibody" as used herein refers to an antibody comprising portions of antibodies of different origin, wherein at least one portion is of human origin. For example, the humanized antibody can comprise portions derived from an antibody of nonhuman origin with the requisite specificity, such as a mouse, and from an antibody of human origin, joined together chemically by conventional techniques (e.g., synthetic) 6'r prepared as a contiguous polypeptide using genetic engineering techniques.
Preferably, a "humanized antibody" has CDRs that originate from a non-human antibody (preferably a mouse monoclonal antibody) while framework and constant region, to the. extent it is present, (or a significant or substantial portion thereof, i.e., at least aboiut 85%, 87%, 90%, 92%, 94%, 95%, 96%, 97%, 98% or 99%) are encoded by nucleic acid sequence information-that occurs in the human germline immunoglobulin region (see, e.g., the International ImMunoGeneTics Database) or in recombined or mutated forms thereof whether or not said antibodies are produced in a human cell. The CDRs of a humanized antibody may be optimized from the CDRs of non-human parent antibody from which they originated to generate desired properties, e.g., specificity, affinity and capacity. Optimized CDRs may have amino acid substitutions, additions and/or deletions when compared to the parent CDRs. For example, the amino acid positions of SEQ
ID
NOs: 57, 30, 58, 59, 60, and 61 that are underlined and in bold print in Fig.
9 are positions which have been optimized from the parent CDRs as shown in Figs 10 and 11.
However, it is contemplated that any non-human anti=myostatin antibody(i) raised against a peptide consisting of a sequence shown in SEQ ID NO: 53 or 62 or (ii) raised against a peptide comprising the sequence shown in SEQ ID NO: 53 or 62 and reactive to a peptide consisting of the sequence shown in SEQ ID NO: 53 or 62, may serve as a parent antibody which may be converted to a humanized or chimeric antibody using standard methods available in the art. - -Humanized forms of non-human (e.g., murine) antibodies include an intact antibody, a substantially intact antibody, a portion of an antibody comprising an antigen-binding site, or a portion of an antibody comprising a Fab fragment, Fab' fragment, F(ab')2, or a single chain Fv fragment. Humanized antibodies preferably contain minimal sequence derived from non-human immunoglobulin. Humanized antibodies may also comprise residues which are found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the amino acids in the CDR regions correspond to those of a non-human immunoglobulin and all or substantially all of the aniino acids in the FR regions are those of a human immunoglobulin consensus sequence. The humanized antibody 5 optimally also comprises at least a portion of an immunoglobulin constant region (Fc), typically that of a human immunoglobulin. [Jones et al., Nature, 321:522-525 (1986);
Riechmann et al., Nature, 332:323-329 (1988); and Presta, Curr. Op. Struct.
Biol., 2:593-596 (1992).]
Humanized antibodies may be subjected to in vitro mutagenesis using methods of 10 routine use in the art (or, when an animal transgenic for human Ig sequences is used, in vivo somatic mutagenesis) and, thus, the framework region amino acid sequences of the HCVR and LCVR regions of the humanized recombinant antibodies are sequences that, while derived from those related to human germline HCVR and LCVR sequences, may not naturally exist within the human antibody germline repertoire in vivo. It is 15 contemplated that such amino acid sequences of the HCVR and LCVR framework regions of the humanized recombinant antibodies are at least 85%, 87%, 90%, 92%, 94%, 95%, 96%, 98% or most preferably at least 99% identical to a human germline sequence.
Preferably, those framework residues of the parent antibody (e.g., murine antibody or generally the antibody from which the humanized antibody is derived) which maintain or 20 affect combining-site structures will be retained. These residues may be identified e.g., by X-ray crystallography of the parent antibody or Fab fragment, thereby identifying the three-dimensional structure of the antigen-binding site.
The humanized antibody of the present invention may comprise or be derived from a human germline light chain framework. In particular embodiments, the light chain 25 germline sequence is selected from human VK sequences including, but not limited to, Al, A10, All, A14, A17, A18, A19, A2, A20, A23, A26, A27, A3, A30, A5, A7, B2, B3, L1, L10, L11, L12, L14, L15, L16, L18, L19, L2, L20, L22, L23, L24, L25, L4/18a, L5, L6, L8, L9, 01, 011, 012, 014, 018, 02, 04, and 08. In certain embodiments, this light chain human germline framework is selected from V1-11, V1-13, V1-16, V1-17, V1-18, 30 V1-19, V1-2, V1-20, V1-22, V1-3, V1-4, V1-5, V1-7, V1-9, V2-1, V2-1 1, V2-13, V2-14, V2-15, V2-17, V2-19, V2-6, V2-7, V2-8, V3-2, V3-3, V3-4, V4-1, V4-2, V4-3, V4-4, V4-6, V5-1, V5-2, V5-4, and V5-6. See PCT WO 2005/005604 for a description of the different germline sequences.
In other embodiments, the humanized antibody of the present invention may comprise or be derived from a human ger.mline heavy chain framework. In particular embodiments, this heavy chain human germline framework is selected from VH1-18, VHl-2, VH1-24, VH1-3, VH1-45, VH1-46, VH1-58, VH1-69, VH1-8, VH2-26, VH2-5, VH2-70, VH3-i l, VH3-13, VH3-15, VH3-16, VH3-20, VH3-21, VH3-23, VH3-30, VH3-33, VH3-35, VH3-38, VH3-43, VH3-48, VH3-49, VH3-53, VH3-64, VH3-66, VH3-7, VH3-72, V113-73, VH3-74, VH3-9, VH4-28, VH4-31, VH4-34, VH4-39, VH4-4, VH4-59, VH4-61, VH5-51, VH6-1, and VH7-81. See PCT WO 2005/005604 for a description of the different germline sequences.
In particular embodiments, the light chain variable region and/or heavy chain variable region comprises a framework region or at least a portion of a framework region (e.g., containing 2 or 3 subregions, such as FR2 and FR3). In certain embodiments, at least FRLl, FRL2, FRL3, or FRL4 is fully human. In other embodiments, at least FRHI, FRH2, FRH3, or FRH4 is fully human. In some embodiments, at least FRLl, FRL2, FRL3, or FRL4 is a gern-fline sequence (e.g., human germline) or comprises human consensus sequences for the particular framework. In other.embodiments, at least FRH1, FRH2, FRH3, or FRH4 is a germline sequence (e.g., human gern-dine) or comprises human cons-ensus sequences-for the particular framework.- In preferred embodiments, the framework region is a human framework region.
In general, humanized antibodies may be produced by obtaining nucleic acid sequences encoding the HCVR and LCVR of an antibody, e.g., a murine antibody or antibody made by a hybridoma, which binds a myostatin epitope of the invention, identifying the CDRs in said HCVR and LCVR (nonhuman), and grafting such CDR-encoding nucleic acid sequences onto selected human framework-encoding nucleic acid sequences. Optionally, a CDR region may be optimized by mutagenizing randoinnly or at particular locations in order to substitute, delete, or add one or more amino acids in the CDR prior to grafting the CDR region into the framework region. Alternatively, a CDR
region may be optimized subsequent to insertion into the human framework region using methods available to one of skill in the art. Preferably, the human framework amino acid sequences are selected such that the resulting antibody is likely to be suitable for in vivo administration in humans. This can be determined, e.g., based on previous usage of antibodies containing such human framework sequence. Preferably, the human framework sequence will not itself be significantly immunogenic.
Alternatively, the amino acid sequences of the frameworks for the antibody to be humanized may be compared to those of known human framework sequences the human framework sequences to be used for CDR-grafting and selected based on their comprising sequences highly similar to those of the parent antibody, e.g., a murine antibody which binds myostatin. Numerous human framework sequences have been isolated and their sequences reported in the art. This enhances the likelihood that the resultant CDR-grafted humanized antibody, which contains CDRs of the parent (e.g., murine) or optimized CDRs of the parent antibody grafted onto selected human frameworks (and possibly also the human constant region) will substantially retain the antigen binding structure and thus retain the binding affinity of the parent antibody. To retain a significant degree of antigen binding affinity, the selected human framework regions will preferably be those that are expected to be suitable for in vivo administration, i.e., not immunogenic.
In either method, the DNA sequence encoding the HCVR and LCVR regions of the preferably murine anti-myostatin antibody are obtained. Methods for cloning nucleic acid sequences encoding immunoglobulins are well known in the art. Such methods may, -for example, involve the amplification of the immunoglobulin-encoding sequences to be --cloned-using appropriate primers by polymerase chain reaction (PCR). Primers suitable for amplifying immunoglobulin nucleic acid sequences, and specifically murine HCVR
and LCVR sequences have been reported in the literature. After such immunoglobulin-encoding sequences have been cloned, they will be sequences by methods well known in the art.

After the CDR-encoding sequences are grafted onto the selected human framework encoding sequences, the resultant DNA sequences encoding the "humanized"
variable heavy and variable light sequences are then expressed to produce a humanized Fv or humanized antibody that binds myostatin. The humanized HCVR and LCVR may be expressed as part of a whole anti-myostatin antibody molecule, i.e., as a fusion protein with human constant domain sequences whose encoding DNA sequences have been obtained from a commercially available library or which have been obtained using, e.g., one of the above described methods for obtaining DNA sequences, or are in the art.

