WO2012138575A1 - Compositions and methods for increasing muscle function and mass - Google Patents

Compositions and methods for increasing muscle function and mass Download PDF

Info

Publication number
WO2012138575A1
WO2012138575A1 PCT/US2012/031630 US2012031630W WO2012138575A1 WO 2012138575 A1 WO2012138575 A1 WO 2012138575A1 US 2012031630 W US2012031630 W US 2012031630W WO 2012138575 A1 WO2012138575 A1 WO 2012138575A1
Authority
WO
WIPO (PCT)
Prior art keywords
gremlin
antibody
subject
muscle
protein
Prior art date
Application number
PCT/US2012/031630
Other languages
French (fr)
Inventor
Maria DEATO
Jun Zhang
Hui Tian
Original Assignee
Ngm Biopharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ngm Biopharmaceuticals, Inc. filed Critical Ngm Biopharmaceuticals, Inc.
Publication of WO2012138575A1 publication Critical patent/WO2012138575A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1136Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against growth factors, growth regulators, cytokines, lymphokines or hormones
    • 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
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed
    • C12N2310/531Stem-loop; Hairpin

Definitions

  • Muscle wasting is associated with a number of diseases and conditions. Currently there are few effective treatments for such disorders.
  • the present disclosure provides compositions and methods for reducing levels and/or activity of Gremlin-2.
  • the present disclosure provides compositions and methods for increasing muscle function and/or muscle mass.
  • the present methods involve using an agent that targets Gremlin-2 polypeptide.
  • Subject compositions and methods are useful for treating various conditions and disorders characterized by loss of muscle function and/or muscle mass.
  • AAV adeno-associated virus
  • Figure 3 shows body weight, lean mass, and fat mass of mice that were injected with AAV expressing a protein of the present disclosure (mouse ortholog) at day 1-3 by
  • TA Tibialis Anterior
  • Figure 6 shows different tissue weights of 14 week old mice that were injected with AAV expressing a protein of the present disclosure (mouse ortholog) at day 1-3 by
  • Figure 7 shows serum phosphate, calcium, iron, cholesterol, low-density lipoprotein
  • LDL low-density lipoprotein
  • TRIGL fatty acid, triglyceride
  • ALT alanine transaminase
  • AST aspartate transaminase
  • GLU glucose
  • Figure 8 shows levels of message RNA of the present disclosure (mouse ortholog) in
  • Figure 9 shows levels of a protein of the present disclosure (mouse ortholog) in Tibialis Anterior muscle and serum in a 4 week old mouse model for human Duchenne Muscular Dystrophy (DMD).
  • Figure 10 shows levels of the message RNA of present disclosure (mouse ortholog) at day 7 post-cardiotoxin injection in Tibialis Anterior muscle of 13 week old mice.
  • Figure 11 shows levels of a protein of the present disclosure (mouse ortholog) at day 7 post-cardiotoxin injection in Tibialis Anterior muscle and serum of 13 week old mice.
  • AAV adeno-associated virus
  • Figure 14 shows body weight, lean mass and fat mass of 14 week old mice that were
  • mice ortholog a nucleic acid encoding a protein of the present disclosure
  • Figure 15 provides an alignment of Gremlin-2 amino acid sequences.
  • patient or “subject” as used interchangeably herein in the context of therapy, refer to a human and non-human animal, as the recipient of a therapy or preventive care.
  • indicators include, but are not limited to, Gremlin-2 circulating levels, Gremlin-2 tissue levels, Gremlin-2 activity, muscle function, and muscle mass.
  • Gremlin-2 also known as “cysteine knot superfamily 1 bone morphogenic protein
  • Neuroblastoma- 1 domain family member 3 (DAND3); "protein related to DAN and Cerberus” (PRDC); FLJ21195; and DCR5) encompasses murine and human proteins that are encoded by a GREM2 gene or a gene homologue of GREM2.
  • Gremlin-2 is found in many mammals (e.g. human, non-human primates, and mouse). See Figure 15 for an alignment of amino acid sequences of various Gremlin-2 polypeptides.
  • homologues or “variants” refers to protein or DNA sequences that are similar based on their amino acid or nucleic acid sequences, respectively. Homologues or variants encompass naturally occurring DNA sequences and proteins encoded thereby and their isoforms. The homologues also include known allelic or splice variants of a protein/gene. Homologues and variants also encompass nucleic acid sequences that vary in one or more bases from a naturally-occurring DNA sequence but still translate into an amino acid sequence that correspond to the naturally-occurring protein due to degeneracy of the genetic code. Homologues and variants may also refer to those that differ from the naturally-occurring sequences by one or more conservative substitutions and/or tags and/or conjugates.
  • polypeptide refers to a polymeric form of amino acids of any length, which can include genetically coded and non-genetically coded amino acids, chemically or biochemically modified or derivatized amino acids, and polypeptides having modified peptide backbones.
  • the term includes fusion proteins, including, but not limited to, fusion proteins with a heterologous amino acid sequence, fusions with heterologous and homologous leader sequences, with or without N-terminal methionine residues; immunologically tagged proteins; and the like.
  • nucleic acid molecule and “polynucleotide” are used interchangeably and refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof.
  • Non-limiting examples of polynucleotides include linear and circular nucleic acids, messenger RNA (mRNA), cDNA, recombinant polynucleotides, vectors, probes, and primers.
  • heterologous refers to two components that are defined by structures derived from different sources.
  • heterologous is used in the context of a polypeptide, where the polypeptide includes operably linked amino acid sequences that can be derived from different polypeptides (e.g., a first component consisting of a recombinant peptide and a second component derived from a native Gremlin-2 polypeptide).
  • heterologous in the context of a polynucleotide encoding a chimeric polypeptide includes operably linked nucleic acid sequence that can be derived from different genes (e.g., a first component from a nucleic acid encoding a peptide according to an
  • heterologous nucleic acids include expression constructs in which a nucleic acid comprising a coding sequence is operably linked to a regulatory element (e.g., a promoter) that is from a genetic origin different from that of the coding sequence (e.g., to provide for expression in a host cell of interest, which may be of different genetic origin relative to the promoter, the coding sequence or both).
  • a T7 promoter operably linked to a polynucleotide encoding a Gremlin-2 polypeptide or domain thereof is said to be a heterologous nucleic acid.
  • “Heterologous” in the context of recombinant cells can refer to the presence of a nucleic acid (or gene product, such as a polypeptide) that is of a different genetic origin than the host cell in which it is present.
  • operably linked refers to functional linkage between molecules to provide a desired function.
  • operably linked in the context of nucleic acids refers to a functional linkage between nucleic acids to provide a desired function such as
  • a functional linkage between a nucleic acid expression control sequence such as a promoter, signal sequence, or array of transcription factor binding sites
  • a second polynucleotide wherein the expression control sequence affects transcription and/or translation of the second polynucleotide.
  • "Operably linked" in the context of a polypeptide refers to a functional linkage between amino acid sequences (e.g., of different domains) to provide for a described activity of the polypeptide.
  • N-terminus and C- terminus refer to the extreme amino and carboxyl ends of the polypeptide, respectively, while “N-terminal” and “C-terminal” refer to relative positions in the amino acid sequence of the polypeptide toward the N-terminus and the C-terminus, respectively, and can include the residues at the N-terminus and C-terminus, respectively.
  • "Immediately N- terminal” or “immediately C-terminal” refers to a position of a first amino acid residue relative to a second amino acid residue where the first and second amino acid residues are covalently bound to provide a contiguous amino acid sequence.
  • "Derived from” in the context of an amino acid sequence or polynucleotide sequence is meant to indicate that the polypeptide or nucleic acid has a sequence that is based on that of a reference polypeptide or nucleic acid (e.g., a naturally occurring Gremlin-2 polypeptide or Gremlin- 2-encoding nucleic acid), and is not meant to be limiting as to the source or method in which the protein or nucleic acid is made.
  • Isolated refers to a protein of interest (e.g., a Gremlin-2 polypeptide; an anti-Gremlin-2 antibody) that, if naturally occurring, is in an environment different from that in which it may naturally occur. "Isolated” is meant to include proteins that are within samples that are substantially enriched for the protein of interest and/or in which the protein of interest is partially or substantially purified. Where the protein is not naturally occurring, “isolated” indicates the protein has been separated from an environment in which it was made by either synthetic or recombinant means.
  • a protein of interest e.g., a Gremlin-2 polypeptide; an anti-Gremlin-2 antibody
  • Enriched means that a sample is non-naturally manipulated (e.g., by an experimentalist or a clinician) so that a protein of interest (e.g., a Gremlin-2 polypeptide; an anti-Gremlin-2 antibody) is present in a greater concentration (e.g., at least a three-fold greater, at least 4- fold greater, at least 8-fold greater, at least 64-fold greater, or more) than the concentration of the protein in the starting sample, such as a biological sample (e.g., a sample in which the protein naturally occurs or in which it is present after administration), or in which the protein was made (e.g., as in a bacterial protein and the like).
  • a protein of interest e.g., a Gremlin-2 polypeptide; an anti-Gremlin-2 antibody
  • a greater concentration e.g., at least a three-fold greater, at least 4- fold greater, at least 8-fold greater, at least 64-fold greater, or more
  • substantially pure indicates that an entity (e.g., a Gremlin-2 polypeptide; an anti- Gremlin-2 antibody) makes up greater than about 50% of the total content of the composition (e.g., total protein of the composition), or greater than about 60% of the total protein content.
  • a “substantially pure” refers to compositions in which at least 75%, at least 85%, at least 90% or more of the total composition is the entity of interest (e.g. 95%, 98%, 99%, greater than 99%), of the total protein.
  • the protein can make up greater than about 90%, or greater than about 95% of the total protein in the
  • antibody encompasses polyclonal and monoclonal antibody preparations where the antibody may be of any class of interest (e.g., IgM, IgG, and subclasses thereof), as well as preparations including hybrid antibodies, altered antibodies, F(ab') 2 fragments, F(ab) molecules, Fv fragments, single chain Fv fragments (scFv), single chain antibodies, single domain antibodies (VRH), chimeric antibodies, humanized antibodies, and antigen-binding fragments thereof which exhibit immunological binding properties of the parent antibody molecule.
  • Antibodies that have inhibitory functions for their targets are of particular interest.
  • the antibodies described herein may be detectably labeled, e.g., with a radioisotope, an enzyme which generates a detectable product, a fluorescent protein, and the like.
  • the antibodies may be further conjugated to other moieties, such as members of specific binding pairs, e.g., biotin (member of biotin-avidin specific binding pair), and the like.
  • the antibodies may also be bound to a support (e.g., a solid support), such as a polystyrene plate or bead, test strip, and the like.
  • binding refers to a direct association between two molecules, due to, for
  • a subject anti- Gremlin-2 antibody binds specifically to an epitope within a Gremlin-2 polypeptide, e.g., with an affinity of at least about 10 "7 M, at least about 10 ⁇ 8 M, at least about 10 "9 M, at least about 10 "10 M, at least about 10 "11 M, or at least about 10 "12 M, or greater than 10 "12 M.
  • a subject antibody binds to an epitope present on a Gremlin-2 polypeptide with an affinity of from about 10 "7 M to about 10 ⁇ 8 M, from about 10 "8 M to about 10 "9 M, from about 10 "9 M to about 10 "10 M, from about 10 "10 M to about 10 "11 M, or from about 10 "11 M to about 10 "12
  • Non-specific binding would refer to binding with an affinity of less than about 10 "7 M, e.g., binding with an affinity of 10 "6 M, 10 "5 M, 10 "4 M, etc.
  • Immunoglobulin polypeptides include the kappa and lambda light chains and the alpha, gamma (IgG 1 ; IgG 2 , IgG 3 , IgG 4 ), delta, epsilon and mu heavy chains or equivalents in other species.
  • Full-length immunoglobulin "light chains” comprise a variable region at the NH 2 -terminus and a kappa or lambda constant region at the COOH-terminus.
  • Full-length immunoglobulin "heavy chains” similarly comprise a variable region and one of the aforementioned heavy chain constant regions, e.g., gamma.
  • An immunoglobulin light or heavy chain variable region is composed of a "framework" region (FR) interrupted by three hypervariable regions, also called “complementarity determining regions” or "CDRs". CDRs have been described by Kabat et al., J. Biol.
  • the framework region of an antibody that is the combined framework regions of the constituent light and heavy chains, serves to position and align the CDRs.
  • the CDRs are primarily responsible for binding to an epitope of an antigen.
  • the term "monoclonal antibody” refers to an antibody composition having a homogeneous antibody population. The term is not limited by the manner in which it is made. The term encompasses whole immunoglobulin molecules, as well as Fab molecules, F(ab')2 fragments, Fv fragments, scFv, fusion proteins comprising an antigen-binding portion of an antibody and a non-antibody protein, and other molecules that exhibit immunological binding properties of the parent monoclonal antibody molecule. Methods of making polyclonal and monoclonal antibodies are known in the art.
  • Antibody fragments comprise a portion of an intact antibody, for example, the antigen binding or variable region of the intact antibody.
  • antibody fragments include Fab, Fab', F(ab') 2 , and Fv fragments; diabodies; linear antibodies (Zapata et al., Protein Eng. 8(10): 1057-1062 (1995)); single-chain antibody molecules; and multispecific antibodies formed from antibody fragments.
  • Papain digestion of antibodies produces two identical antigen-binding fragments, called "Fab” fragments, each with a single antigen- binding site, and a residual "Fc” fragment, a designation reflecting the ability to crystallize readily.
  • Pepsin treatment yields an F(ab') 2 fragment that has two antigen combining sites and is still capable of cross-linking antigen.
  • Fv is the minimum antibody fragment which contains a complete antigen-recognition and -binding site. This region consists of a dimer of one heavy- and one light-chain variable domain in tight, non-covalent association. It is in this configuration that the three CDRS of each variable domain interact to define an antigen-binding site on the surface of the V R -V L dimer. Collectively, the six CDRs confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
  • the "Fab" fragment also contains the constant domain of the light chain and the first
  • constant domain CH of the heavy chain.
  • Fab fragments differ from Fab' fragments by the addition of a few residues at the carboxyl terminus of the heavy chain CHi domain including one or more cysteines from the antibody hinge region.
  • Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear a free thiol group.
  • F(ab') 2 antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
  • Single-chain Fv or “sFv” antibody fragments comprise the V H and V L domains of
  • the Fv polypeptide further comprises a polypeptide linker between the V H and V L domains, which enables the sFv to form the desired structure for antigen binding.
  • diabodies refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (V H ) connected to a light-chain variable domain (V L ) in the same polypeptide chain (V R -V L ).
  • V H heavy-chain variable domain
  • V L light-chain variable domain
  • V R -V L polypeptide chain
  • the present disclosure provides compositions and methods for reducing levels and/or activity of Gremlin-2.
  • the present disclosure provides compositions and methods for increasing muscle function and/or muscle mass.
  • the present methods involve using an agent that targets a Gremlin-2 polypeptide.
  • Subject compositions and methods are useful for treating various conditions and disorders characterized by loss of muscle function and/or muscle mass.
  • Gremlin-2 encompass Gremlin-2, Gremlin-2 genes and/or proteins encoded thereby, and are useful for treating individuals having a deficiency in muscle function and/or having reduced muscle mass, e.g., for treating disorders, diseases, and conditions in which reduced muscle function and/or mass is a result, a sequela, or a symptom of the disorder, disease, or condition.
  • Gremlin-2 protein was administered (as exemplified by expression from an AAV) to wild-type mice, loss of muscle mass and reduced grip strength were observed.
  • mice showed elevated muscle levels of Gremlin-2 mRNA, and elevated muscle and serum levels of Gremlin-2 protein.
  • targeting a Gremlin-2 protein to reduce circulating and/or tissue levels of Gremlin-2 and/or to reduce Gremlin-2 activity, can be used to increase muscle function and or muscle mass in an individual.
  • Targeting a Gremlin-2 protein can be used to treat disorders, diseases, and conditions in which reduced muscle function (e.g., muscle weakness) and/or reduced muscle mass is a result, a sequela, or a symptom of the disorder, disease, or condition.
  • the present disclosure provides methods of reducing levels and/or activity of Gremlin-2 in an individual.
  • the present disclosure also provides methods for increasing muscle function and/or muscle mass in an individual having a deficiency in muscle function and/or having reduced muscle mass, e.g., in an individual having a condition, disease, or disorder in which reduced muscle function and/or reduced muscle mass is a result, a sequela, or a symptom of the disorder, disease, or condition.
  • a subject method generally involves administering to an individual an effective amount of an agent that reduces Gremlin-2 levels and/or activity.
  • Suitable agents include, e.g., an antibody that specifically binds Gremlin-2; and a short interfering nucleic acid (siNA) that specifically reduces the level of Gremlin-2.
  • Agents that reduce levels and/or activity of Gremlin-2 include agents that reduce Gremlin- 2 circulating and/or tissue levels of Gremlin-2; and agents that reduce one or more biological activities of Gremlin-2. Circulating levels of Gremlin-2 include levels of Gremlin-2 in the blood or any extracellular fluid.
  • Agents that reduce circulating and/or tissue levels of Gremlin-2 polypeptide, and that are suitable for use in a subject method includes agents that, when administered to an individual, reduce circulating levels of Gremlin-2 polypeptide in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or more than 70%, compared to the circulating level of Gremlin-2 polypeptide in the individual not treated with the agent.
  • a suitable agent is one that, when administered to an individual, reduces circulating levels of Gremlin-2 polypeptide in the individual to a normal control level.
  • Circulating levels of Gremlin-2 include serum levels. Circulating levels of Gremlin-2 polypeptide can be readily determined, using any known method, e.g., an immunological method employing anti-Gremlin-2 antibody. Suitable immunological methods include, e.g., an enzyme-linked immunosorbent assay (ELISA), a
  • RIA radioimmunoassay
  • Agents that reduce circulating and/or tissue levels of Gremlin-2 polypeptide, and that is suitable for use in a subject method includes agents that, when administered to an individual, reduce tissue levels of Gremlin-2 polypeptide in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or more than 70%, compared to the tissue level of Gremlin-2 polypeptide in the individual not treated with the agent.
  • an agent suitable for use in a subject method includes an agent that reduces muscle levels (e.g., skeletal muscle levels) of Gremlin-2 polypeptide in an individual.
  • a suitable agent is one that, when administered to an individual, reduces tissue levels of Gremlin-2 polypeptide in the individual to a normal control level.
  • Agents that reduce one or more Gremlin-2 activities in an individual include, e.g., agents that reduce (e.g., inhibit) binding of Gremlin-2 to a Gremlin-2 binding partner, e.g., a Gremlin-2 binding partner present in a muscle cell, a Gremlin-2 binding partner present in a cell membrane, or an extracellular Gremlin-2 binding partner.
  • agents that reduce (e.g., inhibit) binding of Gremlin-2 to a Gremlin-2 binding partner e.g., a Gremlin-2 binding partner present in a muscle cell, a Gremlin-2 binding partner present in a cell membrane, or an extracellular Gremlin-2 binding partner.
  • an agent suitable for use in a subject method can include an agent that inhibits binding of a
  • Gremlin-2 polypeptide to a Gremlin-2 binding partner by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or more than 70%, compared to the binding of the Gremlin-2 polypeptide to the Gremlin-2 binding partner in the absence of the agent.
  • Increasing muscle mass includes: a) an increase in muscle mass that results from generation of new muscle tissue; and b) an increase in muscle mass that results from repair of existing muscle tissue that has been damaged (e.g., due to disease or injury).
  • a subject method involves administering an anti-Gremlin-2 antibody to a subject who has a disease, disorder, or condition resulting in or associated with reduced muscle function and/or reduced muscle mass (e.g., a disease, disorder, or condition in which reduced muscle function and/or reduced muscle mass is a result, a sequela, or a symptom of the disorder, disease, or condition).
  • the methods of the present disclosure can comprise administering one or more anti-Gremlin-2 antibodies in the context of a variety of diseases, disorders, and conditions resulting in or associated with reduced muscle function and/or reduced muscle mass.
  • a subject method involves administering a short interfering nucleic acid (siNA) specific for Gremlin-2 to a subject who has a disease, disorder, or condition resulting in or associated with reduced muscle function and/or muscle mass (e.g., a disease, disorder, or condition in which reduced muscle function and/or reduced muscle mass is a result, a sequela, or a symptom of the disorder, disease, or condition).
  • a disease, disorder, or condition in which reduced muscle function and/or reduced muscle mass is a result, a sequela, or a symptom of the disorder, disease, or condition.
  • the methods of the present disclosure can include administering one or more Gremlin-2-specific siNA in the context of a variety of diseases, disorders, and conditions resulting in or associated with reduced muscle function and/or muscle mass.
  • condition resulting in or associated with reduced muscle function and/or muscle mass are contemplated for therapy and diagnosis described herein.
  • treatment is meant that at least an amelioration of the symptoms associated with the condition afflicting the host is achieved, where amelioration refers to at least a reduction in the magnitude of a parameter, e.g. symptom, associated with the condition being treated.
  • amelioration refers to at least a reduction in the magnitude of a parameter, e.g. symptom, associated with the condition being treated.
  • treatment includes situations where the condition, or at least symptoms associated therewith, are reduced or avoided.
  • treatment includes: (i) prevention, that is, reducing the risk of development of clinical symptoms, including causing the clinical symptoms not to develop, e.g., preventing disease progression to a harmful or otherwise undesired state; (ii) inhibition, that is, arresting the development or further development of clinical symptoms, e.g., mitigating or completely inhibiting an active disease (e.g., so as to increase muscle function and/or muscle mass).
  • prevention that is, reducing the risk of development of clinical symptoms, including causing the clinical symptoms not to develop, e.g., preventing disease progression to a harmful or otherwise undesired state
  • inhibition that is, arresting the development or further development of clinical symptoms, e.g., mitigating or completely inhibiting an active disease (e.g., so as to increase muscle function and/or muscle mass).
  • antibody compositions or siNA compositions described herein can be administered to a subject (e.g. a human patient) to, for example, increase muscle function to a range found in a healthy individual.
  • Subjects for treatment include those having a disease, disorder, or condition resulting in or associated with reduced muscle function and/or mass, as described herein.
  • an effective amount of an anti-Gremlin-2 antibody or a Gremlins- specific siNA is an amount that is effective to reduce muscle atrophy, e.g., an effective amount of an anti-Gremlin-2 antibody or a Gremlin-2-specific siNA reduces muscle atrophy by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or at least about 80%, or more than 80%, compared to the degree of atrophy in the absence of treatment with the anti-Gremlin-2 antibody or the Gremlin-2- specific siNA.
  • an effective amount of an anti-Gremlin-2 antibody or a Gremlins- specific siNA is an amount that is effective to increase muscle mass (e.g., skeletal muscle mass), e.g., an effective amount of an anti-Gremlin-2 antibody or a Gremlin-2- specific siNA increases muscle mass by at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, or at least about 5-fold, or more than 5-fold, compared to the muscle mass in the absence of treatment with the anti- Gremlin-2 antibody or the Gremlin-2- specific siNA.
  • increasing muscle mass includes: a) an increase in muscle mass that results from generation of new muscle tissue; and b) an increase in muscle mass that results from repair of existing muscle tissue that has been damaged (e.g., due to disease or injury).
  • Whether atrophy is reduced, and whether muscle mass is increased can be determined using any known method, including, e.g., magnetic resonance imaging (MRI), dual energy x-ray absorptiometry (DEXA), and computed tomography (CT).
  • MRI magnetic resonance imaging
  • DEXA dual energy x-ray absorptiometry
  • CT computed tomography
  • a method of the present disclosure can provide for improved muscle
  • muscle function includes, e.g., muscle endurance, muscle strength, muscle force, muscle fatigability, etc.
  • "Improved" muscle function includes increased muscle endurance, increased muscle strength, increased muscle force, and decreased muscle fatigability.
  • treatment with an agent that reduces Gremlin-2 levels and/or activity results in an increase in one or more of muscle endurance, muscle strength, and muscle force of at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, or at least about 5- fold, or more than 5-fold, compared to the muscle endurance, muscle strength, or muscle force in the absence of treatment with the agent.
  • treatment with an agent that reduces Gremlin-2 levels and/or activity results in a decrease in muscle fatigability, e.g., results in an increase in the amount of time to reach a fatigued state, such that muscle fatigability is reduced by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or more than 80%, compared to the muscle fatigability in the absence of treatment with the agent.
  • an effective amount of an anti-Gremlin-2 antibody is an amount that is effective to improve function (e.g., increase one or more of muscle endurance, muscle strength, muscle force, etc.; and/or decrease muscle fatigability, etc.).
  • an effective amount of an anti-Gremlin-2 antibody improves muscle function by at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 2- fold, at least about 2.5-fold, or at least about 5-fold, or more than 5-fold, compared to the muscle function in the absence of treatment with the anti-Gremlin-2 antibody.
  • an effective amount of a Gremlin-2-specific siNA improves muscle function by at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, or at least about 5-fold, or more than 5-fold, compared to the muscle function in the absence of treatment with the Gremlin- specific siNA.
  • Muscle strength can be measured using any known method, including, e.g., a grip strength test. See, e.g., Geere et al. ((2007) BMC Musculoskelet. Disord. 8: 114), and references cited therein.
  • a field test such as the one -repetition maximum (1-RM) test, can also be used.
  • the 1-RM test measures dynamic strength by determining how much weight an individual can lift during a single repetition. The amount can be divided by body weight to give 1-RM/BW.
  • Muscle strength and function can be assessed by standard performance tests such as knee flexor and extensor strength, repeated sit-to-stand test, and timed up & go (TUG).
  • Muscle strength can be measured as knee extensor and flexor in Newtons (kiloponds).
  • TUG is a measure of functional mobility including muscle strength, gait speed, and balance and is assessed in seconds.
  • the repeated sit-to-stand is a functional test and measured in seconds.
  • Muscle force expressed as tetanic force, can be measured using any known method.
  • Various types of contractions can be measured, including isotonic contraction, concentric contraction, eccentric contraction, and isometric contraction.
  • Methods of measuring muscle contraction are known in the art, and any such method can be used to measure muscle contraction. Suitable methods include, e.g., mechanomyography, ultrasound myography, acoustic myography, electromyography, and the like.
  • Muscle fatigability and muscle endurance can be measured in humans using a treadmill test, e.g., where the treadmill is inclined or is horizontal. Muscle fatigability and muscle endurance can be measured in rodents (e.g., mice, rats, etc.) using a rotarod test or a wire hang test. For example, in the rotarod test, mice (or rats) are placed on an elevated accelerating rod and the rod is rotated at a certain speed (e.g., 4 rotations per minute (rpm) to 40 rpm). The rodents are then scored for their latency (e.g., in seconds) to fall. A reduction in the time to fall is an indication of an increase in muscle endurance or a reduction in muscle fatigability.
  • rodents e.g., mice, rats, etc.
  • a rotarod test mice (or rats) are placed on an elevated accelerating rod and the rod is rotated at a certain speed (e.g., 4 rotations per minute (rpm) to 40 r
  • Methods and compositions are provided herein to reduce levels and/or activity of Gremlin- 2 and/or to increase muscle function and/or muscle mass in a subject.
  • the subject methods and/or compositions target Gremlin-2 so as to decrease the amount and/or the activity of the mature Gremlin-2 protein.
  • the present disclosure provides methods for reducing (e.g., inhibiting, or neutralizing) Gremlin-2 activity in a subject having a disease, disorder, or condition that is associated with or that results in reduced muscle function and/or mass.
  • One way Gremlin-2 can be inhibited is to administer anti-Gremlin-2 antibodies (also referred to as "anti-Gremlin-2 antibodies"), where such antibodies include a whole antibody (e.g. IgG), an antigen- binding fragment thereof, single-chain Fab, or a synthetic anti-Gremlin-2 antibody that comprise portions of an antibody.
  • Gremlin-2 is the target of such antibodies. Details on Gremlin-2 are provided below.
  • Gremlin-2 may exacerbate symptoms of a disease, disorder, or condition associated with reduced muscle function and/or mass. Accordingly, decreasing the amount and/or activity of Gremlin-2 can serve to treat such diseases, disorders, and conditions.