However, the HCVR and LCVR sequences can also be expressed in the absence of constant sequences to produce a humanized anti7myostatin Fv. Nevertheless, fusion of human constant sequences onto the variable region is potentially desirable because the resultant humanized anti-myostatin antibody may possess human effector functions.
Methods for synthesizing DNA encoding a protein of known sequence are well kiiown in the art. Using such methods, DNA sequences which encode the subject humanized HCVR and LCVR sequences'(with or without constant regions) are synthesized, and then expressed in a vector system suitable for expression of recombinant antibodies. This may be effected in any vector system which provides for the subject ' humanized HCVR and LCVR sequences to be expressed as a fusion protein with human constant domain sequences- and to associate to produce functional (antigen binding) antibodies or antibody fragments.
Human constant domain sequences are well known in the art, and have been reported in the literature. Preferred human constant light chain sequences include the kappa and lambda constant light chain sequences. Preferred human constant heavy chain sequences include human IgGI, human IgG2, human IgG3, human IgG4, and mutated versions thereof which provide for altered effector function, e.g., enhanced in vivo half-life, reduced Fc receptor binding or altered deamidation profile.
If present, human framework regions are preferably derived from a human antibody variable region having sequence similarity to the analogous or equivalent region of the antigen binding region donor (i.e., the parent antibody). Other sources of frainework regions for portions of human origin of a humanized antibody include human variable consensus sequences (see e.g., Kettleborough, C.A. et al. Protein Engineering 4:773-783 (1991); Carter et al., WO 94/04679. For example, the sequence of the antibody or variable region used to obtain the-nonhuman portion can be compared to human sequences as described in Kabat et al. Sequences of Proteins of Ininiunological Interest, Fifth Edition, NIH, U.S. Government Printing Office (1991). In a particularly preferred embodiment, the framework regions of a humanized antibody chain are derived from a human variable region having at least about 60% overall sequence identity, preferably at least about 70% overall sequence identity and more preferably at least about 85% overall sequence identity, with the variable region of the nonhuman donor.
A human portion can also be derived from a human antibody having at least about 65%
sequence identity, and preferably at least about 70% sequence identity, within the particular portion (e.g., FR) being used, when compared to the equivalent portion (e.g., FR) of the nonhuman donor.
References further describing methods involved in humanizing a mouse antibody that may be used are e.g., Queen et al., Proc. Natl. Acad. Sci. USA 88:2869, 1991; U.S.
Pat. No. 5,693,761; U.S. Pat. No. 4,816,397; U.S. Pat. No. 5,225,539; computer programs ABMOD and ENCAD as 'described in Levitt, M., J. Mol. Biol. 168:595-620, 1983;
humanization can be essentially performed following the method of Winter and co-workers [Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)], Human Antibodies As an alternative to humanization, human antibodies can be generated. Human antibodies can be produced using various techniques known in the art, including phage display libraries. Hoogenboom and Winter, J. Mol. Biol., 227:381 (1991); Marks et al., J.
Mol. Biol., 222:581 (1991). The techniques of Cole et al. and Boerner et al.
are also available for the preparation of human monoclonal antibodies. Cole et al., Monoclonal Antibodies and Caizcer Tlzerapy, Alan R. Liss, p. 77 (1985) and Boemer et al., J.
Immunol., 147(l):86-95 (1991). Similar-ly, human antibodies can be made by introducing -human immunoglobulin loci into transgenic animals, e.g., mice in which the endogenous inlmunoglobulin genes have been partially or complete inactivated. Upon immunization, e.g., with an antigen comprising an immunogenic epitope of the invention, a full repertoire of human antibody production is obtained, which closely resembles that seen in humans in all respects, including gene rearrangement, assembly and antibody repertoire.
This approach is described, for example, in U.S. Pat. Nos. 5,545,807;
5,545,806;
5,569,825; 5,589,369; 5,591,669; 5,625,126; 5,633,425; 5,661,016, and in-the following scientific publications: Marks et al., BioTechrzology 10:779-783, 1992;
Lonberg et al., Nature 368: 856-859, 1994; Morrison, Nature 368: 812-13, 1994; Fishwild et al., Nature Biotechnology 14:845-51, 1996; Neuberger, Nature Biotechnology 14: 826 (1996);
Lonberg and Huszar, b2tem. Rev. Itnmun.ol. 13: 65-93 (1995) and Jobkobovits et al., Proc.
Natl. Acad. Sci. USA, 90:2551, 1993.

Human immunoglobulin genes introduced into the mouse thus creating transgenic mice capable of responding to antigens with antibodies having human sequences are also described in Bruggemann et al. Proc. Nat'l. Acad. Sci. USA 86:6709-6713 (1989)]. There are several strategies that exist for the generation of mammals that produce human 5 antibodies. In particular, the're is the "minilocus" approach in which an exogenous Ig locus is mimicked through the inclusion of pieces (individual genes) from the Ig locus (see, e.g., U.S. Pat. Nos. 5,545,807, 5,545,806, 5,625,825, 5,625,126, 51633,425, 5,661,016, 5,770,429, 5,789,650, and 5,814,318, 5,612,205, 5,721,367, 5,789,215), YAC
introduction of large andsubstantially germline fragments of the Ig loci [See Mendez et 10 al. Nature Genetics 15:146-156 (1997), Green and Jakobovits J. Exp. Med.
188:483-495 (1998)], and-introduct-ion of entire orsubstantially entire loci through the use of microcell fusion (see European Patent Application No. EP 0 843 961 Al).
Any transgenic mouse capable of responding to immunization with antibodies having human sequences may be used to produce an anti-myostatin antibody of the 15 invention when using methods available to one skilled in the art, e.g., when such mouse is -, _ immunized with a polypeptide comprising an immunogenic epitope of the invention.

Uses Antibodies of the present invention are useful in therapeutic, prophylactic, 20 diagnostic and research applications as described herein. An antibody of the invention may be used to diagnose a disorder or disease associated with the expression of human myostatin. In a similar manner, the antibody of the invention can be used in an assay to monitor myostatin levels in a subject being treated for a myostatin-associated condition.
Diagnostic assays include methods that utilize the antibody of the invention and a label to 25 - detect myostatin--in a-sample, e.g., in a human-body fluid or in a cell or tissue extract.
Antibodies of the invention may be used with or without modification, and are labeled by covalent or non-covalent attachment of a detectable moiety. The detectable moiety can be any one that is capable of producing, either directly or indirectly, a detectable signal. For example, the detectable moiety may be a radioisotope such as, e.g., 3H,14C, 32P, 35S, or 30 125 1, a fluorescent or chemiluminescent compound, such as fluorescein isothiocyanate, rhodamine, or luciferin; or an exzyme, such as alkaline phosphatase, beta-galactosidase, or horseradish peroxidase. Any method known in the art for separately conjugating the antibody to the detectable moiety may be employed, including those methods described by Hun.ter, et al., Nature 144:945, 1962; David, et al., Biochemistry 13: 1014, 1974; Pain, et al., J. Iinmunol. Meth. 40: 219, 1981; and Nygren, J. Histochern. And Cytocheln. 30: 407, 1982.

A variety of conventional protocols for measuring myostatin, including e.g., ELISAs, RIAs, and FACS, are known in the art and provide a basis for diagnosing altered or abnormal levels of myostatin expression. Normal or standard expression values are established using any art known technique, e.g., by combining a sample comprising a myostatin polypeptide with, e.g., antibodies under conditions suitable to form a antigen:antibody complex. The antibody is directly or indirectly labeled with a detectable substance to facilitate detection of the bound or unbound antibody. Suitable detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, (3-galactosidase, or acetylcholinesterase;
examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an exainple of a luminescent material includes lununol; and examples of a radioactive material include 125I, 131I335S, or 3H. (See, e.g., Zola, Monoclonal Antibodies:
A Manudl of Techniques, CRC Press, Inc. (1987)).

The amount of a standard complex formed is quantitated by various methods, such as, e.g., photometric means. Amounts of myostatin polypeptide expressed in subject, control, and samples (e.g., from biopsied tissue) are then compared with the standard values. Deviation between standard and subject values establishes parameters for correlating a particular disorder, state, condition, syndrome, or disease with a certain level of expression (or lack thereof) for a myostatin polypeptide.
Once the presence of a disorder, state, condition, syndrome, or disease is established and a treatment protocol is initiated, assays are repeated on a regular basis to monitor the level of myostatin expression. The results obtained from successive assays are used to show the efficacy of treatment over a period ranging. from several days to months. With respect to a particular disorders (e.g., frailty or cachexia) the presence of an altered amount of myostatin in biopsied tissue or fluid (e.g., serum or urine) from a subject may indicate a predisposition for the development of a disorder, state, condition, syndrome, or disease or it may provide a means for detecting such a disorder, state, condition, syndrome, or disease prior to the appearance of actual clinical symptoms or it may define a population more likely to respond therapeutically to an antibody of the invention. A more definitive initial detection may allow earlier treatment thereby preventing and/or aineliorating further progression of a disease or disorder associated with myostatin expression, e.g., muscle or bone deterioration.
As a matter of convenience, the antibody of the present invention can be provided in a kit, a packaged combination of reagents in predetermined amounts with instructions for performing the diagnostic assay. Where the antibody is labeled with an enzyme, the kit will include substrates and cofactors required by the enzyme (e.g., a substrate precursor which provides the detectable chromophore or fluorophore). In addition, other additives may be included such as stabilizers, buffers (e.g., a blocking buffer or lysis buffer) and the like. The relative amounts of the various reagents may be varied widely to provide for concentrations in solution of the reagents which substantially optimize the sensitivity of the assay. Particularly, 'the reagents may be provided as dry powders, usually lyophilized, including excipients which on dissolution will provide a reagent solution having the appropriate concentration.

--- Therapeutic Uses for the Antibody Myostatin plays a role in muscle development and a number of related disorders or diseases. In adults, myostatin mRNA is primarily detected in skeletal muscle although lower coiicentrations are also found in adipose tissue and cardiac tissue (Sharma, M., et al, J. Cell Physiol. 180:1, 1999). Myostatin knockout mice have two- to three-fold greater - -muscle mass than their wild type littermates. The increased muscle mass is the result of fiber hypertrophy and hyperplasia (McPherron, A., et al. Nature 387:83-90, 1997 and Zhu, X. et al., FEBS Letters 474:71). In addition, the myostatin knockout mice accumulate less fat than their wild type littermates but otherwise appear normal and healthy. Myostatin has also been recently shown to be an important regulator of adipogenesis (Rebbapragada, A., et al., Mol. ai2d Cell. Bio. 23:7230-7242, 2003).
Additionally, bone structure and content has been recently studied in myostatin deficient mice (Hamrick M.W., et al., J. Orthopaedic Research 21:1025, 2003; Hamrick, M.W., et al., Calcif Tissue Int 71:63, 2002.
Therefore, a pharmaceutical composition comprising an anti-myostatin monoclonal antibody of the invention may be used to increase muscle mass, increase bone density, decrease muscle wasting, or may be useful for the treatment or prevention of conditions wherein the presence of myostatin causes or contributes to undesirable pathological effects or decrease of myostatin levels has a therapeutic benefit in mammals, preferably humans, including, but not limited to, muscle wasting, muscle injury, surgery, repair of damaged muscle, frailty, age-related sarcopenia, disuse atrophy,.osteoporosis, osteoarthritis, ligament growth and repair, obesity, suppression of body fat accumulation, obesity, muscular-dystrophy of any type, critical care myopaythy, alcoholic myopathy, cachexia (e.g., cancer-related or HIV-induced, or resulting from COPD, chronic lung disease, recovery from sepsis, renal failure, liver failure, cardiac failure or disease), metabolic syndrome, post-burn muscle wasting, and Type II diabetes. Disuse atrophy may result from numerous causes or incidents including any disorder or disease or state which leads to prolonged immobility or disuse or bed rest including, but not limited to, solid organ transplant, joint replacement, stroke, spinal cord injury, recovery from severe burn, sedentary chronic hemodialysis, post-sepsis recovery and exposure to microgravity.
Since myostatin is highly conserved in sequence and function across species, the antibodies of the invention may be used to increase muscle mass, increase bone density or treat or prevent conditions in non-human mammals or avian species [e.g., domestic animals (e.g., canine and feline), sports animals (e.g., equine), food-source animals (e.g., bovine, porcine and ovine), avian species (e.g., chicken, turkey, other game birds or poultry)] wherein the presence of myostatin causes or contributes to undesirable - pathological effects or decrease of myostatin levels has a therapeutic benefit.
The use of an anti-myostatin monoclonal antibody of the present invention for treating or preventing of at least one of the aforementioned disorders in which myostatin activity is detrimental or which benefits for decreased levels of bioactive myostatin is contemplated herein. Additionally, the use of an anti-myostatin monoclonal antibody of the present invention for use in the manufacture of a medicament for the treatment of at least one of the aforementioned disorders is contemplated.

As used herein, the terms "treatment", "treating'", and the like, refer to obtaining a desired pharmacologic and/or physiologic effect. The effect may be prophylactic in terms of completely or partially preventing a disease or symptom thereof and/or may be therapeutic in terms of a partial or complete cure for a disease and/or adverse affect attributable to the disease. "Treatment", as used herein, includes administration of a compound of the present invention for treatment of a disease or condition in a mammal, particularly in a human, and includes: (a)'preventing the disease from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it; (b). inhibiting the disease, i.e., arresting its development; and (c) relieving the disease;
i.e., causing regression of the disease or disorder or alleviating symptoms or complications thereof. Dosage regimens may be adjusted to provide the optimum desired response (e.g., a therapeutic or prophylactic response). For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation.

Phannaceutical Composition An antibody of the invention can be incorporated into pharmaceutical compositions suitable for administration to a subject. The compounds of the invention may be administered alone or in combination with a pharmaceutically acceptable carrier, diluent, and/or excipients, in single or multiple doses. The pharmaceutical compositions for administration are designed to be appropriate for the selected mode of administration, and pharmaceutically acceptable diluents, carrier, and/or excipients such as dispersing agents, buffers, surfactants, preservatives, solubilizing agents, isotonicity agents, stabilizing agents-and the like are used as appropriate.- Said compositions are designed in accordance with conventional techniques as in e.g., Remington, The Science and Practice of Pharnzacy, 19"' Edition, Gennaro, Ed., Mack Publishing Co., Easton, PA 1995 which provides a compendium of formulation techniques as are generally known to practitioners.
A pharmaceutical composition comprising an anti-myostatin monoclonal antibody of the present invention can be administered to a subject at risk for or exhibiting pathologies as described herein using standard administration techniques including oral, intravenous, intraperitoneal, subcutaneous, pulmonary, transdermal, intiamuscular, intranasal, buccal, sublingual, or suppository administration.
A pharmaceutical composition of the invention preferably is a "therapeutically effective amount" or a 'prophylactically effective amount" of an antibody of the 5 invention. A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result.
A
therapeutically effective amount of the aritibody may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the antibody or antibody portion to elicit.a desired response in the individual. A
therapeutically effective 10 amount is also one in which any toxic or detrimental effect of the antibody, are outweighed-by-the therapeutically-beneficial effects. -A "prophylactically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result. Typically, since a prophylactic dose is used in subjects prior to or at an earlier stage of disease, the prophylactically effective amount 15 will be less than the therapeutically effective amount.
A therapeutically-effective or'prophylactically-effective amount is at least the minimal dose, but less than a toxic dose, of an active agent which is necessary to impart therapeutic benefit to a subject. Stated another way, a therapeutically-effective amount of an antibody of the invention is an amount which in mammals, preferably humans, 20 increases muscle mass;-increases-bone-density, or treats-conditions wherein the presence of myostatin causes or contributes to undesirable pathological effects or decrease in myostatin levels results in a beneficial therapeutic effect in a mammal, preferably a human, including, but not limited to, muscle wasting, muscle injury, surgery frailty, age-related sarcopenia, disuse atrophy, osteoporosis, osteoarthritis, ligament growth and 25 -r-epair, obesity; suppression of body fat -accumulation, muscular dystrophy of any type, critical care myopaythy, cachexia (e.g., cancer-related or HIV-induced, or resulting from COPD, renal failure, liver failure, cardiac failure or disease), metabolic syndrome and Type II diabetes. Disuse atrophy may result from numerous causes or incidents including any disorder or disease or state which leads to prolonged immobility or disuse or bed rest 30 including, but not limited to, solid organ transplant, joint replacement, stroke, spinal cord injury, recovery from severe burn, sedentary chronic hemodialysis, post-sepsis recovery and exposure to microgravity.