  • Targeting Gremlin-2 as provided by the present methods can encompass inhibiting other biomolecules ("binding partners") that normally interact with Gremlin-2 from binding to Gremlin-2 or sequestering Gremlin-2 from its signaling partners.
  • binding partners biomolecules
  • a subject anti-Gremlin-2 antibody can decrease Gremlin-2 activity.
  • a subject anti-Gremlin-2 antibody can decrease the ability of Gremlin-2 to bind to a binding partner for Gremlin-2, e.g., a naturally- occurring cellular binding partner for Gremlin-2.
  • the binding of a subject anti-Gremlin-2 antibody may be competitive or non-competitive with the endogenous binding partners of Gremlin-2.
  • a subject anti-Gremlin-2 antibody may also modify the activity and/or structure of Gremlin-2.
  • the methods can further encompass decreasing the level of Gremlin-2 expression and/or amount of mature, active Gremlin-2.
  • the method can also increase turnover of Gremlin-2.
  • a subject anti-Gremlin-2 antibody can reduce binding of a Gremlin-2
  • a subject antibody can reduce binding of a Gremlin-2 polypeptide to a Gremlin-2 binding partner by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more, compared to the degree of binding between the Gremlin-2 polypeptide and the binding partner in the absence of the antibody.
  • an antibody comprising: a) a variable domain comprising: i. a
  • CDR1 region that is identical in amino acid sequence to the heavy chain CDR1 region of an anti-Gremlin-2 antibody
  • a CDR2 region that is identical in amino acid sequence to the heavy chain CDR2 region of the anti-Gremlin-2 antibody
  • a CDR3 region that is identical in amino acid sequence to the heavy chain CDR3 region of the anti-Gremlin-2 antibody
  • a light chain variable domain comprising: i. a CDR1 region that is identical in amino acid sequence to the light chain CDR1 region of the anti-Gremlin-2 antibody; ii.
  • a CDR2 region that is identical in amino acid sequence to the light chain CDR2 region of the anti-Gremlin-2 antibody; and iii. a CDR3 region that is identical in amino acid sequence to the light chain CDR3 region of the anti-Gremlin-2 antibody; or b) a variant of the variable domain of part a) that is otherwise identical to the variable domain of part a) except for a number of (e.g., 1, 2, 3, 4, 5, 6, 7 or 8) amino acid substitutions in the CDR regions, where the antibody binds a Gremlin-2 polypeptide.
  • a subject antibody comprises: a) a light chain region comprising: i) one, two, or three complementarity determining regions (CDRs) from a mouse monoclonal anti-Gremlin-2 antibody light chain variable region sequence; and ii) a light chain framework region, e.g., a framework region from a human immunoglobulin light chain; and b) a heavy chain region comprising: i) one, two, or three CDRs from the mouse monoclonal anti-Gremlin-2 antibody heavy chain variable region sequence; and ii) a heavy chain framework region, e.g., a framework region from a human immunoglobulin heavy chain.
  • CDRs complementarity determining regions
  • a subject anti-Gremlin-2 antibody can find use in a variety of applications, including use in various methods of treating a host suffering from a disease, disorder, or condition associated with reduced muscle function and/or mass, as well as in diagnosis of various diseases and conditions associated with Gremlin-2 expression.
  • a subject anti-Gremlin-2 antibody may be recombinant.
  • the antibody may contain a light and/or heavy chain.
  • Methods for producing recombinant antibodies are known in the art.
  • the nucleic acids encoding the antibody, or at least a complementary determining region (CDR) of a heavy chain polypeptide or at least a CDR of a light chain polypeptide are introduced directly into a host cell, and the cell incubated under conditions sufficient to induce expression of the encoded antibody.
  • the recombinant antibody may be glycosylated by an endogenous glycosylase in the host cells, unglycosylated, or may have an altered glycosylation pattern.
  • the antibody may be chimeric.
  • Chimeric antibodies are immunoglobulin molecules comprising human and non-human portions. More specifically, the antigen combining region (or variable region) of a humanized chimeric antibody is derived from a non-human source (e.g. murine), and the constant region of the chimeric antibody (which confers biological effector function to the immunoglobulin) is derived from a human source.
  • the chimeric antibody can have the antigen binding specificity of the non-human antibody molecule and the effector function conferred by the human antibody molecule.
  • a large number of methods of generating chimeric antibodies are well known to those of skill in the art.
  • An alternative approach is the generation of humanized antibodies by linking the CDR regions of non-human antibodies to human constant regions by recombinant DNA techniques.
  • a recombinant fusion antibody that is specific for a Gremlin-2 polypeptide is
  • the antibody is modified to include a heterologous protein.
  • a heavy chain polypeptide and/or light chain polypeptide may be joined to a reporter protein or to a protein having a desired therapeutic effect.
  • the reporter protein may be a fluorescent protein.
  • the antibody may also be conjugated to a second antibody (or at least an antigen-binding portion thereof).
  • a subject anti-Gremlin-2 antibody will in some embodiments be humanized.
  • Amino acids may be substituted in the framework regions of a parent non-human (e.g., mouse monoclonal) antibody to produce a modified antibody that is less immunogenic in a human than the parent non-human antibody.
  • Antibodies can be humanized using a variety of techniques known in the art. Framework substitutions are identified by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions.
  • the antibody may also be a fully human antibody. Human antibodies are primarily
  • a subject human antibody can be produced by a wide variety of methods.
  • human antibodies can be produced initially in trioma cells (descended from three cells, two human and one mouse). Genes encoding the antibodies are then cloned and expressed in other cells, particularly non-human mammalian cells.
  • trioma cells descend from three cells, two human and one mouse.
  • Genes encoding the antibodies are then cloned and expressed in other cells, particularly non-human mammalian cells.
  • the general approach for producing human antibodies by trioma technology has been described in the art.
  • the present disclosure contemplates a DNA molecule comprising a nucleic acid sequence encoding an antibody that binds to a Gremlin-2 polypeptide.
  • the disclosure further contemplates recombinant host cells containing an exogenous polynucleotide encoding at least a CDR of a heavy chain polypeptide or at least a CDR of a light chain polypeptide of the subject antibody.
  • a subject antibody comprises anti-Gremlin-2 antibody heavy chain CDRs and anti-Gremlin-2 antibody light chain CDRs in a single polypeptide chain, e.g., in some embodiments, a subject antibody is a scFv.
  • a subject antibody comprises, in order from N-terminus to C-terminus: a first amino acid sequence of from about 5 amino acids to about 25 amino acids in length; a heavy chain CDR1 of an anti- Gremlin-2 antibody; a second amino acid sequence of from about 5 amino acids to about 25 amino acids in length; a heavy chain CDR2 of an anti-Gremlin-2 antibody; a third amino acid sequence of from about 5 amino acids to about 25 amino acids in length; a heavy chain CDR3 of an anti-Gremlin-2 antibody; a fourth amino acid sequence of from about 5 amino acids to about 25 amino acids in length; a light chain CDR1 of an anti- Gremlin-2 antibody; a fifth amino acid sequence of from about 5 amino acids to about 25 amino acids in length; a light chain CDR2 an anti-Gremlin-2 antibody; a sixth amino acid sequence of from about 5 amino acids to about 25 amino acids in length; a light chain CDR3 an anti-Gremlin-2 antibody; and
  • a subject anti-Gremlin-2 antibody comprises scFv multimers.
  • a subject antibody is an scFv dimer (e.g., comprises two tandem scFv (scFv 2 )), an scFv trimer (e.g., comprises three tandem scFv (scFv 3 )), an scFv tetramer (e.g., comprises four tandem scFv (scFv 4 )), or is a multimer of more than four scFv (e.g., in tandem).
  • the scFv monomers can be linked in tandem via linkers of from about 2 amino acids to about 10 amino acids in length, e.g., 2 aa, 3 aa, 4 aa, 5 aa, 6 aa, 7 aa, 8 aa, 9 aa, or 10 aa in length.
  • Suitable linkers include, e.g., (Gly) x , where x is an integer from 2 to 10. Other suitable linkers are those discussed above.
  • each of the scFv monomers in a subject scFV multimer is humanized, as described above.
  • a subject anti-Gremlin-2 antibody can comprises one or more modifications.
  • a subject antibody comprises a free thiol (-SH) group at the
  • the free thiol group can be used to attach the antibody to a second polypeptide (e.g., another antibody, including a subject antibody), a scaffold, a carrier, etc.
  • a second polypeptide e.g., another antibody, including a subject antibody
  • a subject antibody comprises one or more non-naturally occurring amino acids.
  • the non-naturally encoded amino acid comprises a carbonyl group, an acetyl group, an aminooxy group, a hydrazine group, a hydrazide group, a semicarbazide group, an azide group, or an alkyne group. See, e.g., U.S. Patent No. 7,632,924 for suitable non-naturally occurring amino acids.
  • Inclusion of a non- naturally occurring amino acid can provide for linkage to a polymer, a second polypeptide, a scaffold, etc.
  • a subject antibody linked to a water-soluble polymer can be made by reacting a water-soluble polymer (e.g., poly(ethylene glycol) (PEG)) that comprises a carbonyl group to an the subject antibody that comprises a non-naturally encoded amino acid that comprises an aminooxy, hydrazine, hydrazide or semicarbazide group.
  • a subject antibody linked to a water-soluble polymer can be made by reacting a subject antibody that comprises an alkyne-containing amino acid with a water-soluble polymer (e.g., PEG) that comprises an azide moiety; in some embodiments, the azide or alkyne group is linked to the PEG molecule through an amide linkage.
  • non-naturally encoded amino acid refers to an amino acid that is not one of the 20 common amino acids or pyrolysine or selenocysteine.
  • Other terms that may be used synonymously with the term “non-naturally encoded amino acid” are “non-natural amino acid,” “unnatural amino acid,” “non-naturally-occurring amino acid,” and variously hyphenated and non- hyphenated versions thereof.
  • the term “non-naturally encoded amino acid” also includes, but is not limited to, amino acids that occur by modification (e.g.
  • a naturally encoded amino acid including but not limited to, the 20 common amino acids or pyrolysine and selenocysteine
  • examples of such non-naturally-occurring amino acids include, but are not limited to, N- acetylglucosaminyl-L-serine, N-acetylglucosaminyl-L-threonine, and O-phosphotyrosine.
  • a subject antibody is linked (e.g., covalently linked) to a polymer (e.g., a polymer other than a polypeptide).
  • Suitable polymers include, e.g., biocompatible polymers, and water-soluble biocompatible polymers. Suitable polymers include synthetic polymers and naturally-occurring polymers. Suitable polymers include, e.g., substituted or unsubstituted straight or branched chain polyalkylene, polyalkenylene or polyoxyalkylene polymers or branched or unbranched polysaccharides, e.g. a homo- or hetero- polysaccharide. Suitable polymers include, e.g., ethylene vinyl alcohol copolymer
  • polybutylmethacrylate poly(hydroxyvalerate); poly(L-lactic acid); polycaprolactone; poly(lactide-co-glycolide); poly(hydroxybutyrate); poly(hydroxybutyrate-co-valerate); polydioxanone; polyorthoester; polyanhydride; poly(glycolic acid); poly(D,L-lactic acid); poly(glycolic acid-co-trimethylene carbonate); polyphosphoester; polyphosphoester urethane; poly(amino acids); cyanoacrylates; poly(trimethylene carbonate);
  • poly(iminocarbonate); copoly(ether-esters) e.g., poly(ethylene oxide)-poly(lactic acid) (PEO/PLA) co-polymers
  • polyalkylene oxalates polyphosphazenes
  • biomolecules such as fibrin, fibrinogen, cellulose, starch, collagen and hyaluronic acid
  • polyurethanes polyurethanes
  • silicones such as polyvinyl chloride; polyvinyl ethers, such as polyvinyl methyl ether; polyvinylidene halides, such as polyvinylidene fluoride and polyvinylidene chloride; polyacrylonitrile; polyvinyl ketones; polyvinyl aromatics, such as polystyrene; polyvinyl esters, such as polyvinyl acetate; copolymers of vinyl monomers with each other and olefins, such as ethylene-methyl methacrylate copolymers, acrylonitrile-styrene copolymers, ABS resins, and ethylene-vinyl acetate copolymers; polyamides, such as Nylon 66 and
  • polycaprolactam alkyd resins; polycarbonates; polyoxymethylenes; polyimides;
  • polyethers epoxy resins; polyurethanes; rayon; rayon-triacetate; cellulose; cellulose acetate; cellulose butyrate; cellulose acetate butyrate; cellophane; cellulose nitrate;
  • cellulose propionate cellulose ethers; amorphous Teflon; poly(ethylene glycol); and carboxymethyl cellulose.
  • Suitable synthetic polymers include unsubstituted and substituted straight or branched chain poly(ethyleneglycol), poly(propyleneglycol) poly(vinylalcohol), and derivatives thereof, e.g., substituted poly(ethyleneglycol) such as methoxypoly(ethyleneglycol), and derivatives thereof.
  • Suitable naturally-occurring polymers include, e.g., albumin, amylose, dextran, glycogen, and derivatives thereof.
  • Suitable polymers can have an average molecular weight in a range of from 500 Da to
  • the PEG or methoxypoly(ethyleneglycol) polymer can have a molecular weight in a range of from about 0.5 kiloDaltons (kDa) to 1 kDa, from about 1 kDa to 5 kDa, from 5 kDa to 10 kDa, from 10 kDa to 25 kDa, from 25 kDa to 40 kDa, or from 40 kDa to 60 kDa.
  • kDa kiloDaltons
  • a subject antibody is covalently linked to a PEG polymer.
  • a subject scFv multimer is covalently linked to a PEG polymer. See, e.g., Albrecht et al. (2006) J. Immunol. Methods 310: 100. Methods and reagents suitable for PEGylation of a protein are well known in the art and may be found in, e.g., U.S. Pat. No. 5,849,860.
  • PEG suitable for conjugation to a protein is generally soluble in water at room temperature, and has the general formula R(0-CH 2 -CH 2 ) n O-R, where R is hydrogen or a protective group such as an alkyl or an alkanol group, and where n is an integer from 1 to 1000. Where R is a protective group, it generally has from 1 to 8 carbons.