The route of administration of an antibody of the present invention may be oral, parenteral, by inhalation, or topical. Preferably, the antibodies of the invention can be incorporated into a pharmaceutical composition suitable for parenteral administration.
The term parenteral as used herein includes intravenous, intramuscular, subcutaneous, rectal, vaginal, or intraperitoneal administration. Peripheral systemic delivery by intravenous or intraperitoneal or subcutaneous injection is preferred.
Suitable vehicles for such injections are straightforward in the art.
The pharmaceutical composition typically must be sterile and stable under the conditions of manufacture and storage in the container provided, including e.g., a sealed vial or syringe. Therefore, pharmaceutical compositions may be sterile filtered after making the formulation, or otherwise made microbiologically acceptable. A
typical composition for intravenous infusion could have a volume as much as 250-1000 ml of fluid, such as sterile Ringer's solution, physiological saline, dextrose solution and Hank's solution and.a therapeutically effective dose, (e.g., 1 to 100 mg/mL, or more) of antibody concentration. Dose may vary depending on the type and severity of the disease. As is well known in the medical arts, dosages for any one subject depends upon many factors, including the patient's size, body surface area, age, the particular compound to be administered, sex, tinle and route of adnunistration, general health, and other drugs being administered concurrently. A typical dose can be, for example, in the range of 0.001 to 1000 g; however, doses below or above this exemplary range are envisioned, especially considering the aforementioned factors. The daily parenteral dosage regimen can be about 0.1 g/kg to about 100 mg/kg of total body weight, preferably from about 0.3 g/kg to about 10 mg/kg and more preferably from about 1 g/kg to 1 mg/kg, even more preferably from about 0.5 to 10 mg/kg body weight per day. Progress may be monitored by periodic assessment. For repeated administrations over several days or longer, depending on the condition, the treatment is repeated until a desired suppression of disease symptoms occurs. However, other dosage regimens may be useful and are not excluded herefrom. The desired dosage can be delivered by a single bolus administration, by multiple bolus administrations, or by continuous infusion administration of antibody, depending on the pattern of pharmacokinetic decay that the practitioner wishes to achieve.
These suggested amounts of antibody are subject to a great deal of therapeutic discretion._ The key factor in selecting an appropriate dose and scheduling is the result obtained. Factors for consideration in this context include the particular disorder being treated; the particular mammal being treated, the clinical condition of the individual patient, the cause of the disorder, the_site of delivery of the antibody, the particular type of antibody, the method of administration, the scheduling of administration, and other factors known to medical pr'actitioners.
Therapeutic agents of the invention may be frozen or lyophilized for storage and reconstituted in a suitable sterile carrier p'rior to use. Lyophilization and reconstitution can lead to varying degrees of antibody activity loss. Dosages may have to be adjusted to compensate. Generally, pH between 6 and 8 is preferred.
Articles of Manufacture:
In another embodiment of the invention, an article of manufacture containing materials useful for the treatment or prevention of the disorders or conditions described above is provided. The article of manufacture comprises a container and a label. Suitable containers include, for. example, bottles, vials, syringes, and test tubes.
The containers may be formed from a variety of materials such as glass or plastic. The container holds a composition of the invention which is effective for preventing or treating the disorder or condition and may have a sterile access port (for example the container may be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). The active -agent in the composition is an-anti-myostatin antibody of the invention. The label on, or associated with, the container indicates that the composition is used for treating the condition of choice. The article of manufacture may further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution and dextrose solution. It may further include other materials-desi-r-able-from a commercial and user-standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for use.
The following examples are offered for illustrative purposes only, and are not intended to limit the scope of the present invention in any way.

EXAMPLES
Example 1: ELISA Assays A. Anti-myostatin Fabs preferentially bind myostatin over GDF-11.
Anti-myostatin Fabs (or full-length mAbs) of the present invention are tested in an ' ELISA assay, in which binding of the Fab to mature myostatin (dimeric form) coated at various concentrations on a 96-well plate is measured. Binding of the Fabs to GDF-11 is also tested.

Each well of two 96-well plates is coated with 200 ng/well recombinant mouse myostatin (R&D systems, Cat. #788-G8/CF, carrier-free, in carbonate buffer, pH
9.6) or 200 ng/well recombinant human GDF-11 (Peprotech, Inc., Cat. # 120-11, carrier-free, in carbonate buffer, pH 9.6). The plates are incubated at 4 C overnight. The wells are aspirated and washed twice with washing buffer (20 mM Tris (hydroxymethyl) aminomethane, pH 7.4, 0.15 M NaC1,-0.1% Tween-20). The plates are blocked with l blocking buffer per well (5% Carnation Instant milk in the above washing buffer) for 5 hours.

Fabs or mabs to be tested are diluted into blocking buffer at various concentrations, e.g., 10 g/ml, 2 g/ml, 0.4 g/ml, 0.08 g/ml, and 0.016 g/ml. Fifty microliters of each Fab solution is added to the GDF-8 and GDF-11 coated wells in duplicate. The plates are incubated for 1 hour at room temperature. The wells are then washed 3 times with washing buffer. AP-conjugated secondary antibody (50 l goat anti-kappa AP (Southern Biotech), diluted 1:1000 in blocking buffer) is added to each well and incubated for 1 hour at room temperature. The wells are then washed 3 times with washing buffer. Fifty microliters of chromogenic substrate (i.e., AP
substrate) is added to each well and allowed to develop at room temperature. The absorbance of the wells is read at OD of 560 nm. The average absorbance from duplicate wells is determined.
All antibodies of the invention are contemplated to bind to plate-bound human mature myostatin and preferentially bind to myostatin when compared to GDF-1 1 binding. In a GDF-8 down ELISA, Fab C12 has an IC50 of 0.25 g/ml and Fab has an IC50 of 0.27 Rg/ml.

B. Humanized-optimized anti-myostatin antibodies bind myostatin peptide In an ELISA assay, antibodies of the invention bind a purified, biotinylated peptide with the sequence that spans amino acids 40-64 of mature human myostatin (SEQ
ID NO: 53) with the exception that an aminobutyric acid is at position 47 in-stead of the cysteine residue. Antibodies of the invention also bind a peptide with the sequence shown in SEQ, ID NO: 62 (with cysteine or serine at position 47 of mature myostatin) as well as the mature form of myostatin in monomeric and dimeric forms.
In an exemplary ELISA assay, 50 l of a 1 g/mi solution of goat anti-human kappa antibody (UNLB, Southern Biotech 2060-01) in 50mM sodium bicarbonate buffer, pH 8.0 is used to coat each well of a high-bind microtiter plate (Greiner EK
20061). The wells are aspirated and washed with PBST (PBS, 0.1% Tween) and then blocked for 1 hour with PBST-BSA (PBS, 1% BSA, 0.1% Tween). The wells are then washed again with PBST. All steps of the assay are performed at room temperature.
Titrations of humanized-optimized anti-myostatin antibody (full-length antibody comprising an IgG4 -fic-region,-tr-ansiently expressed in 293 cells), diluted in PBST-BSA
beginning at 50 ng/well are added to the wells and the plate is further incubated for 1 hr, washed as above and probed with 50 l of the biotinylated peptide described above at a concentration of 50 nM (diluted in PBST-BSA). Then 50 l of a 2 g/ml NeutrAvidin-AP (1:1000 in PBST-BSA, Pierce 31002) is added and detection is achieved by addition of AP
substrate according to manufacturer's instructions (Pierce 31002). Buffer alone is used as a control. Absorbance is read at 560 nm. The 510C2 and C12 Fabs bind the peptide with IC50 values of 6.73 and 5.42 ng/well respectively.
Antibodies of the invention are able to bind a peptide consisting of amino acids 40-64 (SEQ ID NO: 53) of mature myostatin and/or a peptide consisting of amino acids 43-57 (SEQ ID NO: 62) of mature myostatin (with a cysteine or serine amino acid at position 47). However, antibodies of the invention are not able"to bind (at levels significantly greater than background), peptides consisting of amino acids 60-73, 74-86, 87-97, 96-108, 16-29, 1-15 or 30-42 of mature myostatin. The murine antibody named --Mab-3-(see U.S. patent applications 60/559,621 and 60/555,456 incorporated herein), comprising murine variable regions (see Figs 10 and 11) operably attached to murine IgGl Fc region - does not bind peptides consisting of amino acids 60-73, 74-86, *87-97, 96-108, 16-29, 1-15 or 30-42 of mature myostatin and is the parent antibody for antibodies of the invention.

Humanized antibodies 510C2 and C12 bind peptides consisting of amino acids 43-57 and 45-59 of mature myostatin (as shown in Table 1 below) to a similar degree.
This demonstrates that the epitope of the invention is more specifically localized within the peptide spanning amino acids 45-57 of mature myostatin. Binding is slightly reduced when the antibodies are tested for binding to a peptide consisting of amino acids 47-61 of mature myostatin, demonstrating importance of amino acids 45-46 of mature myostatin for binding or for preserving conformation of the peptide that allows for binding. Binding 5 is further reduced when antibodies are tested for binding to a peptide consisting of amino acids 49-63 of mature myostatin, demonstrating importance of amino acids 47-48 of mature myostatin for binding or for preserving conformation of the peptide that allows for binding. Binding is reduced to background levels when the antibodies are tested for binding a peptide consisting of amino acids 39-53 of mature myostatin, suggesting the 10 importance of the region from residues 54-57.
Table 1 Relative Seq Peptide Residues Binding Id No:
15 ANYCSGECEFVFLQKYPHTHLVHQA 40-64 +++ 53 CSGES*EFVFLQKYPH 43-57 +++ 62 GECEFVFLQKYPHTH 45-59 +++ 92 CEFVFLQKYPHTHLV 47-61 ++ 93 FVFLQKYPHTHLVHQ 49-63 + 94 (* residue is C in myostatin) C. Myostatin Humanized-Optimized Mab ELISA AssaX

25 Humanized anti-human myostatin full-length monoclonal antibodies 510C2 and C12 are tested in an ELISA assay, in which binding of the antibody to the GDF-8 antigen coated on a plate is measured. Cross-reactivity of the Mabs to GDF-11 is also tested. A
no Mab condition, an isotype control antibody, and an irrelevant protein (HGF) are used as negative controls.