  • the PEG conjugated to the subject antibody can be linear.
  • the PEG conjugated to the subject protein may also be branched. Branched PEG derivatives such as those described in U.S. Pat. No. 5,643,575, "star-PEG's" and multi-armed PEG's such as those described in Shearwater Polymers, Inc. catalog "Polyethylene Glycol Derivatives 1997-1998.” Star PEGs are described in the art including, e.g., in U.S. Patent No. 6,046,305.
  • a subject antibody can be glycosylated, e.g., a subject antibody can comprise a covalently linked carbohydrate or polysaccharide moiety.
  • Glycosylation of antibodies is typically either N-linked or O-linked.
  • N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue.
  • the tripeptide sequences asparagine-X- serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain.
  • the presence of either of these tripeptide sequences in a polypeptide creates a potential glycosylation site.
  • O-linked glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine, 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 an 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 O-linked glycosylation sites).
  • removal of the amino acid sequence such that it contains one or more of the above-described tripeptide sequences (for N-linked glycosylation sites).
  • glycosylation sites can be accomplished by amino acid alteration within the native glycosylation sites of an antibody.
  • a subject antibody will in some embodiments comprise a "radiopaque" label, e.g. a label that can be easily visualized using for example x-rays.
  • Radiopaque materials are well known to those of skill in the art. The most common radiopaque materials include iodide, bromide or barium salts. Other radiopaque materials are also known and include, but are not limited to organic bismuth derivatives (see, e.g., U.S. Pat. No. 5,939,045), radiopaque multiurethanes (see U.S. Pat. No. 5,346,981), organobismuth composites (see, e.g., U.S. Pat. No.
  • a subject antibody can be covalently linked to a second moiety (e.g., a lipid, a polypeptide other than a subject antibody, a synthetic polymer, a carbohydrate, and the like) using for example, glutaraldehyde, a homobifunctional cross-linker, or a heterobifunctional cross- linker.
  • Glutaraldehyde cross-links polypeptides via their amino moieties.
  • Homobifunctional cross-linkers e.g., a homobifunctional imidoester, a homobifunctional N-hydroxysuccinimidyl (NHS) ester, or a homobifunctional sulfhydryl reactive cross- linker
  • a homobifunctional imidoester e.g., a homobifunctional N-hydroxysuccinimidyl (NHS) ester, or a homobifunctional sulfhydryl reactive cross- linker
  • Homobifunctional NHS ester and imido esters cross-link amine containing polypeptides. In a mild alkaline pH, imido esters react only with primary amines to form imidoamides, and overall charge of the cross-linked polypeptides is not affected.
  • Homobifunctional sulfhydryl reactive cross-linkers includes bismaleimidhexane (BMH), l,5-difluoro-2,4-dinitrobenzene (DFDNB), and l,4-di-(3',2'-pyridyldithio) propinoamido butane (DPDPB).
  • BMH bismaleimidhexane
  • DFDNB l,5-difluoro-2,4-dinitrobenzene
  • DPDPB l,4-di-(3',2'-pyridyldithio) propinoamido butane
  • Heterobifunctional cross-linkers have two or more different reactive moieties (e.g., amine reactive moiety and a sulfhydryl-reactive moiety) and are cross-linked with one of the polypeptides via the amine or sulfhydryl reactive moiety, then reacted with the other polypeptide via the non-reacted moiety.
  • Multiple heterobifunctional haloacetyl cross- linkers are available, as are pyridyl disulfide cross -linkers.
  • Carbodiimides are a classic example of heterobifunctional cross-linking reagents for coupling carboxyls to amines, which results in an amide bond.
  • a subject antibody can be immobilized on a solid support.
  • Suitable supports are well known in the art and comprise, inter alia, commercially available column materials, polystyrene beads, latex beads, magnetic beads, colloid metal particles, glass and/or silicon chips and surfaces, nitrocellulose strips, nylon membranes, sheets, duracytes, wells of reaction trays (e.g., multi-well plates), plastic tubes, etc.
  • a solid support can comprise any of a variety of substances, including, e.g., glass, polystyrene, polyvinyl chloride, polypropylene, polyethylene, polycarbonate, dextran, nylon, amylose, natural and modified celluloses, polyacrylamides, agaroses, and magnetite.
  • Solid supports can be soluble or insoluble, e.g., in aqueous solution.
  • a suitable solid support is generally insoluble in an aqueous solution.
  • a subject antibody will in some embodiments comprise a detectable label.
  • Suitable detectable labels include any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means. Suitable include, but are not limited to, magnetic beads (e.g.
  • DynabeadsTM fluorescent dyes (e.g., fluorescein isothiocyanate, texas red, rhodamine, a green fluorescent protein, a red fluorescent protein, a yellow fluorescent protein, and the like), radiolabels (e.g., 3 H, 125 I, 35 S, 14 C, or 32 P), enzymes (e.g., horse radish peroxidase, alkaline phosphatase, luciferase, and others commonly used in an enzyme-linked immunosorbent assay (ELISA)), and colorimetric labels such as colloidal gold or colored glass or plastic (e.g. polystyrene, polypropylene, latex, etc.) beads.
  • fluorescent dyes e.g., fluorescein isothiocyanate, texas red, rhodamine, a green fluorescent protein, a red fluorescent protein, a yellow fluorescent protein, and the like
  • radiolabels e.g., 3 H, 125
  • a subject antibody comprises a contrast agent or a
  • radioisotope where the contrast agent or radioisotope is one that is suitable for use in imaging, e.g., imaging procedures carried out on humans.
  • labels include radioisotope such as 131 I (iodine), 18 F (fluorine), 99 Tc (technetium), U1 ln (indium), and 67 Ga (gallium), and contrast agent such as gadolinium (Gd), dysprosium, and iron.
  • Radioactive Gd isotopes ( 153 Gd) also are available and suitable for imaging procedures in non-human mammals.
  • a subject antibody can be labeled using standard techniques.
  • a subject antibody can be iodinated using chloramine T or 1,3,4,6-tetrachloro- 3a,6a-dephenylglycouril.
  • fluorination fluorine is added to a subject antibody during the synthesis by a fluoride ion displacement reaction. See, Muller-Gartner, H., TIB Tech., 16: 122-130 (1998) and Saji, H., Crit. Rev. Ther.
  • a subject antibody can also be labeled with a contrast agent through standard techniques.
  • a subject antibody can be labeled with Gd by conjugating low molecular Gd chelates such as Gd diethylene triamine pentaacetic acid (GdDTPA) or Gd tetraazacyclododecanetetraacetic (GdDOTA) to the antibody.
  • GdDTPA Gd diethylene triamine pentaacetic acid
  • GdDOTA Gd tetraazacyclododecanetetraacetic
  • a subject antibody can be labeled with Gd by, for example, conjugating polylysine-Gd chelates to the antibody. See, for example, Curtet et al., Invest. Radiol., 33(10):752-761 (1998).
  • a subject antibody can be labeled with Gd by incubating paramagnetic polymerized liposomes that include Gd chelator lipid with avidin and biotinylated antibody. See, for example, Sipkins et al., Nature Med., 4:623-626 (1998).
  • Suitable fluorescent proteins that can be linked to a subject antibody include, but are not limited to, a green fluorescent protein from Aequoria victoria or a mutant or derivative thereof e.g., as described in U.S. Patent No. 6,066,476; 6,020,192; 5,985,577; 5,976,796; 5,968,750; 5,968,738; 5,958,713; 5,919,445; 5,874,304; e.g., Enhanced GFP, many such GFP which are available commercially, e.g., from Clontech, Inc.; a red fluorescent protein; a yellow fluorescent protein; any of a variety of fluorescent and colored proteins from Anthozoan species, as described in, e.g., Matz et al. (1999) Nature Biotechnol. 17:969-973; and the like.
  • a subject antibody will in some embodiments be linked to (e.g., covalently or non- covalently linked) a fusion partner, e.g., a ligand; an epitope tag; a peptide; a protein other than an antibody; and the like.
  • a fusion partner e.g., a ligand; an epitope tag; a peptide; a protein other than an antibody; and the like.
  • Suitable fusion partners include peptides and polypeptides that confer enhanced stability in vivo (e.g., enhanced serum half-life); provide ease of purification, e.g., (His) n , e.g., 6His, and the like; provide for secretion of the fusion protein from a cell; provide an epitope tag, e.g., GST, hemagglutinin (HA; e.g., CYPYDVPDYA; SEQ ID NO://), FLAG (e.g., DYKDDDDK; SEQ ID NO://), c-myc (e.g.,
  • CEQKLISEEDL SEQ ID NO://
  • a detectable signal e.g., an enzyme that generates a detectable product (e.g., ⁇ -galactosidase, luciferase), or a protein that is itself detectable, e.g., a green fluorescent protein, a red fluorescent protein, a yellow fluorescent protein, etc.
  • a detectable product e.g., ⁇ -galactosidase, luciferase
  • a protein that is itself detectable e.g., a green fluorescent protein, a red fluorescent protein, a yellow fluorescent protein, etc.
  • provides for multimerization e.g., a multimerization domain such as an Fc portion of an immunoglobulin; and the like.
  • the fusion may also include an affinity domain, including peptide sequences that can interact with a binding partner, e.g., such as one immobilized on a solid support, useful for identification or purification.
  • a binding partner e.g., such as one immobilized on a solid support
  • Consecutive single amino acids, such as histidine when fused to a protein, can be used for one-step purification of the fusion protein by high affinity binding to a resin column, such as nickel sepharose.
  • affinity domains include His5 (HHHHH) (SEQ ID NO://), HisX6 (HHHHHH) (SEQ ID NO://), C-myc (EQKLISEEDL) (SEQ ID NO://), Flag (DYKDDDDK) (SEQ ID NO://), StrepTag (WSHPQFEK) (SEQ ID NO://), hemagglutinin, e.g., HA Tag (YPYDVPDYA; SEQ ID NO://), glutathinone-S-transferase (GST), thioredoxin, cellulose binding domain, RYIRS (SEQ ID NO://), Phe-His-His-Thr (SEQ ID NO://), chitin binding domain, S-peptide, T7 peptide, SH2 domain, C-end RNA tag, WEAAAREACCRECCARA (SEQ ID NO://), metal binding domains, e.g., zinc binding domains or calcium binding domains such as those from calcium-binding proteins, e.g., calmodul
  • a subject antibody is modified to include a carbohydrate moiety, where the carbohydrate moiety can be covalently linked to the antibody.
  • a subject antibody is modified to include a lipid moiety, where the lipid moiety can be covalently linked to the antibody.
  • Suitable lipid moieties include, e.g., an N- fatty acyl group such as N-lauroyl, N-oleoyl, etc.; a fatty amine such as dodecyl amine, oleoyl amine, etc.; a C3-C16 long-chain aliphatic lipid; and the like. See, e.g., U.S. Pat. No. 6,638,513).
  • a subject antibody is incorporated into a liposome.
  • a subject anti-Gremlin-2 antibody can be modified to include a moiety that
  • the modified antibody modifies cellular uptake relative to unconjugated material.
  • the modified antibody may exhibit increased cellular uptake relative to unconjugated material.
  • the modified antibody exhibits decreased cellular uptake relative to unmodified antibody.
  • the efficiency of cellular uptake can be increased or decreased by linking to peptides or proteins that facilitate endocytosis.
  • a given antibody can be linked to a ligand for a target receptor or large molecule that is more easily engulfed by endocytotic mechanisms, such as another antibody.
  • the antibody or other ligand can then be internalized by endocytosis and the payload released by acid hydrolysis or enzymatic activity when the endocytotic vesicle fuses with lysosomes.
  • the conjugate may be one that increases endocytosis relative to unconjugated antibody.
  • the modified antibody can include a ligand that retains the antibody on the surface of a cell, which can be useful as a control for cellular uptake, or in some instances decrease uptake in one cell type while increasing it in others.
  • a subject anti-Gremlin-2 antibody can comprise one or more moieties, which
  • moieties may be linked (e.g., covalently or non-covalently linked) to the anti-Gremlin-2 antibody, either directly or via a linker, e.g. a flexible linker.
  • a linker e.g. a flexible linker.
  • the heterologous fusion partner can be linked to the anti-Gremlin-2 antibody via a linker.
  • Linkers suitable for use in attaching a moiety to a subject anti-Gremlin-2 antibody include "flexible linkers". If present, the linker molecules are generally of sufficient length to permit the anti-Gremlin-2 antibody and a linked carrier to allow some flexible movement between the anti-Gremlin-2 antibody and the carrier. The linker molecules are generally about 6-50 atoms long. The linker molecules may also be, for example, aryl acetylene, ethylene glycol oligomers containing 2-10 monomer units, diamines, diacids, amino acids, or combinations thereof. Other linker molecules which can bind to polypeptides may be used in light of this disclosure.
  • Suitable linkers can be readily selected and can be of any of a suitable of different lengths, such as from 1 amino acid (e.g., Gly) to 20 amino acids, from 2 amino acids to 15 amino acids, from 3 amino acids to 12 amino acids, including 4 amino acids to 10 amino acids, 5 amino acids to 9 amino acids, 6 amino acids to 8 amino acids, or 7 amino acids to 8 amino acids, and may be 1, 2, 3, 4, 5, 6, or 7 amino acids.
  • Exemplary flexible linkers include glycine polymers (G) n , glycine-serine polymers
  • glycine- alanine polymers including, for example, (GS) n , GSGGS n (SEQ ID NO: 1) and GGGS n (SEQ ID NO: 2), where n is an integer of at least one
  • Glycine and glycine-serine polymers are of interest since both of these amino acids are relatively unstructured, and therefore may serve as a neutral tether between components.
  • Glycine polymers are of particular interest since glycine accesses significantly more phi-psi space than even alanine, and is much less restricted than residues with longer side chains (see Scheraga, Rev. Computational Chem.
  • Exemplary flexible linkers include, but are not limited GGSG (SEQ ID NO:3), GGSGG (SEQ ID NO:4), GSGSG (SEQ ID NO: 5), GSGGG (SEQ ID NO: 6), GGGSG (SEQ ID NO: 7), GSSSG (SEQ ID NO: 8), and the like.