30 Each well of 96-well plates is coated with 70 gl of human GDF-8 (carrier-free, lug/ml in carbonate buffer, pH 9.6), recombinant human GDF-11 (from Peprotech, Inc., catalog # 120-11, carrier-free, lug/ml in carbonate buffer, pH 9.6), or recombinant human HGF (from R&D Systems, catalog # 294-HG/CF, carrier-free, lug/ml in carbonate buffer, pH 9.6). The plates are incubated at 4 C overnight. The wells are aspirated and washed 35 twice with washing buffer (20 mM Tris (hydroxymethyl) aminomethane, pH 7.4, 0.15 M
NaCl, 0.1% Tween-20). The plates are blocked with 200u1 blocking buffer per well (5%

Carnation Instant milk in the above washing buffer) for 4 hours at room temperature.
Plates are washed twice with washing buffer.
Mabs are diluted into blocking buffer at 1 g/ml, 0.2 g/ml, 0.04 ug/ml, 0.008 ug/ml, 0.0016 g/ml, and 0.00032 g/ml. A no Mab control is used, which consists of blocking buffer alone. An isotype control antibody is also used as a negative control, at 1ug/ml. Then 50 l of each Mab solution is added to the GDF-8 and GDF-11 coated wells in duplicate. The plates are incubated for 1 hour at room temperature.
The wells are then washed 3 times with washing buffer.
Then 50 1 peroxidase-conjugated secondary antibody (goat anti-human IgG HRP, Fc gamma fragment specific, Jackson ImmunoResearch, catalog # 109-035-008), diluted 1:2000 in blocking buffer, is added to each well and incubated for 1 hour at room temperature. The wells are then washed 3 times with washing buffer. Then 50 l of chromogenic substrate (i.e., OPD substrate) is added to each well and allowed to develop at room temperature for 2.5 minutes. The reaction is stopped by adding 100 l 1N HCl to each well. The absorbance of the wells is read at 490 nm. The average absorbance from duplicate wells is determined, and these values are listed in Table 2, below.
These data demonstrate that Mabs 510C2 and C12 bind to plate-bound myostatin.
With regards to specificity, these Mabs preferentially bind GDF-8 over GDF-11. Mab 510C2 shows approximately 25-fold greater binding to myostatin (GDF-8) than to GDF-11. Mab C 12 shows approximately 5-fold greater binding to myostatin than to GDF- 11.
Table 2 1 ug/m1 1.62275 0.67645 0.0723 1.5785 1.3021 0.0665 0.2ug/mI 1.20585 0.24605 0.071 1.37095 0.87605 0.0543 0.04ug/ml 0.55695 0.11485 0.07525 0.8177 0.34495 0.0564 0.008ug/ml 0.2482 0.09125 0.07105 0.32805 0.11915 0.0507 0.0016ug/ml 0.11425 0.09385 0.07255 0.1257 0.07 0.058 0.00032ug/ml 0.0876 0.08245 0.0722 0.07415 0.05475 0.0554 no Mab 0.08645 0.0936 0.0721 0.0544 0.05185 0.0522 isotype 0.09075 0.0919 0.0804 0.05185 0.0487 0.049 control Furthermore, full-length mab C12 was demonstrated in an ELISA assay to not bind other TGF-(3 superfamily members BMP-2, BMP-3, BMP-3b/GDF-10, BMP-4, BMP-7, BMP-8B, Activin A, Activin B, TGF-a, TGF- (31, TGF- (32 and GDF-3; it demonstrated minimal binding to BMP-5. It is contemplated that other anti-myostatin antibodies of the invention similarly do, not bind or m.inimally bind these other TGF-(3 superfamily members.

Example 3 Myostatin Neutralization AssU
Ectodermal explants. are removed from stage 8-9 blastula Xenopus embryos by standard procedures and cultured in 0.5X MBS (1X MBS: 88 mM NaCI, 1 niM KCI, 0.7 mM CaClz, 1 mM MgSO4, 5 mM HEPES, 2.5 mM NaHCO3; 1:1000 v/v gentamycin, 0.1% bovine serum albumin) with the addition of growth factor (GDF8 or GDF1.1) plus or indicated, for 18 hours at 18 C, by which time control embryos reach the early neurula stage (stage 15-16). Explants are photographed and the length of each explant is measured using an image analysis algorithm designed for animal cap quantitation.
Explants not treated with either growth factor or Fab (controls), round into balls of epidermis. Myostatin and GDF-11 induce mesoderm in these ectodermal explants which causes the explants to elongate and form dumbbell-like structures. Antibodies or Fabs, when tested for neutralizing activity, are added to the culture medium containing myostatin for the entire length of the culture period and their ability to inhibit the growth factor-induced elongation movements is assessed. Myostatin is added to the explants at ng/.ml. Antibodies or Fabs to be tested are added at 20 g/ml. A Fab generated to an irrelevant antigen is used as a control. A commercially available monoclonal anti-mouse 20 GDF8 antibody may be tested as a control, this antibody is produced in goats inununized with purified mouse GDF8 and demonstrated by the manufacturer to neutralize elongation of Xenopus animal caps elicited by 25 ng/ml of murine GDF8 when present at about 10-20 g/m1(R&D Systems Cat. #MAB788).
ImagePro (v4.5.1.22, from Media Cybernetics) is used for the image processing.
25 A macro is written to automate the image processing. The macro processes the image and records length in units of bits. Alternative measuring methods may be used as known in the art. Antibodies of the invention are contemplated to neutralize GDF8 activity in the animal cap assay and have no GDF- 11 neutralization activity.

Example 4: Affinity Measurement of Fabs The affinity (KD) and koõ and koff rates of anti-myostatin Fabs of the present invention are measured using a BIAcore" 2000 instrument containing a CM4 sensor chip.

The BlAcore utilizes the optical properties of surface plasmon resonance to detect alterations in protein concentration of interacting molecules within a dextran biosensor matrix. Except where noted, all reagents.and materials are purcliased from BIAcore AB
(Upsala, Sweden). All measurements are performed at 25 C. Samples containing Fabs are dissolved in HBS-EP buffer (150 mM sodium chloride, 3 mM EDTA, 0.05% (w/v) surfactant P-20, and 10 mM HEPES, pH 7.4). Myostatin or GDF-11 (R&D Systems) is immobilized onto flow cells of a CM4 chip using amine-coupling chemistry. Flow cells (1-4) are activated with a 1:1 mixture of 0.1 M N-hydroxysuccinimide and 0.1 M
3-(N,N-dimethylamino)propyl-N-ethylcarbodiimide at a flow rate of 20 l/min.
Myostatin or GDF-1 1 (2.5 g/mL in 10mM sodium acetate, pH 4.5) is manually injected over individual flow cells at a flow rate of 10 L/min. The surface density is monitored and until each flow cell reaches a surface density of - 150 response units (RU).
Surfaces are blocked with a 50 l injection of 1 M ethanolanline-HCI, pH 8.5 (10 L/min).
To ensure complete removal of any noncovalently bound myostatin or GDF-11, 15 l of 10 mM
glycine, pH 1.5 is injected twice. Running buffer used for kinetic experiments contained 10n-ilVl HEPES, pH 7.4, 150mM NaCl, 0.005% P20.
Collection of kinetic binding data is performed at maximum flow rate (100 gl/min). Each analysis cycle consists of (i) 250 l injection of a Fab (concentration range of 50 nM to 0.4 nM in 2-fold dilution increments) over all 4 flow cells with flow cell 1 as the reference flow cell, (ii) 20 min dissociation (buffer flow), (iii) regeneration of the GDF-8 or GDF-11 surface with a two 15 l injection of 10 mM glycine, pH 1.5, (iv) a 15 l blank injection of running buffer, and (v) a 2 min stabilization time before start of next cycle. Signal is monitored as flow cell 2 minus flow cell 1, flow ce113 minus flow cell 1 and flow ce114 minus flow cell 1. Samples and a buffer blank are injected in duplicate in a random order. Data are processed using SCRUBBER (Center for Biomolecular Interaction Analysis, Univ. of Utah) software. Association and dissociation rates for each cycle are determined by fitting of the biosensor data using to a simple association model using ClampXP (Center for Biomolecular Interaction Analysis, Univ. of Utah) to extract the ko,t and k~ff rate constants; the equilibrium binding constant Kd is calculated using the relationship Kd = k~fj/ko,,. The affinity data measured for myostatin binding to the 510C2 Fab are: koõ of 5.4 x 105 M-Is koff of 4.67 x 10-4 sec"1, and Kd (calc) of 0.87 nM. Binding to GDF-11 by 510C2 Fab was not observed.

Example 5: Affinity Measurement of Mabs Binding affinity measurements for full-length monoclonal antibodies of the invention are determined using a Sapidyne KINEXA assay. NHS-activated fast-flow sepharose beads (GE Healthcare) are pre-coated with an antibody of the invention (50 g anti-myostatin antibody per ml of beads) and blocked with 10 mg/ml BSA in 1 M
Tris-HCl, pH 8Ø Then 20 pM and 50 pM of an antibody of the invention is incubated with various concentration (e.g., 2.4 pM to 20 nM, serial dilutions) of myostatin in running buffer (PBS, 0.005% (v/v) Tween-20, 1 mg/ml ovalbumin) for 10 hours at room temperature. To determine the free antibody present at equilibrium, each sample is passed through the myostatin-coated beads. The amount of bead-bound antibody is then quantified by passing a solution of fluorescent (Cy5) labeled goat ant-human Fc antibody (Jackson Immuno Research) diluted 1:4000 in running buffer over the beads. The measured fluorescence signal is proportional to the concentration of free antibody at equilibrium. Each concentration of myostatin is measured in duplicate. The equilibrium dissociation constant (KD) is obtained from non-linear regression of the competition curves using a multiple-curve, one-site homogeneous binding model (KINEXA
software) and data is presented with 95% confidence intervals (CI).
The association rate constant (koõ) for GDF-8 binding is also determined using a Sapidyne KINEXA assay. Twenty pM antibody is mixed with 1 nM GDF-8 using the same conditions described above. At various time points, samples are probed for free antibody using the conditions described above for equilibrium binding, and then the resulting time dependence is fit using the KINEXA software to determine the association rate (kon). The dissociation rate constant (koff) is calculated using the expression koff = KD
x ko,,. When full-length 510C2 and C12 (operably linked to a IgG4 Fc region) is measured using the described assay, the results are as stated in Table 3 below.
Table 3 Mab Kd, pM (95CI) kon, M- , s" (95% CI) koff, s, calculated 510C2 80(41-134) 1.38 x 10 1.11 x 10"
(1.08-1.75 x 105) C12 108 (23-294) 7.42 x 10 8.02 x 10 (6.83-8.01 x 104) Example 6 Myostatin/SBE Reporter Assay In this reporter assay, a plasmid encoding a reporter gene, i.e., luciferase gene, downstream of a SMAD binding element ("SBE"), more specifically (CAGA)12 expresses 5 luciferase protein when a molecule such as myostatin, GDF- 11, or other TGF-(3 superfamily member binds its own receptor, thereby triggering SMAD signaling which results in a phosphorylated SMAD complex which is capable of binding the SBE.
The CAGA sequence was previously reported to be a TGF-(3 responsive sequence within the promoter of the TGF-0 induced gene PAI-1 (Denner et al., EMBO J., 17:3091-3 100, 10 1998). The amount of active myostatin exposed to the cells is directly proport ional to the quantity of luciferase enzyme produced which is directly proportional to the quantity of light produced and measurable. The presence of an inhibitor (e.g., an antibody that binds myostatin) reduces the quantity of myostatin able to activate the SBE which ultimately results in a reduced production of light. This assay is also described in International 15 Publication Number WO 2004/037861 incorporated herein.
It is contemplated that a Myostatin/SBE Reporter Assay not be limited to the exact conditions described herein, other types of cells may be used, e.g., 293HEK
(ATCC) or A204 Rhabsomyosarcoma cells (see, e.g., Whittemore, et al. BBRC, 200:965-71, 2003);
other types of reporters may be used, e.g., CAT, (3-gal, GFP, and other growth conditions 20 for the cells and assay conditions including varying amounts of myostatin in the reaction may be used. One of skill in the art would readily be able to discern if an assay falls within the scope of a myostatin/SBE reporter assay for it would have a vector comprising an SBE element upstream of a reporter gene introduced into a host cell, wherein the SBE
element used is responsive to the SMAD produced in response to myostatin binding the 25 myostatin receptor. R.S. Thies, et al., Growtlz Factors, 18:251-259, 2001, describes a similar assay while, Wittemore, L. et al., BBRC, 300:965-971, 2003 describes the SBE
element response to the SMAD produced in response to myostatin binding its receptor.
In this assay, 293E cells (Edge Biosystems) in DMEM/F12 media (3:1) (Gibco 93-0152DK), 10% FBS, 20mM Hepes, 4mM L-glutamine are seeded at about 25000 cells per 30 well in poly-lysine coated inner wells of a 96-well plate (BD Biocoat 35-4461) and incubated overnight at 37 C. The following day, the cells are washed in PBS
and 50 l OptiMEM I (Gibco 31985-070) is added per well. The cells are transfected with 50 l of the following SBE-luciferase DNA mixture: (i) 80 l Lipofectamine (Gibco 11668-combined with 1.5 ml OptiMEM and allowed to sit for 5 minutes then added to (ii) a tube in which 20 g SBE luciferase DNA is combined with 1.5 ml OptiMEM and 200 l Plus reagent, mixed and allowed to sit for 5 minutes. After the two mixtures are added together, the solution is mixed vigorously and allowed to stand for 30 minutes before 50 l is added to each well. The cells are then incubated overnight at 37 C in 5%
CO2. For each plate of cells, 5 ml of myostatin (R&D Systems 788-G8) is diluted to 20 ng/ml in .complete medium. Each antibody of the inveintion to be tested is titrated in complete medium, e.g., from about 40 g/ml to about 50 ng/ml. The transfection media is removed from the wells and 50 l of an antibody dilution is added per well and 50 l of GDF-8 (myostatin) or GDF-11 (R&D Systems) is added per well. The plate of cells is then incubated overnight at 37 C in 5% CO2. The following day, the media is aspirated, the cells are washed in PBS and 75 [tl lysis buffer (Promega E266A) is added. The luciferase activity in the cell lysate is measured using Luciferase Reagent according to manufacturer's instructions (Promega E2620). Luminescence is plotted against Loglo Mab concentration ([tg/ml) and the IC50 for each Mab for myostatin and GDF-11 is calculated.
Monoclonal antibodies 510C2, C12 when tested in these conditions with GDF-8 yield IC50 values (from an average of two tests) of 5.15 nM (5.74 and 4.57 nM) and 16.07 nM (23.04 and 9.12 nM) respectively. Neither 510C2 nor C12 mAbs demonstrate neutralization activity in this assay when tested with GDF-11 instead of GDF-8.
Example 7 Pharmacokinetics The pharmacokinetics (PK) of the antibodies of the invention may be evaluated in C57B6/SCID mice at a dose of 1 mg/kg after a single intravenous (IV) or intraperitoneal (IP) administration. The animals receive a mixture of unlabeled and I''5I-labeled antibody at a dose described above and serum concentration is determined based on I'5I
radioactivity in the serum and the specific activity of the injected dose.
Serum concentration of the antibody administered either IV or IP versus time is plotted.
Exampie 8 In vivo effect on Muscle Mass and Strength To determine whether an antibody of the invention blocks myostatin activity in vivo, an antibody of the invention may be tested in adult SCID mice. SCID mice suffer from severe combined immune deficiency, and therefore do not.generate an immunological reaction following injection of an antibody of the invention.
Muscle mass is used as an indicator for myostatin activity in mice treated with an antibody of the invention.
Male C57B6 SCID eight weeks old mice are weighed and evenly distributed with respect to body weight into groups of eight. An antibody of the invention in PBS buffer is injected into the mice IP at various doses (e.g., 60, 10, 5 and 1 mg/kg) weekly. PBS-treated or untreated mice are used as controls. The treatments continue for 4-12 weeks.
Muscle mass is assessed by dissecting and weighing the gastrocnemious and quadriceps muscles following treatment.
To determine muscle strength, the strength of the front limb is measured with a grip strength. test meter (e.g., model 1027 csx, Columbus Instruments).

DEMANDE OU BREVET VOLUMINEUX

LA PRESENTE PARTIE DE CETTE DEMANDE OU CE BREVET COMPREND
PLUS D'UN TOME.

NOTE : Pour les tomes additionels, veuillez contacter le Bureau canadien des brevets JUMBO APPLICATIONS/PATENTS

THIS SECTION OF THE APPLICATION/PATENT CONTAINS MORE THAN ONE
VOLUME

NOTE: For additional volumes, please contact the Canadian Patent Office NOM DU FICHIER / FILE NAME:

NOTE POUR LE TOME / VOLUME NOTE:

Claims (21)