  • GGSG SEQ ID NO:3
  • GGSGG SEQ ID NO:4
  • GSGSG SEQ ID NO: 5
  • GSGGG SEQ ID NO: 6
  • GGGSG SEQ ID NO: 7
  • GSSSG SEQ ID NO: 8
  • Gremlin-2 include nucleic acid agents, e.g., a short interfering nucleic acid (siNA) agent that specifically targets Gremlin-2, reduces expression of Gremlin-2, and reduces the level of Gremlin-2.
  • siNA short interfering nucleic acid
  • Short interfering nucleic acids include small nucleic acid molecules, such as a short interfering RNA (siRNA), a double- stranded RNA (dsRNA), a micro-RNA (miRNA), and a short hairpin RNA (shRNA).
  • siRNA short interfering RNA
  • dsRNA double- stranded RNA
  • miRNA micro-RNA
  • shRNA short hairpin RNA
  • short interfering nucleic acid short interfering nucleic acid
  • siNA short interfering nucleic acid
  • siRNA short interfering RNA
  • RNA short interfering nucleic acid molecule
  • oligonucleotide molecule short interfering oligonucleotide molecule
  • chemically- modified short interfering nucleic acid molecule refer to any nucleic acid molecule capable of inhibiting or down regulating gene expression, for example by mediating RNA interference "RNAi” or gene silencing in a sequence- specific manner. Design of RNAi molecules, given a target gene, is routine in the art. See also US 2005/0282188 (which is incorporated herein by reference) as well as references cited therein. See, e.g., Pushparaj et al. Clin Exp Pharmacol Physiol.
  • siNAs including siRNAs
  • Gremlin-2 gene for the purposes disclosed herein will be readily apparent to the ordinarily skilled artisan, as are methods of production of siNAs having modifications (e.g., chemical modifications) to provide for, e.g., enhanced stability, bioavailability, and other properties to enhance use as therapeutics.
  • modifications e.g., chemical modifications
  • methods for formulation and delivery of siNAs to a subject are also well known in the art. See, e.g., US 2005/0282188; US 2005/0239731; US 2005/0234232; US 2005/0176018; US 2005/0059817; US 2005/0020525; US 2004/0192626;
  • DEQOR therefore, can help to predict (i) regions in a gene that show high silencing capacity based on the base pair composition and (ii) siRNAs with high silencing potential for chemical synthesis.
  • each siRNA arising from the input query is evaluated for possible cross-silencing activities by performing BLAST searches against the transcriptome or genome of a selected organism.
  • DEQOR can therefore predict the probability that an mRNA fragment will cross-react with other genes in the cell and helps researchers to design experiments to test the specificity of siRNAs or chemically designed siRNAs.
  • Suitable Gremlin-2 gene targets include, e.g., a contiguous stretch of from about 10 nucleotides (nt) to about 15 nt, from about 15 nt to about 20 nt, from about 20 nt to about 25 nt, from about 25 nt to about 30 nt, from about 30 nt to about 35 nt, from about 35 nt to about 40 nt, from about 40 nt to about 50 nt, from about 50 nt to about 60 nt, from about 60 nt to about 70 nt, from about 70 nt to about 80 nt, from about 80 nt to about 90 nt, or from about 90 nt to about 100 nt, of a Gremlin-2 mRNA, e.g., of nucleotides 1-4199 of a Gremlin-2 mRNA provided in GenBank NM_022469 ⁇ Homo sapiens Gremlin-2 mRNA),
  • siNA e.g., siRNA, e.g., shRNA. e.g. dsRNA
  • oligonucleotides used would comprise a sequence complementary to the target.
  • siNA molecules can be of any of a variety of forms.
  • the siNA can be a double- stranded polynucleotide molecule comprising self-complementary sense and antisense regions, wherein the antisense region comprises nucleotide sequence that is complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region having nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof.
  • siNA can also be assembled from two separate oligonucleotides, where one strand is the sense strand and the other is the antisense strand, wherein the antisense and sense strands are self-complementary.
  • each strand comprises nucleotide sequence that is complementary to nucleotide sequence in the other strand; such as where the antisense strand and sense strand form a duplex or double stranded structure, for example wherein the double stranded region is about 15 to about 30, e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 base pairs;
  • the antisense strand comprises nucleotide sequence that is complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense strand comprises nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof (e.g., about 15 to about 25 or more nucleotides of the siNA molecule are complementary to the target nucleic acid
  • the siNA can be assembled from a single oligonucleotide, where the self-complementary sense and antisense regions of the siNA are linked by a nucleic acid- based or non-nucleic acid-based linker(s).
  • the siNA can be a polynucleotide with a duplex, asymmetric duplex, hairpin or asymmetric hairpin secondary structure, having self- complementary sense and antisense regions, wherein the antisense region comprises nucleotide sequence that is complementary to nucleotide sequence in a separate target nucleic acid molecule or a portion thereof and the sense region having nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof.
  • the siNA can be a circular single- stranded polynucleotide having two or more loop structures and a stem comprising self-complementary sense and antisense regions, wherein the antisense region comprises nucleotide sequence that is complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region having nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof, and wherein the circular polynucleotide can be processed either in vivo or in vitro to generate an active siNA molecule capable of mediating RNAi.
  • the siNA can also comprise a single stranded polynucleotide having nucleotide sequence complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof (e.g., where such siNA molecule does not require the presence within the siNA molecule of nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof), wherein the single stranded polynucleotide can further comprise a terminal phosphate group, such as a 5'- phosphate (see for example Martinez et al., 2002, Cell., 110, 563-574 and Schwarz et al., 2002, Molecular Cell, 10, 537-568), or 5',3'-diphosphate.
  • a terminal phosphate group such as a 5'- phosphate (see for example Martinez et al., 2002, Cell., 110, 563-574 and Schwarz et al., 2002, Molecular Cell, 10, 537-568), or 5',3'-di
  • the siNA molecule contains separate sense and antisense sequences or regions, wherein the sense and antisense regions are covalently linked by nucleotide or non-nucleotide linkers molecules as is known in the art, or are alternately non-covalently linked by ionic interactions, hydrogen bonding, van der Waals interactions, hydrophobic interactions, and/or stacking interactions.
  • the siNA molecules comprise nucleotide sequence that is complementary to nucleotide sequence of a target gene.
  • the siNA molecule interacts with nucleotide sequence of a target gene in a manner that causes inhibition of expression of the target gene.
  • siNA molecules need not be limited to those molecules containing only RNA, but further encompasses chemically-modified nucleotides and non-nucleotides.
  • the short interfering nucleic acid molecules of the invention lack 2' -hydroxy (2'-OH) containing nucleotides.
  • siNAs do not necessarily require the presence of nucleotides having a 2'-hydroxy group for mediating RNAi and as such, siNA molecules of the invention optionally do not include any ribonucleotides (e.g., nucleotides having a 2'-OH group).
  • siNA molecules that do not require the presence of ribonucleotides within the siNA molecule to support RNAi can however have an attached linker or linkers or other attached or associated groups, moieties, or chains containing one or more nucleotides with 2'-OH groups.
  • siNA molecules can comprise ribonucleotides at about 5, 10, 20, 30, 40, or 50% of the nucleotide positions.
  • the modified short interfering nucleic acid molecules of the invention can also be referred to as short interfering modified oligonucleotides "siMON.”
  • siNA is meant to be equivalent to other terms used to
  • RNAi sequence specific RNA interference
  • siRNA short interfering RNA
  • dsRNA double- stranded RNA
  • miRNA micro-RNA
  • shRNA short hairpin RNA
  • RNAi short interfering oligonucleotide
  • short interfering nucleic acid short interfering modified oligonucleotide
  • chemically-modified siRNA post-transcriptional gene silencing RNA (ptgsRNA)
  • ptgsRNA post-transcriptional gene silencing RNA
  • ptgsRNA post-transcriptional gene silencing RNA
  • siNA molecules of the invention can be used to epigenetically silence a target gene at the post-transcriptional level or the pre-transcriptional level.
  • epigenetic regulation of gene expression by siNA molecules of the invention can result from siNA mediated modification of chromatin structure or methylation pattern to alter gene expression (see, for example, Verdel et al., 2004, Science, 303, 672-676; Pal-Bhadra et al., 2004, Science, 303, 669-672; Allshire, 2002, Science, 297, 1818-1819; Volpe et al., 2002, Science, 297, 1833-1837; Jenuwein, 2002, Science, 297, 2215-2218; and Hall et al., 2002, Science, 297, 2232-2237).
  • siNA molecules contemplated herein can comprise a duplex forming
  • siNA molecules also contemplated herein include multifunctional siNA, (see, e.g., WO 05/019453 and US 2004/0249178).
  • multifunctional siNA can comprise sequence targeting, for example, two regions of PKDl.
  • siNA molecules contemplated herein can comprise an asymmetric hairpin or
  • asymmetric hairpin as used herein is meant a linear siNA molecule comprising an antisense region, a loop portion that can comprise nucleotides or non-nucleotides, and a sense region that comprises fewer nucleotides than the antisense region to the extent that the sense region has enough complementary nucleotides to base pair with the antisense region and form a duplex with loop.
  • an asymmetric hairpin siNA molecule can comprise an antisense region having length sufficient to mediate RNAi in a cell or in vitro system (e.g.
  • the asymmetric hairpin siNA molecule can also comprise a 5'-terminal phosphate group that can be chemically modified.
  • the loop portion of the asymmetric hairpin siNA molecule can comprise nucleotides, non-nucleotides, linker molecules, or conjugate molecules as described herein.
  • asymmetric duplex as used herein is meant a siNA molecule having two
  • an asymmetric duplex siNA molecule of the invention can comprise an antisense region having length sufficient to mediate RNAi in a cell or in vitro system (e.g.
  • nucleotides about 15 to about 30, or about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides
  • a sense region having about 3 to about 25 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) nucleotides that are complementary to the antisense region.
  • siNAs e.g., siRNAs, shRNAs, etc.
  • modifications base, sugar and/or phosphate
  • serum ribonucleases which can increase their potency
  • oligonucleotides can be modified to enhance stability and/or enhance biological activity by modification with nuclease resistant groups, for example, 2'-amino, 2'-C-allyl, 2'-fluoro, 2'-0-methyl, 2'-0-allyl, 2'-H, nucleotide base modifications (for a review see Usman and Cedergren, 1992, TIBS. 17, 34; Usman et al., 1994, Nucleic Acids Symp. Ser. 31, 163; Burgin et al., 1996, Biochemistry, 35, 14090).
  • nuclease resistant groups for example, 2'-amino, 2'-C-allyl, 2'-fluoro, 2'-0-methyl, 2'-0-allyl, 2'-H, nucleotide base modifications (for a review see Usman and Cedergren, 1992, TIBS. 17, 34; Usman et al., 1994, Nucleic Acids Symp. Ser. 31, 163; Burgin e
  • Short interfering nucleic acid (siNA) molecules having chemical modifications that maintain or enhance activity are contemplated herein. Such a nucleic acid is also generally more resistant to nucleases than an unmodified nucleic acid. Accordingly, the in vitro and/or in vivo activity should not be significantly lowered. Nucleic acid molecules delivered exogenously are generally selected to be stable within cells at least for a period sufficient for transcription and/or translation of the target RNA to occur and to provide for modulation of production of the encoded mRNA and/or polypeptide so as to facilitate reduction of the level of the target gene product.
  • RNA and DNA molecules can be accomplished synthetically and can provide for introduction of nucleotide modifications to provide for enhanced nuclease stability, (see, e.g., Wincott et al., 1995, Nucleic Acids Res. 23, 2677; Caruthers et al., 1992, Methods in Enzymology 211, 3-19, incorporated by reference herein.
  • nucleic acids of the present disclosure include one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) G-clamp nucleotides, which are modified cytosine analogs which confer the ability to hydrogen bond both Watson-Crick and Hoogsteen faces of a complementary guanine within a duplex, and can provide for enhanced affinity and specificity to nucleic acid targets (see, e.g., Lin et al. 1998, J. Am. Chem. Soc, 120, 8531- 8532).
  • nucleic acid molecules can include one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) LNA "locked nucleic acid" nucleotides such as a 2',4'-C methylene bicyclo nucleotide (see, e.g., Wengel et al., WO 00/66604 and WO 99/14226).
  • LNA "locked nucleic acid" nucleotides such as a 2',4'-C methylene bicyclo nucleotide (see, e.g., Wengel et al., WO 00/66604 and WO 99/14226).
  • siNA molecules can be provided as conjugates and/or complexes, e.g., to facilitate delivery of siNA molecules into a cell.
  • exemplary conjugates and/or complexes include those composed of an siNA and a small molecule, lipid, cholesterol, phospholipid, nucleoside, antibody, toxin, negatively charged polymer (e.g., protein, peptide, hormone, carbohydrate, polyethylene glycol, or polyamine).
  • the transporters described are designed to be used either individually or as part of a multi-component system, with or without degradable linkers. These compounds can improve delivery and/or localization of nucleic acid molecules into cells in the presence or absence of serum (see, e.g., US 5,854,038).
  • Conjugates of the molecules described herein can be attached to biologically active molecules via linkers that are biodegradable, such as biodegradable nucleic acid linker molecules.
  • Interfering RNAs may be generated exogenously by chemical synthesis, by in vitro transcription, or by cleavage of longer double- stranded RNA with dicer or another appropriate nuclease with similar activity.
  • DNA/RNA synthesizer may be obtained from commercial suppliers such as Ambion Inc. (Austin, Tex.), Invitrogen (Carlsbad, Calif.), or Dharmacon (Lafayette, Colo.). Interfering RNAs are purified by extraction with a solvent or resin, precipitation, electrophoresis, chromatography, or a combination thereof, for example. Alternatively, interfering RNA may be used with little if any purification to avoid losses due to sample processing.
  • Interfering RNAs can also be expressed endogenously from plasmid or viral
  • RNA expression vectors or from minimal expression cassettes for example, polymerase chain reaction (PCR)-generated fragments comprising one or more promoters and an appropriate template or templates for the interfering RNA.
  • PCR polymerase chain reaction
  • the present disclosure provides a recombinant expression vector comprising a nucleotide sequence encoding an interfering RNA.