1. An anti-myostatin monoclonal antibody that specifically binds myostatin with an affinity no greater than about 3 × 10 -8 M, wherein said antibody neutralizes or antagonizes at least one myostatin activity.
2. The monoclonal antibody of Claim 1, wherein the monoclonal antibody binds myostatin with an affinity no greater than about 9 × 10 -10 M.
3. An anti-myostatin monoclonal antibody that has an IC50 of less than or equal to about 25 nM in an in vitro myostatin/SBE assay.
4. The monoclonal antibody according to any one of Claims 1 to 3, wherein the monoclonal antibody binds myostatin within the domain spanning amino acids 40 to 64 of mature myostatin.
5. The monoclonal antibody according to any one of Claims 1 to 3 wherein the monoclonal antibody binds a polypeptide consisting of an amino acid sequence as shown in the group consisting of: ANYCSGECEFVFLQKYPHTHLVHQA (SEQ
ID NO: 53) and CSGECEFVFLQKYPH (SEQ ID NO: 62).
6. A monoclonal antibody according to any one of Claims 1 to 5, which preferentially binds a myostatin polypeptide over a GDF-1 1 polypeptide.
7. The monoclonal antibody according to any one of Claims 1 to 6, wherein said antibody is cross-reactive at 35% or less with a GDF-11 polypeptide.
8. The monoclonal antibody according to any one of claims 1 to 7, wherein said antibody is a chimeric, humanized or fully human antibody.
9. The monoclonal antibody according to any one of claims 1 to 8, wherein said antibody competes for binding myostatin in a competition ELISA assay with an antibody comprising two polypeptides with the sequences shown in the group consisting of: (i) SEQ ID NOs: 5 and 15, (ii) SEQ ID NOs: 5 and 16, and (iii) SEQ
ID NOs: 10 and 26.
10. The monoclonal antibody according to any one of claims 1 to 9 comprising two polypeptides with a sequence selected from the group consisting of:
(i) SEQ ID NO: 6 and SEQ ID NO: 14;
(ii) SEQ ID NO: 5 and SEQ ID NO: 13, 14, 15, 16, 17, 18, 19 or 20;
(iii) SEQ ID NO: 7 and SEQ ID NO: 21, 22, 23, 24, 25 or 56;
(iv) SEQ ID NO: 8, 9, 10, 11 or 12 and SEQ ID NO: 55.
11. The monoclonal antibody according to any one of Claims 1 to 9, wherein said antibody comprises a HCVR and a LCVR, wherein the HCVR comprises a peptide at CDRH1 with a sequence as shown in SEQ ID NO: 59, a peptide at CDRH2 with a sequence as shown in SEQ ID NO: 60, and a peptide at CDRH3 with a sequence as shown in SEQ ID NO: 61, and wherein the LCVR comprises a peptide at CDRL1 with a sequence as shown in SEQ ID NO: 57, a peptide at CDRL2 with a sequence as shown in SEQ ID NO: 30 or SEQ ID NO: 52, and a peptide at CDRL3 with a sequence as shown in SEQ ID NO: 58.
12. The monoclonal antibody of Claim 11, further comprising a human framework region.
13. The monoclonal antibody of either of Claims 11 or 12, wherein said CDRH1, CDRH2, CDRH3, CDRL1, CDRL2 and CDRL3 comprise a peptide as shown in Figure 9 herein in the corresponding CDR position.
14. The monoclonal antibody according to any one of claims 1 to 13, wherein the monoclonal antibody is a full-length antibody, a substantially intact antibody, a Fab fragment, a F(ab')2 fragment or a single chain Fv fragment.
15. The monoclonal antibody according to any one of claims 1 to 13, wherein the monoclonal antibody further comprises a heavy chain constant region selected from the group consisting of IgG1, IgG2, IgG3, IgG4, IgA, IgE, IgM and IgD.
16. The monoclonal antibody according to any one of claims 1 to 15 wherein the constant region present in the antibody originates from the genome of an animal selected from the group consisting of domestic animals, sports animals and food-source animals.
17. A pharmaceutical composition comprising the antibody according to any one of claims 1 to 16.
18. The pharmaceutical composition of claim 17 further comprising a pharmaceutically acceptable carrier.
19. An antibody according to any one of claims 1 to 16 for use as a medicament.
20. Use of an effective amount of antibody according to any one of claims 1 to 16 in the preparation of a medicament for increasing muscle mass or increasing bone density in a subject in need thereof.
21. Use of an effective amount of antibody according to any one of claims 1 to 14 in the preparation of a medicament for the treatment or prevention of one or more conditions selected from frailty, cachexia, muscle wasting, muscle weakness, myopathy, muscular dystrophy, osteoporosis, COPD, renal failure or disease, liver failure or disease, cardiac failure, type II diabetes or metabolic syndrome.
CA002624976A 2005-10-06 2006-10-05 Anti-myostatin antibodies Abandoned CA2624976A1 (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US72467005P 2005-10-06 2005-10-06
US60/724,670 2005-10-06
US72523505P 2005-10-11 2005-10-11
US60/725,235 2005-10-11
US72606205P 2005-10-12 2005-10-12
US60/726,062 2005-10-12
PCT/US2006/038818 WO2007044411A2 (en) 2005-10-06 2006-10-05 Anti-myostatin antibodies

Publications (1)

Publication Number Publication Date
CA2624976A1 true CA2624976A1 (en) 2007-04-19

Family

ID=37943347

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002624976A Abandoned CA2624976A1 (en) 2005-10-06 2006-10-05 Anti-myostatin antibodies

Country Status (13)

Country Link
US (2) US7635760B2 (en)
EP (1) EP1951756B1 (en)
JP (1) JP5052517B2 (en)
KR (1) KR101135220B1 (en)
CN (1) CN101277976B (en)
AU (1) AU2006302494B2 (en)
BR (1) BRPI0616923A2 (en)
CA (1) CA2624976A1 (en)
EA (1) EA015589B1 (en)
ES (1) ES2533464T3 (en)
IL (1) IL189579A0 (en)
NO (1) NO20082073L (en)
WO (1) WO2007044411A2 (en)

Families Citing this family (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2533464T3 (en) * 2005-10-06 2015-04-10 Eli Lilly And Company Anti-myostatin antibodies
UA92504C2 (en) 2005-10-12 2010-11-10 Эли Лилли Энд Компани Anti-myostatin monoclonal antibody
WO2007114319A1 (en) 2006-03-31 2007-10-11 Chugai Seiyaku Kabushiki Kaisha Method for control of blood kinetics of antibody
PL2066695T3 (en) * 2006-09-05 2013-08-30 Lilly Co Eli Anti-myostatin antibodies
US11103497B2 (en) 2007-06-01 2021-08-31 Wyeth Llc Treatment of imatinib resistant leukemia
EP2202245B1 (en) 2007-09-26 2016-08-24 Chugai Seiyaku Kabushiki Kaisha Method of modifying isoelectric point of antibody via amino acid substitution in cdr
KR100857861B1 (en) * 2007-10-15 2008-09-11 주식회사 바이오리더스 Surface expression vector for fusion protein of myo-2 peptide multimer and myostatin, and microorganism transformed by therof
PE20091163A1 (en) 2007-11-01 2009-08-09 Wyeth Corp ANTIBODIES FOR GDF8
SI2275443T1 (en) 2008-04-11 2016-03-31 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
CA2735193A1 (en) * 2008-08-26 2010-03-11 Macrogenics, Inc. T-cell receptor antibodies and methods of use thereof
TW201029662A (en) 2008-12-19 2010-08-16 Glaxo Group Ltd Novel antigen binding proteins
JO3340B1 (en) * 2010-05-26 2019-03-13 Regeneron Pharma Antibodies to human gdf8
AR081556A1 (en) 2010-06-03 2012-10-03 Glaxo Group Ltd HUMANIZED ANTIGEN UNION PROTEINS
MX2013001845A (en) * 2010-08-16 2013-08-27 Amgen Inc Antibodies that bind myostatin, compositions and methods.
AR082641A1 (en) 2010-09-07 2012-12-19 Imclone Llc ANTI-VEGFR-3 ANTIBODY COMPOSITIONS
TWI654204B (en) 2010-11-30 2019-03-21 中外製藥股份有限公司 Antibody with calcium-dependent antigen binding ability
CN102212545B (en) * 2011-04-07 2013-05-08 北京济福霖生物技术有限公司 Method for knocking out cattle beta-lactoglobulin gene by using zinc finger nucleases (ZFNs)
CN102260711B (en) * 2011-06-24 2013-09-18 北京济福霖生物技术有限公司 Method for knocking out bovine myostatin gene by using zinc finger nuclease
US8871209B2 (en) 2011-11-14 2014-10-28 Regeneron Pharmaceuticals, Inc. Compositions and methods for increasing muscle mass and muscle strength by specifically antagonizing GDF8 and or Activin A
CN102660577B (en) * 2012-04-18 2013-12-25 山东省农业科学院奶牛研究中心 Method for knocking out beta 6 subunit genes of bovine integrins by utilizing zinc finger nucleases (ZFNs)
JP6774164B2 (en) 2012-08-24 2020-10-21 中外製薬株式会社 Mouse FcγRII specific Fc antibody
NZ705370A (en) 2012-08-24 2018-06-29 Chugai Pharmaceutical Co Ltd Fcγriib-specific fc region variant
MY189682A (en) 2012-09-13 2022-02-25 Bristol Myers Squibb Co Fibronectin based scaffold domain proteins that bind to myostatin
JPWO2014119634A1 (en) * 2013-01-30 2017-01-26 国立研究開発法人国立精神・神経医療研究センター Muscle increasing agent and pharmaceutical composition containing the same
EP3783017A1 (en) 2013-04-02 2021-02-24 Chugai Seiyaku Kabushiki Kaisha Fc region variant
HUE052232T2 (en) * 2013-05-06 2021-04-28 Scholar Rock Inc Compositions and methods for growth factor modulation
TW201920262A (en) 2013-07-30 2019-06-01 美商再生元醫藥公司 Anti-activin A antibodies and uses thereof
EP3119432B1 (en) 2014-03-19 2020-01-08 MacKay Medical Foundation the Presbyterian Church in Taiwan MacKay Memorial Hospital Antibodies against immunogenic glycopeptides, composition comprising the same and use thereof
EP2960653A1 (en) * 2014-06-24 2015-12-30 Stichting Kwaliteitsgarantie Vleeskalversector Diagnostic kit and method for the identification of the manipulation of muscle mass in a domestic animal
CA3005158A1 (en) 2014-11-06 2016-05-12 Scholar Rock, Inc. Anti-pro/latent-myostatin antibodies and uses thereof
KR101860280B1 (en) 2014-12-19 2018-05-21 추가이 세이야쿠 가부시키가이샤 Anti-myostatin antibodies, polypeptides containing variant fc regions, and methods of use
KR20170110129A (en) 2015-02-05 2017-10-10 추가이 세이야쿠 가부시키가이샤 Antibodies comprising ionic concentration dependent antigen binding domains, Fc region variants, antibodies that bind to IL-8, and their use
EP3283519A1 (en) 2015-04-15 2018-02-21 Regeneron Pharmaceuticals, Inc. Methods of increasing strength and functionality with gdf8 inhibitors
KR20230155021A (en) 2015-09-15 2023-11-09 스칼러 락, 인크. Anti-pro/latent-myostatin antibodies and uses thereof
EP3394098A4 (en) 2015-12-25 2019-11-13 Chugai Seiyaku Kabushiki Kaisha Anti-myostatin antibodies and methods of use
WO2018025982A1 (en) 2016-08-05 2018-02-08 中外製薬株式会社 Composition for prophylaxis or treatment of il-8 related diseases
ES2944357T3 (en) 2017-01-06 2023-06-20 Scholar Rock Inc Treatment of metabolic diseases by inhibiting myostatin activation
EP3593144A1 (en) * 2017-03-10 2020-01-15 UCL Business Ltd Method relating to myostatin pathway inhibition
CN111787981A (en) 2018-03-01 2020-10-16 瑞泽恩制药公司 Method of altering body composition
US20220220213A1 (en) 2018-12-18 2022-07-14 Regeneron Pharmaceuticals, Inc. Compositions and Methods for Enhancing Body Weight and Lean Muscle Mass Using Antagonists Against Leptin Receptor, GDF8 and Activin A
WO2024064842A1 (en) 2022-09-21 2024-03-28 Regeneron Pharmaceuticals, Inc. Methods of treating obesity, diabetes, and liver dysfunction

Family Cites Families (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPH09507829A (en) * 1993-03-19 1997-08-12 ジョーンズ ホプキンス ユニバーシティー スクール オブ メディシン Growth differentiation factor-8
US7393682B1 (en) 1993-03-19 2008-07-01 The Johns Hopkins University School Of Medicine Polynucleotides encoding promyostatin polypeptides
US5994618A (en) * 1997-02-05 1999-11-30 Johns Hopkins University School Of Medicine Growth differentiation factor-8 transgenic mice
EP2272959A1 (en) * 1993-05-12 2011-01-12 Genetics Institute, LLC BMP-11 compositions
CA2194660C (en) * 1994-07-08 2009-09-29 Se-Jin Lee Growth differentiation factor-11
US6008434A (en) * 1994-07-08 1999-12-28 Johns Hopkins University School Of Medicine Growth differentiation factor-11 transgenic mice
WO1999006559A1 (en) 1997-08-01 1999-02-11 The Johns Hopkins University School Of Medicine Methods to identify growth differentiation factor (gdf) receptors
US6696260B1 (en) * 1997-08-01 2004-02-24 The Johns Hopkins University School Of Medicine Methods to identify growth differentiation factor (GDF) binding proteins
US6656475B1 (en) * 1997-08-01 2003-12-02 The Johns Hopkins University School Of Medicine Growth differentiation factor receptors, agonists and antagonists thereof, and methods of using same
AU1276399A (en) 1997-11-07 1999-05-31 Genetics Institute Inc. Neuronal uses of bmp-11
GB2333706A (en) 1998-02-02 1999-08-04 Merck & Co Inc Method for increasing muscle mass in animals
CA2319703C (en) 1998-02-05 2005-09-20 The Johns Hopkins University School Of Medicine Growth differentiation factor-8
AU765214B2 (en) * 1998-05-06 2003-09-11 Metamorphix International, Inc. Methods for treating diabetes by inhibiting gdf-8
US7504490B1 (en) * 1998-10-16 2009-03-17 Oscient Pharmaceuticals Corporation Nucleic acid and amino acid sequences relating to Apergillus fumigatus for diagnostics and therapeutics
WO2000043781A2 (en) 1999-01-21 2000-07-27 Metamorphix, Inc. Growth differentiation factor inhibitors and uses therefor
US7320789B2 (en) * 2001-09-26 2008-01-22 Wyeth Antibody inhibitors of GDF-8 and uses thereof
EP2192129A1 (en) * 2002-09-16 2010-06-02 Johns Hopkins University Metalloprotease activation of myostatin, and methods of modulating myostatin activity
US7261893B2 (en) * 2002-10-22 2007-08-28 Wyeth Neutralizing antibodies against GDF-8 and uses therefor
EP2272864A3 (en) * 2002-12-20 2011-02-16 Amgen Inc. Binding agents which inhibit myostatin
CA2526669A1 (en) * 2003-06-02 2004-12-16 Wyeth Use of myostatin (gdf8) inhibitors in conjunction with corticosteroids for treating neuromuscular disorders
NZ529860A (en) * 2003-11-28 2006-10-27 Ovita Ltd Muscle growth regulator mighty and use in promoting muscle mass and treating muscle wasting diseases
JP2007535912A (en) * 2003-12-31 2007-12-13 シェーリング−プラウ・リミテッド Neutralizing epitope-based growth-enhancing vaccine
MXPA06010929A (en) * 2004-03-23 2007-01-16 Lilly Co Eli Anti-myostatin antibodies.
US20060034831A1 (en) * 2004-08-12 2006-02-16 Wyeth Combination therapy for diabetes, obesity and cardiovascular diseases using GDF-8 inhibitors
NZ538097A (en) * 2005-02-07 2006-07-28 Ovita Ltd Method and compositions for improving wound healing
MX2007011400A (en) * 2005-03-23 2007-10-11 Wyeth Corp Detection of gdf-8 modulating agents.
EA015534B1 (en) * 2005-04-25 2011-08-30 Пфайзер Инк. Antibodies to myostatin and methods of use thereof
WO2006128190A2 (en) * 2005-05-27 2006-11-30 The Regents Of The University Of California Compositions and methods for treatment of neural disorders using transforming growth factor-beta superfamily proteins and their antagonists
DK1915397T3 (en) * 2005-08-19 2015-04-20 Wyeth Llc Antagonist AGAINST GDF-8 AND USES IN TREATMENT OF ALS AND OTHER GDF-8-ASSOCIATED DISEASES
ES2533464T3 (en) * 2005-10-06 2015-04-10 Eli Lilly And Company Anti-myostatin antibodies
UA92504C2 (en) * 2005-10-12 2010-11-10 Эли Лилли Энд Компани Anti-myostatin monoclonal antibody
MX2008007324A (en) * 2005-12-06 2009-03-04 Amgen Inc Uses of myostatin antagonists.
US20070190056A1 (en) * 2006-02-07 2007-08-16 Ravi Kambadur Muscle regeneration compositions and uses therefor
PL2066695T3 (en) 2006-09-05 2013-08-30 Lilly Co Eli Anti-myostatin antibodies
US7947646B2 (en) 2007-03-06 2011-05-24 Amgen Inc. Variant activin receptor polypeptides

Also Published As

Publication number Publication date
JP5052517B2 (en) 2012-10-17
EP1951756A2 (en) 2008-08-06
KR101135220B1 (en) 2012-04-24
ES2533464T3 (en) 2015-04-10
US20080299126A1 (en) 2008-12-04
KR20080045733A (en) 2008-05-23
NO20082073L (en) 2008-06-17
US7745583B2 (en) 2010-06-29
US20100087631A1 (en) 2010-04-08
JP2009511480A (en) 2009-03-19
IL189579A0 (en) 2008-08-07
WO2007044411A3 (en) 2007-08-16
WO2007044411A2 (en) 2007-04-19
AU2006302494B2 (en) 2011-01-06
CN101277976A (en) 2008-10-01
CN101277976B (en) 2012-04-11
EP1951756B1 (en) 2015-01-07
EA015589B1 (en) 2011-10-31
US7635760B2 (en) 2009-12-22
AU2006302494A1 (en) 2007-04-19
BRPI0616923A2 (en) 2011-07-05
EA200801027A1 (en) 2008-10-30

Similar Documents

Publication Publication Date Title
AU2006302494B2 (en) Anti-myostatin antibodies
US8066995B2 (en) Anti-myostatin antibodies
JP5058261B2 (en) Anti-myostatin antibody
JP5063612B2 (en) Anti-IL-17 antibody
JP2009519348A5 (en)
BRPI0619792B1 (en) HUMANIZED ANTI-IL-17 MONOCLONAL ANTIBODY, COMPOSITION AND USE OF SUCH ANTIBODY

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued

Effective date: 20140325