  • the nucleotide sequence encoding the interfering RNA can be operably linked to a promoter, e.g., an inducible promoter, a muscle- specific promoter, a constitutive promoter, etc.
  • Examples of commercially available plasmid-based expression vectors for shRNA include members of the pSilencer series (Ambion, Austin, Tex.) and pCpG-siRNA (InvivoGen, San Diego, Calif.).
  • Viral vectors for expression of interfering RNA may be derived from a variety of viruses including adenovirus, adeno-associated virus, lenti virus (e.g., human immunodeficiency virus-based vectors, feline immunodeficiency virus-based vectors, and EIAV), and herpes virus.
  • Examples of commercially available viral vectors for shRNA expression include pSilencer adeno (Ambion, Austin, Tex.) and
  • kits for production of PCR- generated shRNA expression cassettes include Silencer Express (Ambion, Austin, Tex.) and siXpress (Minis, Madison, Wis.).
  • an interfering RNA-encoding nucleotide sequence is
  • a muscle-specific control element e.g., a promoter, an enhancer
  • Muscle-specific promoters and other control elements are known in the art.
  • Suitable muscle-specific control sequences include, but are not limited to, a Pitx3 promoter (see, e.g., Coulon et al. (2007) J. Biol. Chem. 282:33192); a desmin promoter (see, e.g., Talbot et al. (2010) Mol. Ther. 18:601); a muscle creatine kinase promoter (see, e.g., Wang et al. (2008) Gene Ther. 15: 1489); a smooth muscle a-actin promoter; an a- myosin heavy chain promoter (see, e.g., Franz et al.
  • a myosin light chain 2 promoter a mef2c promoter; a fast skeletal troponin T promoter (see, e.g., Stefancsik et al. (2003) Comp. Funct. Genomics 4:609); a SM22a promoter (see, e.g., Akyurek et al. (2000) Mol. Med. 6:983; and U.S. Patent No. 7,169,874); a smoothelin promoter (see, e.g., WO 2001/018048); an a-smooth muscle actin promoter; and the like. See also, e.g., Pacak et al.
  • An interfering RNA can be delivered in a delivery system that provides tissue
  • a suitable formulation for an interfering nucleic acid can include one or more additional properties: 1) nucleic acid binding into a core that can release the siRNA into the cytoplasm; 2) protection from non-specific interactions; 3) and tissue targeting that provides cell uptake.
  • the composition comprises a modular polymer conjugate targeting muscle cells by coupling a peptide ligand specific for those cells to one end of a protective polymer, coupled at its other end to a cationic carrier for nucleic acids.
  • a suitable polymer conjugate can have three functional domains: peptide ligand specific for a target cell; protective polymer; and cationic carrier for nucleic acids.
  • Another suitable formulation includes surface coatings attached to a preformed nanoparticle.
  • Suitable formulations for delivery of an interfering nucleic acid include polymers, polymer conjugates, lipids, micelles, self-assembly colloids, nanoparticles, sterically stabilized nanoparticles, and ligand-directed nanoparticles.
  • a subject interfering nucleic acid composition can comprise, in addition to a subject interfering nucleic acid, one or more of: a salt, e.g., NaCl, MgCl, KC1, MgS0 4 , etc.; a buffering agent, e.g., a Tris buffer, N-(2-Hydroxyethyl)piperazine-N'-(2- ethanesulfonic acid) (HEPES), 2-(N-Morpholino)ethanesulfonic acid (MES), 2-(N- Morpholino)ethanesulfonic acid sodium salt (MES), 3-(N-Morpholino)propanesulfonic acid (MOPS), N-tris[Hydroxymethyl]methyl-3-aminopropanesulfonic acid (TAPS), etc.; a solubilizing agent; a detergent,
  • the present disclosure further provides pharmaceutical compositions comprising a subject interfering nucleic acid.
  • the present disclosure provides a pharmaceutical composition comprising a subject interfering nucleic acid and a pharmaceutically acceptable excipient.
  • Pharmaceutically acceptable excipients are described below.
  • An agent e.g., an anti-Gremlin-2 antibody; a Gremlin-2-specific siNA
  • reduces circulating and/or tissue levels of Gremlin-2 and/or that reduces Gremlin-2 activity can be provided in a pharmaceutical composition, for administration to an individual in need thereof.
  • composition comprising an agent that reduces circulating and/or tissue levels of
  • Gremlin-2 and/or that reduces Gremlin-2 activity can comprise a pharmaceutically acceptable excipient, a variety of which are known in the art and need not be discussed in detail herein.
  • Pharmaceutically acceptable excipients have been amply described in a variety of publications, including, for example, "Remington: The Science and Practice of Pharmacy", 19 th Ed. (1995), or latest edition, Mack Publishing Co; A. Gennaro (2000) “Remington: The Science and Practice of Pharmacy", 20th edition, Lippincott, Williams, & Wilkins; Pharmaceutical Dosage Forms and Drug Delivery Systems (1999) H.C. Ansel et al., eds 7 th ed., Lippincott, Williams, & Wilkins; and Handbook of Pharmaceutical
  • a subject pharmaceutical composition can comprise an agent that reduces
  • a subject pharmaceutical circulating and/or tissue levels of Gremlin-2 and/or that reduces Gremlin-2 activity, and a pharmaceutically acceptable excipient.
  • a subject pharmaceutical circulating and/or tissue levels of Gremlin-2 and/or that reduces Gremlin-2 activity, and a pharmaceutically acceptable excipient.
  • composition will be suitable for injection into a subject, e.g., will be sterile.
  • a subject pharmaceutical composition will be suitable for injection into a human subject, e.g., where the composition is sterile and is free of detectable pyrogens and/or other toxins.
  • a subject pharmaceutical composition may comprise other components, such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium, carbonate, and the like.
  • the compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, toxicity adjusting agents and the like, for example, sodium acetate, sodium chloride, potassium chloride, calcium chloride, sodium lactate, hydrochloride, sulfate salts, solvates (e.g., mixed ionic salts, water, organics), hydrates (e.g., water), and the like.
  • routes of administration may vary.
  • An agent that reduces circulating and/or tissue levels of Gremlin-2 and/or that reduces Gremlin-2 activity can be delivered by a route that provides for delivery of the agent to the bloodstream (e.g., by parenteral administration, such as intravenous administration, intramuscular
  • Injection can be used to accomplish parenteral administration.
  • a specific tissue e.g., muscle tissue.
  • Injection can be used to accomplish parenteral administration.
  • an anti-Gremlin-2 antibody is delivered by a route that provides for delivery of the antibody directly into affected muscle tissue, e.g., by intramuscular injection.
  • a therapeutically effective amount of an agent that reduces the level and/or activity of Gremlin-2 is administered to a subject in need thereof.
  • an agent that reduces levels and/or activity of Gremlin-2 can increase muscle function and/or muscle mass, and can in some cases cause a return to a normal level of muscle function and/or muscle mass relative to a healthy individual when the agent is delivered to the bloodstream or directly into muscle tissue in an effective amount to a patient who previously did not have a normal level of muscle function and/or muscle mass relative to a healthy individual prior to being treated.
  • the amount administered varies depending upon the goal of the administration, the health and physical condition of the individual to be treated, age, the degree of resolution desired, the formulation of a subject antibody, the activity of the subject antibody employed, the treating clinician's assessment of the medical situation, the condition of the subject, and the body weight of the subject, as well as the severity of the disease, disorder, or condition, and other relevant factors.
  • the size of the dose will also be determined by the existence, nature, and extent of any adverse side-effects that might accompany the administration of a particular antibody.
  • the amount of an anti-Gremlin-2 antibody employed to increase muscle mass and/or muscle strength or other muscle function is not more than about the amount that could otherwise be irreversibly toxic to the subject (i.e., maximum tolerated dose). In other cases, the amount is around or even well below the toxic threshold, but still in an effective concentration range, or even as low as threshold dose.
  • suitable doses and dosage regimens can be determined by comparisons to indicators of normal muscle mass and/or function. Such dosages include dosages which result in increased muscle mass and/or function, for example, comparable to a healthy individual, without significant side effects.
  • Dosage treatment may be a single dose schedule or a multiple dose schedule (e.g., including ramp and maintenance doses).
  • a subject composition may be administered in conjunction with other agents, and thus doses and regimens can vary in this context as well to suit the needs of the subject.
  • measurable effect on the subject and may be determined based on the pharmacokinetics and pharmacology for absorption, distribution, metabolism, and excretion ("ADME") of the subject antibody or its by-products, and thus based on the disposition of the composition within the subject.
  • ADME absorption, distribution, metabolism, and excretion
  • administration of a subject antibody can be via injection, e.g., via intravenous injection, intramuscular injection, or a combination thereof.
  • terapéuticaally effective amount is meant that the administration of that
  • the therapeutically effective amount is effective to increase muscle mass and/or muscle function in a subject.
  • the therapeutically effective amount can be adjusted in connection with dosing regimen and diagnostic analysis of the subject's condition (e.g., monitoring muscle mass) and the like.
  • the effective amount of a dose or dosing regimen can be gauged from the ED 50 of an agent for inducing an action that leads to an increase in muscle mass by a certain amount and/or an increase in muscle function by a certain degree.
  • ED 50 (effective dosage) is the intended dosage which induces a response halfway between the baseline and maximum after some specified exposure time.
  • the ED 50 of a graded dose response curve therefore represents the concentration of an agent (e.g., a subject antibody) where 50% of its maximal effect is observed.
  • ED 50 may be determined by in vivo studies (e.g. animal models) using methods known in the art.
  • an effective amount may not be more than 100X the calculated ED 50 .
  • the amount of an agent e.g., an anti-Gremlin-2 antibody
  • the amount of an agent is less than about 100X, less than about 50X, less than about 40X, 35X, 30X, or 25X and many embodiments less than about 20X, less than about 15X and even less than about 10X, 9X, 9X, 7X, 6X, 5X, 4X, 3X, 2X or IX than the calculated ED 50 .
  • the effective amount is about IX to 30X of the calculated ED 50 , and sometimes about IX to 20X, or about IX to 10X of the calculated ED 50 .
  • the effective amount is the same as the calculated ED 50 , and in certain embodiments the effective amount is an amount that is more than the calculated ED 50 .
  • An effective amount of an agent may also an amount that is effective, when administered in one or more doses, to increase muscle function and/or muscle mass by at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or more than 80%, compared to the level of muscle function and/or the muscle mass in the individual not treated with the agent.
  • dose per administration may be at less than 10 ⁇ g, less than 2 ⁇ g, or less than 1 ⁇ g. Dose per administration may also be more than 50 ⁇ g, more 100 ⁇ g, more than 300 ⁇ g up to 600 ⁇ g or more. An example of a range of dosage per weight is about 0.1 ⁇ g/kg to about 1 ⁇ g/kg, up to about 1 mg/kg or more. Effective amounts and dosage regimen can readily be determined empirically from assays, from safety and escalation and dose range trials, individual clinician-patient relationships, as well as in vitro and in vivo assays known in the art.
  • unit dosage form refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of an active agent (e.g., an agent that reduces circulating and/or tissue levels of Gremlin-2) calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle.
  • an active agent e.g., an agent that reduces circulating and/or tissue levels of Gremlin-2
  • the specifications for the novel unit dosage forms depend on the particular protein employed and the effect to be achieved, and the pharmacodynamics associated with each active agent in the host.
  • any of a variety of therapies directed to increasing muscle function and/or muscle mass can be combined in a composition or therapeutic method with an anti-Gremlin-2 antibody.
  • a subject antibody can also be administered in combination with a modified diet and/or exercise regimen to promote muscle strength and/or muscle mass.
  • Second therapeutic agents that can be administered in combination therapy with an anti-Gremlin-2 antibody include, but are not limited to, follistatin (see, e.g., Kota et al. (2009) Sci. Transl. Med. I:6ral5; and U.S. Patent Publication No. 2010/0178348); a follistatin domain-containing protein other than follistatin (see, e.g., U.S. Patent Publication No. 2011/0020372); a corticosteroid; a myostatin inhibitor (see, e.g., U.S. Patent Publication No. 2010/0330072); an anti-activin receptor IIB antibody (see, e.g., U.S. Patent Publication No. 2010/0272734); a truncated activin receptor IIB (see, e.g., U.S. Patent Publication No. 2011/0034372); and the like.
  • follistatin see, e.g.,
  • a subject antibody is administered in combination with one or more other therapies
  • the combination can be administered anywhere from simultaneously to up to 5 hours or more, e.g., 10 hours, 15 hours, 20 hours or more, prior to or after administration of a subject protein.
  • a subject antibody and other therapeutic intervention are administered or applied sequentially, e.g., where a subject antibody is administered before or after another therapeutic treatment.
  • a subject antibody and other therapy are administered simultaneously, e.g., where a subject antibody and a second therapy are administered at the same time, e.g., when the second therapy is a drug it can be administered along with a subject antibody as two separate formulations or combined into a single composition that is administered to the subject.
  • the treatments are considered to be administered together or in combination for purposes of the present disclosure.
  • function and/or muscle mass include individuals having a deficiency in muscle function and/or having reduced muscle mass.
  • Individuals suitable for treatment with a subject method of increasing muscle function and/or muscle mass include individuals having a disease, disorder, or condition associated with or resulting in reduced muscle function and/or muscle mass, e.g., a disease, disorder, or condition in which reduced muscle function and/or muscle mass is a symptom or a sequela of the disease, disorder, or condition.
  • diseases, disorders, or conditions include immobilization, chronic disease, cancer, and injury.
  • Individuals suitable for treatment with a subject method for reducing levels and/or activity of Gremlin-2 include individuals having a disease, disorder, or condition characterized, at least in part, by elevated circulating and/or tissue levels of Gremlin-2.
  • an individual to be treated using a method of the present disclosure can have elevated Gremlin-2 levels, e.g., elevated circulating and/or tissue levels of Gremlin-2, compared to a normal control level of Gremlin-2.
  • Such individuals can include individuals having a disease, disorder, or condition associated with or resulting in reduced muscle function and/or muscle mass, e.g., a disease, disorder, or condition in which reduced muscle function and/or muscle mass is a symptom or a sequela of the disease, disorder, or condition.
  • a Gremlin-2 polypeptide can be targeted in order to reduce levels and/or activity of
  • Gremlin-2 polypeptide and/or to increase muscle function and/or muscle mass in a subject.
  • Methods and compositions targeting a Gremlin-2 polypeptide find use in reducing circulating and/or tissue levels and/or reducing activity of Gremlin-2 polypeptide, and/or increasing muscle function and/or muscle mass in an individual having a disorder, disease, or condition associated with or resulting in reduced muscle function and/or mass.
  • Gremlin-2 polypeptide encompasses naturally-occurring Gremlin-2 polypeptides and homologues from different species.
  • Gremlin-2 polypeptide encompasses naturally- occurring variants.
  • Gremlin-2 encompasses murine and human variants that are encoded by the GREM2 gene or a gene homologous to GREM2.
  • Gremlin-2 refers to Gremlin-2 proteins or Gremlin-2 DNA sequences, which
  • Gremlin-2 protein also refers to proteins that have one or more alteration in the amino acid residues (e.g. at locations that are not conserved across variants and/or species) while retaining the conserved domains and having the same biological activity as the naturally- occurring Gremlin-2.
  • Gremlin-2 also encompasses nucleic acid sequences that vary in one or more bases from a naturally-occurring DNA sequence but still translate into an amino acid sequence that correspond to the a naturally- occurring protein due to degeneracy of the genetic code.
  • Gremlin-2 may also refer to those that differ from the naturally- occurring sequences of Gremlin-2 by one or more conservative substitutions and/or tags and/or conjugates.
  • Proteins targeted by the method of the present disclosure can contain a contiguous amino acid residues of a length derived from Gremlin-2.
  • a sufficient length of contiguous amino acid residues may vary depending on the specific naturally-occurring amino acid sequence from which the protein is derived.
  • the protein may be at least 100 amino acids to 125 amino acid residues in length, at least 125 amino acids to 150 amino acid residues in length, or at least 165 amino acids up to the full-length protein (e.g., 165 amino acids, 166 amino acids, 167 amino acids, 168 amino acids).
  • the protein may be of about 168 amino acid residues in length when derived from a human Gremlin-2 protein or from a mouse Gremlin-2 protein.
  • a protein containing an amino acid sequence that is substantially similar to the amino acid sequence of a Gremlin-2 polypeptide includes a polypeptide comprising an amino acid sequence having at least about 90%, at least about 94%, at least about 95%, at least about 98%, or at least about 99%, or 100%, amino acid sequence identity to a contiguous stretch of from about 100 amino acids (aa) to about 125 aa, from about 125 aa to about 150 aa, from about 150 aa to about 165 aa, or from about 165 aa up to the full length of a naturally occurring Gremlin-2 polypeptide.
  • a Gremlin-2 polypeptide suitable as a target in a subject method can comprise an amino acid sequence having at least about 90%, at least about 94%, at least about 95%, at least about 98%, or at least about 99%, or 100%, amino acid sequence identity to a contiguous stretch of from about 100 amino acids (aa) to about 125 aa, from about 125 aa to about 150 aa, from about 150 aa to about 165 aa, or from 165 aa up to the full length of the human Gremlin-2 polypeptide amino acid sequence depicted in Figure 15.
  • the protein may lack at least 5, at least 10, up to at least 50 or more aa relative to a naturally-occurring full-length Gremlin-2 polypeptide.
  • the protein may not contain the signal sequence of based on the amino acid sequence of a naturally-occurring Gremlin-2 polypeptide.
  • the Gremlin-2 protein may also contain the same or similar glycosylation pattern as that of a naturally- occurring Gremlin-2 polypeptide, may contain no glycosylation, or may contain the glycosylation pattern of host cells used to produce the protein.
  • proteins targeted in the method of the present disclosure include those
  • contiguous amino acid sequences of any naturally-occurring Gremlin-2 polypeptide as well as those having 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 usually no more than 20, 10, or 5 amino acid substitutions, where the substitution is usually a conservative amino acid substitution.
  • conservative amino acid substitution generally refers to substitution of amino acid residues within the following groups:
  • substitutions, insertion, or deletion may be based on alignments of amino acid sequences of different variant proteins or proteins from different species. For example, at certain residue positions that are fully conserved (*), substitution, deletion or insertion may not be allowed while at other positions where one or more residues are not conserved, an amino acid change can be tolerated. Residues that are semi-conserved (. or :) may tolerate changes that preserve charge, polarity, and/or size.
  • the protein may be isolated from a natural source, e.g., is in an environment other than its naturally-occurring environment.
  • the subject protein may also be recombinantly made, e.g., in a genetically modified host cell (e.g., bacteria; yeast; Pichia; insect cells; and the like), where the genetically modified host cell is genetically modified with a nucleic acid comprising a nucleotide sequence encoding the subject protein.
  • the subject protein encompasses synthetic polypeptides, e.g., a subject synthetic polypeptide is synthesized chemically in a laboratory (e.g., by cell-free chemical synthesis). Methods of productions are described in more detail below.
  • polypeptide to be targeted in the subject methods may be generated using
  • nucleic acids encoding a variety of different Gremlin-2 polypeptides are known and available in the art.
  • Nucleotide sequences encoding Gremlin-2 polypeptides are also provided in GenBank. Several sequences and further information on the nucleic acid and protein sequences can also be found below and in the Examples section.
  • GenBank as accession nos.: 1) Homo sapiens: amino acid sequence NP_071914;
  • nucleotide sequence NM_022469
  • Mus musculus amino acid sequence NP_035955; nucleotide sequence NM_011825
  • Rattus norvegicus amino acid sequence XP_222923; nucleotide sequence XM_222923, Pan troglodytes: amino acid sequence XP_525111; nucleotide sequence XM_525111; and Canis familiaris: amino acid sequence XP_547488; nucleotide sequence XM_547488.
  • Exemplary Gremlin-2 amino acid sequences are depicted in Figure 15.
  • nucleotide sequences encoding the protein may be modified so as to optimize the codon usage to facilitate expression in a host cell of interest (e.g., Escherichia coli, and the like). Methods for production of codon optimized sequences are known in the art.
  • Gremlin-2 polypeptides and anti-Gremlin-2 antibodies can be produced by any suitable method, including recombinant and non-recombinant methods (e.g., chemical synthesis).
  • a polypeptide is chemically synthesized, the synthesis may proceed via liquid-phase or solid-phase.
  • Solid-phase synthesis allows the incorporation of unnatural amino acids, peptide/protein backbone modification.
  • Various forms of SPPS such as Fmoc and Boc, are available for synthesizing peptides of the present invention. Details of the chemical synthesis are known in the art (e.g. Ganesan A. 2006 Mini Rev. Med Chem. 6:3-10 and Camarero JA et al. 2005 Protein Pept Lett. 12:723-8). Briefly, small insoluble, porous beads are treated with functional units on which peptide chains are built.
  • the free N-terminal amine of a solid- phase attached is coupled to a single N-protected amino acid unit. This unit is then deprotected, revealing a new N-terminal amine to which a further amino acid may be attached.
  • the peptide remains immobilized on the solid-phase and undergoes a filtration process before being cleaved off.
  • the proteins and/or antibody may be produced as an intracellular protein or as an secreted protein, using any suitable construct and any suitable host cell, which can be a prokaryotic or eukaryotic cell, such as a bacterial (e.g. E. coli) or a yeast host cell, respectively.
  • any suitable host cell which can be a prokaryotic or eukaryotic cell, such as a bacterial (e.g. E. coli) or a yeast host cell, respectively.
  • eukaryotic cells that may be used as host cells include insect cells, mammalian cells, and/or plant cells. Where mammalian host cells are used, the cells may include one or more of the following: human cells (e.g. HeLa, 293, H9 and Jurkat cells); mouse cells (e.g., NIH3T3, L cells, and C127 cells); primate cells (e.g. Cos 1, Cos 7 and CV1) and hamster cells (e.g., Chinese hamster ovary (CHO) cells).
  • human cells e.g. HeLa, 293, H9 and Jurkat cells
  • mouse cells e.g., NIH3T3, L cells, and C127 cells
  • primate cells e.g. Cos 1, Cos 7 and CV1
  • hamster cells e.g., Chinese hamster ovary (CHO) cells.
  • Methods for introduction of genetic material into host cells include, for example, transformation, electroporation, conjugation, calcium phosphate methods and the like.
  • the method for transfer can be selected so as to provide for stable expression of the introduced Gremlin-2-encoding nucleic acid or the introduced anti-Gremlin-2 antibody-encoding nucleic acid.
  • the polypeptide-encoding nucleic acid can be provided as an inheritable episomal element (e.g., plasmid) or can be genomically integrated.
  • plasmid e.g., plasmid
  • a variety of appropriate vectors for use in production of a polypeptide of interest are available commercially.
  • Vectors can provide for extrachromosomal maintenance in a host cell or can
  • the expression vector provides transcriptional and translational regulatory sequences, and may provide for inducible or constitutive expression, where the coding region is operably linked under the
  • transcriptional and translational regulatory sequences may include, but are not limited to, promoter sequences, ribosomal binding sites, transcriptional start and stop sequences, translational start and stop sequences, and enhancer or activator sequences.
  • Promoters can be either constitutive or inducible, and can be a strong constitutive promoter (e.g., T7, and the like).
  • Expression constructs generally have convenient restriction sites located near the promoter sequence to provide for the insertion of nucleic acid sequences encoding proteins of interest.
  • a selectable marker operative in the expression host may be present to facilitate selection of cells containing the vector.
  • the expression construct may include additional elements.
  • the expression vector may have one or two replication systems, thus allowing it to be maintained in organisms, for example in mammalian or insect cells for expression and in a prokaryotic host for cloning and amplification.
  • the expression construct may contain a selectable marker gene to allow the selection of transformed host cells. Selectable genes are well known in the art and will vary with the host cell used.
  • a protein can be isolated from a lysate of cells genetically modified to express the protein constitutively and/or upon induction, or from a synthetic reaction mixture, by immunoaffinity purification, which generally involves contacting the sample with an anti- protein antibody, washing to remove non- specifically bound material, and eluting the specifically bound protein.
  • the isolated protein can be further purified by dialysis and other methods normally employed in protein purification methods.
  • the protein may be isolated using metal chelate chromatography methods. Protein of the present disclosure may contain modifications to facilitate isolation.
  • subject proteins and/or antibody may be prepared in substantially pure or
  • the protein can present in a composition that is enriched for the polypeptide relative to other components that may be present (e.g., other polypeptides or other host cell components).
  • Purified protein may be provided such that the protein is present in a composition that is substantially free of other expressed proteins, e.g., less than 98%, less than 95%, less than 90%, less than 80%, less than 60%, or less than 50%, of the composition is made up of other expressed proteins.
  • a subject anti-Gremlin-2 antibody can be prepared using a wide variety of
  • a subject anti-Gremlin-2 antibody may be made and isolated using methods of phage display.
  • a subject anti- Gremlin-2 antibody may also be isolated from sera of an animal host immunized with an immunogenic composition containing a Gremlin-2 protein, which encompasses whole proteins and fragments thereof.
  • the antigen that coats the wells for phage display panning or the immunogenic composition used to elicit the antibody of the present disclosure may contain an aggregate of one or more Gremlin-2 polypeptides, as described above.
  • the method may involve exposing antigens to an aggregating condition so as to form an aggregate.
  • the methods of production described may further include a step of forming an aggregate of the isolated antigens.
  • the aggregating conditions include heating, addition of an excipient that facilitates aggregation, and the like.
  • Gremlin-2 antibody may be conjugated to another molecule.
  • the antigen can be conjugated to a second molecule such as a peptide, polypeptide, lipid, carbohydrate and the like that aids in solubility, storage or other handling properties, cell permeability, half- life, controls release and/or distribution such as by targeting a particular cell (e.g., muscle cells, etc.) or cellular location (e.g., lysosome, endosome, mitochondria etc.), tissue or other bodily location (e.g., blood, muscle tissue, particular organs etc.).
  • a particular cell e.g., muscle cells, etc.
  • cellular location e.g., lysosome, endosome, mitochondria etc.
  • tissue or other bodily location e.g., blood, muscle tissue, particular organs etc.
  • a particular embodiment of an antigen conjugated to a second molecule is where the second molecule is an immunomodulator.
  • Immunomodulator is a molecule that directly or indirectly modifies an immune response.
  • a specific class of immunomodulators includes those that stimulate or aid in the stimulation of an immunological response.
  • Non- limiting examples include antigens and antigen carriers such as a toxin or derivative thereof, including tetanus toxoid.
  • Phage display can be used for the high-throughput screening of protein interactions.
  • Phages may be utilized to display antigen-binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine). Phage expressing an antigen binding domain that binds Gremlin-2 can be selected or identified with Gremlin-2, e.g., using labeled Gremlin-2 or Gremlin-2 bound or captured to a solid surface or bead. Phage used in these methods are typically filamentous phage including fd and Ml 3 binding domains expressed from phage with Fab, Fv (individual Fv region from light or heavy chains) or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene III or gene VIII protein.
  • a repertoire or combinatorial antibody library e.g., human or murine.
  • ribosomal display can be used to replace bacteriophage as the display platform.
  • Cell surface libraries may be screened for antibodies. Such procedures provide alternatives to traditional hybridoma techniques for the isolation and subsequent cloning of monoclonal antibodies.
  • the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any other desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria.
  • any desired host including mammalian cells, insect cells, plant cells, yeast, and bacteria.
  • techniques to recombinantly produce Fab, Fab' and F(ab') 2 fragments can also be employed using methods known in the art.
  • the method of eliciting antibodies in a host animal involves administering an
  • Gremlin-2 as antigens described above to the host animal (i.e., a suitable mammal such as a mouse, rabbit or guinea pig, or a suitable avian, such as a chicken) to elicit production of an antibody that specifically binds and inhibits Gremlin-2.
  • a suitable mammal such as a mouse, rabbit or guinea pig, or a suitable avian, such as a chicken
  • Methods of immunizing animal including the adjuvants used, booster schedules, sites of injection, suitable animals, etc. are well understood in the art, e.g., Harlow et al.
  • TK thymidine kinase
  • HGPRT hypoxanthine-guanine phosphoribosyl transferase
  • Anti-Gremlin-2 antibodies may also be produced by genetic engineering. In this technique, as with the standard hybridoma procedure, antibody-producing cells are sensitized to the desired antigen or immunogen. The messenger RNA isolated from the immune spleen cells or hybridomas is used as a template to make cDNA using polymerase chain reaction (PCR) amplification. A library of vectors, each containing one heavy chain gene and one light chain gene retaining the initial antigen specificity, is produced by insertion of appropriate sections of the amplified immunoglobulin cDNA into the expression vectors.
  • PCR polymerase chain reaction
  • combinatorial library can be constructed by combining the heavy chain gene library with the light chain gene library. This results in a library of clones which co-express a heavy and light chain (resembling the Fab fragment or antigen binding fragment of an antibody molecule).
  • the vectors that carry these genes are co-transfected into a host (e.g. bacteria, insect cells, mammalian cells, or other suitable protein production host cell.). When antibody gene synthesis is induced in the transfected host, the heavy and light chain proteins self-assemble to produce active antibodies that can be detected by screening with the antigen or immunogen.
  • antibodies are screened using one or a combination of a variety of assays.
  • these assays are functional assays, and may be grouped as follows: assays that detect an antibody's binding affinity or specificity, and assays that detect the ability of an antibody to initialize or inhibit a process.
  • the antigen e.g. Gremlin-2 polypeptide
  • the antigen is coupled to beads or wells or other solid support and incubated with phage displaying the antibody of interest. After washings, bound phage is then recovered by inoculation of log phase E. coli cells. The cells are grown and expanded with helper phage. Steps are repeated for the amplification of tightly bound phages.
  • the phage-infected E. coli colonies after several round of enrichment are harvested and Fab antibodies are purified from the periplasmic fractions. The purified antibodies are then analyzed in accordance with methods known in the art. Certain exemplary examples are detailed below.
  • the population of antibody isolated from phage-infected cells or hybridomas is further analyzed and/or screened for binding to a single antigen (i.e., antigens that are not mixed with other antigens of the plurality of antigens) of the plurality of antigens in vitro or in situ (e.g. on cells). Immuno specific binding may be carried out according to methods routine and known in the art.
  • the immunoassays which can be used include, but are not limited to, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, and protein A
  • the cells and antibodies may be screened based on the ability of the antibody in the supernatant to perform a specific function (e.g. modulation of Gremlin-2).
  • a subject anti-Gremlin-2 antibody may also be screened in vivo. The method
  • In vivo assays of the invention include controls, where suitable controls include a sample in the absence of the antibody.
  • suitable controls include a sample in the absence of the antibody.
  • a plurality of assay mixtures is run in parallel with different antibody concentrations to obtain a differential response to the various concentrations.
  • one of these concentrations serves as a negative control, i.e., at zero concentration or below the level of detection.
  • a monoclonal antibody of interest is one that modulates, e.g.,
  • an undesired symptom in a non-human animal model disease or condition by at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 80%, at least about 90%, or more, when compared to a control in the absence of the antibody.
  • a monoclonal antibody of interest is one that increases muscle function in a non-human animal model disease by at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 80%, at least about 90%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, or more than 5-fold, when compared to a control in the absence of the antibody.
  • a monoclonal antibody of interest will cause an animal to be more similar to an equivalent animal that is not suffering from the disease or condition.
  • Antibodies that have therapeutic value that have been identified using the methods and compositions of the invention are termed "therapeutic" antibodies.
  • the effect of an anti-Gremlin-2 antibody can be determined by in vivo studies (e.g. non-human animal models) using methods known in the art.
  • Suitable non-human animal models include, e.g., the mdx mouse model for human
  • Duchenne Muscular Dystrophy the mdx/utnr " mouse model of Duchenne Muscular Dystrophy; the BIO 14.6 cardiomyopathic hamster; a dysferlin mouse model of myopathy; a sarcoglycan-disrupted mouse model; a laminin a2-deficicient mouse model of congenital muscular dystrophy; other dystrophic mouse models; a non-human animal model (e.g., a mouse model or a rat model) for muscle disuse immobilization; a non-human animal model of muscle injury such as cardiotoxin-induced injury and regeneration model); and the like. See, e.g., Allamand and Campbell (2000) Hum. Mol. Genet.
  • Selected monoclonal antibodies of interest can be expanded in vitro, using routine tissue culture methods, or in vivo, using mammalian subjects.
  • pristane- primed mice can be inoculated with log phase hybridoma cells in phosphate-buffered saline (PBS) for ascites production.
  • Ascites fluid can be stored at -70° C prior to further purification.
  • Cell expressing a monoclonal antibody of interest contains the immunoglobulin heavy and light chain-encoding expression cassettes.
  • the nucleic acids encoding the monoclonal antibody of interest may be identified.
  • the subject nucleic acids may be identified by a variety of methods known to one of skill in the art. Similar methods are used to identify host cell cultures in monoclonal antibody production using hybridoma technology (Harlow et al., Antibodies: A Laboratory Manual, First Edition (1988) Cold spring Harbor, N.Y.), and rely on an "addressable" host cell and an
  • an "addressable" monoclonal antibody such that once a monoclonal antibody of interest is identified, a host cell address may be determined and the nucleic acid encoding the antibody of interested isolated from the cell.
  • nucleic acids encoding a monoclonal antibody of interest may be recovered, characterized and manipulated from a cell expressing the antibody using techniques familiar to one of skill in the art (Ausubel, et al, Short Protocols in Molecular Biology, 3rd ed., Wiley & Sons, (1995) and Sambrook, et al, Molecular Cloning: A Laboratory
  • compositions comprising an anti-Gremlin-2
  • a subject antibody composition can contain, in addition to a subject antibody, one or more of: a salt, e.g., NaCl, MgCl, KC1, MgS0 4 , etc.; a buffering agent, e.g., a Tris buffer, N-(2-Hydroxyethyl)piperazine-N'-(2-ethanesulfonic acid) (HEPES), 2-(N- Morpholino)ethanesulfonic acid (MES), 2-(N-Morpholino)ethanesulfonic acid sodium salt (MES), 3-(N-Morpholino)propanesulfonic acid (MOPS), N-tris[Hydroxymethyl]methyl-3- aminopropanesulfonic acid (TAPS), etc.; a solubilizing agent; a detergent, e.g., a non-ionic detergent such as Tween-20, etc.; a protease inhibitor; glyce
  • compositions comprising a subject antibody may include a buffer, which is
  • composition may comprise a pharmaceutically acceptable excipient, a variety of which are known in the art and need not be discussed in detail herein.
  • Pharmaceutically acceptable excipients have been amply described in a variety of publications, including, for example, "Remington: The Science and Practice of Pharmacy", 19 Ed. (1995), or latest edition, Mack Publishing Co; A. Gennaro (2000) “Remington: The Science and Practice of Pharmacy", 20th edition, Lippincott, Williams, & Wilkins; Pharmaceutical Dosage Forms and Drug Delivery Systems (1999) H.C. Ansel et al., eds 7 th ed., Lippincott, Williams, &
  • a subject pharmaceutical composition can comprise an anti-Gremlin-2 antibody, and a pharmaceutically acceptable excipient.
  • a subject pharmaceutical composition will be suitable for injection into a subject, e.g., will be sterile.
  • a subject pharmaceutical composition will be suitable for injection into a human subject, e.g., where the composition is sterile and is free of detectable pyrogens and/or other toxins.
  • a subject antibody composition may comprise other components, such as
  • compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, toxicity adjusting agents and the like, for example, sodium acetate, sodium chloride, potassium chloride, calcium chloride, sodium lactate, hydrochloride, sulfate salts, solvates (e.g., mixed ionic salts, water, organics), hydrates (e.g., water), and the like.
  • auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, toxicity adjusting agents and the like, for example, sodium acetate, sodium chloride, potassium chloride, calcium chloride, sodium lactate, hydrochloride, sulfate salts, solvates (e.g., mixed ionic salts, water, organics), hydrates (e.g., water), and the like.
  • compositions may include aqueous solution, powder form, granules, tablets, pills, suppositories, capsules, suspensions, sprays, suppositories, and the like.
  • the composition may be formulated according to the different routes of administration described later below.
  • the antibody is administered as an injectable (e.g. subcutaneously,
  • a formulation can be provided as a ready-to-use dosage form, or as non-aqueous form (e.g. a reconstitutable storage- stable powder) or aqueous form, such as liquid composed of pharmaceutically acceptable carriers and excipients.
  • the antibody-containing formulations may also be provided so as to enhance serum half-life of the subject protein following administration.
  • the antibody may be provided in a liposome formulation, prepared as a colloid, or other conventional techniques for extending serum half-life.
  • a variety of methods are available for preparing liposomes, as described in, e.g., Szoka et al. 1980 Ann. Rev. Biophys. Bioeng. 9:467, U.S. Pat. Nos. 4,235,871, 4,501,728 and 4,837,028.
  • the preparations may also be provided in controlled release or slow-release forms.
  • formulations suitable for parenteral administration include
  • a subject pharmaceutical composition can be present in a container, e.g., a sterile container, such as a syringe.
  • the formulations can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use.
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
  • concentration of a subject antibody in a formulation can vary widely (e.g., from less than about 0.1%, usually at or at least about 2% to as much as 20% to 50% or more by weight) and will usually be selected primarily based on fluid volumes, viscosities, and patient-based factors in accordance with the particular mode of administration selected and the patient's needs.
  • a screening method of the present disclosure can be employed to screen for a
  • the method can involve contacting a Gremlin-2 polypeptide with a candidate agent and detecting binding of the candidate agent with Gremlin-2.
  • the method may also involve contacting a Gremlin-2 polypeptide with a candidate agent in the presence or absence of one or more known binding partners of Gremlin-2 and detecting the effect of the candidate agent on binding of Gremlin-2 with the binding partner.
  • the method may also involve the use of libraries of constructs encoding antibodies, aptamers, and/or libraries of small molecules to screen for a Gremlin-2-binding agent.
  • the binding agent may be selected for its potent inhibition of Gremlin-2 activities, inhibition of the expression of mature Gremlin-2, and/or inhibition of the binding affinity for Gremlin-2-interacting proteins.
  • the method may be executed according to methods known in the art.
  • Gremlin-2 e.g. Gremlin-2 alone or Gremlin-2 complexed with its
  • Candidate agents that are effective in disrupting binding of Gremlin-2 to its interacting molecules are selected to be potential agents to be used in diagnostic and therapeutic compositions and methods of use.
  • Candidate agents that can disrupt binding of Gremlin-2 to its interacting molecules encompass those that can decrease the binding affinity of Gremlin-2 to its interacting partners either competitively or noncompetitively.
  • Gremlin-2 that may be used to screen for potential agent include Gremlin-2 as described above.
  • Examples of Gremlin-2 to be used in the subject screening methods include but are not limited to full-length Gremlin-2, mature Gremlin-2, fragments of Gremlin-2 (e.g., a binding partner-binding fragment of Gremlin-2), Gremlin-2 alone, or Gremlin-2 bound to one or more interacting molecules.
  • Gremlin-2 may be immobilized on an ELISA plate or on beads through a covalent or non-covalent interaction, such as hydrophobic adsorption, biotin-avidin interaction, and Ni 2+ -6xHis interaction.
  • a population of candidate agents is then incubated with the immobilized Gremlin-2, washed, and recovered. During selection, the bound candidate is recovered and identified. Multiple successive selection rounds ensure a selection of a candidate that acts as a specific binding agent for Gremlin-2.
  • Other methods such as surface plasmon resonance, western blot, functional assays (e.g. phosphorylation or dephosphorylation of downstream targets), fluorescence activated cell sorting, etc.
  • Gremlin-2 can also be used to screen and select for agents that can bind Gremlin-2, and/or inhibit its interaction to one or more interacting molecules.
  • Other assays known in the art that involve comparing binding and/or activity of Gremlin-2 in the presence or absence of the candidate agents can be employed.
  • Candidate Gremlin-2-binding agents may also be engineered so that the agent
  • KITS contains sites that are known to have affinity for the ligand-binding site (e.g. Gremlin-2 domain homologous to the Gremlin-2-binding site).
  • kits for using the compositions are provided by the present disclosure.
  • kits may be provided for administration of the subject protein in a subject in need of an increase in muscle function and/or muscle mass.
  • the kit can include one or more of a Gremlin-2 polypeptide and/or an anti-Gremlin-2 antibody disclosed herein, which may be provided in a sterile container, and can be provided in formulation with a suitable a pharmaceutically acceptable excipient for administration to a subject.
  • the proteins e.g., an anti-Gremlin-2 antibody
  • the kit may also provide buffers, pharmaceutically acceptable excipient, and the like, packaged separately from the subject protein.
  • the proteins (e.g., an anti-Gremlin-2 antibody) of the present kit may be formulated separately or in combination with other drugs.
  • a subject kit can include: 1) a first container (e.g., a sterile container) comprising a subject pharmaceutical composition (e.g., a pharmaceutical composition comprising a subject anti-Gremlin-2 antibody); and 2) a second container (e.g., a sterile container) comprising a second agent (e.g., a second agent that treats a disease, disorder, or condition resulting in or associated with reduced muscle function and/or reduced muscle mass).
  • a first container e.g., a sterile container
  • a subject pharmaceutical composition e.g., a pharmaceutical composition comprising a subject anti-Gremlin-2 antibody
  • a second container e.g., a sterile container
  • a second agent e.g., a second agent that treats a disease, disorder, or condition resulting in or associated with reduced muscle function and/or reduced muscle mass.
  • kits can further include
  • the instructions for practicing the subject methods are generally recorded on a suitable recording medium.
  • the instructions may be printed on a substrate, such as paper or plastic, etc.
  • the instructions may be present in the kits as a package insert, in the labeling of the container of the kit or components thereof (i.e., associated with the packaging or subpackaging) etc.
  • the instructions are present as an electronic storage data file present on a suitable computer readable storage medium, e.g. CD-ROM, diskette, etc.
  • the actual instructions are not present in the kit, but means for obtaining the instructions from a remote source, e.g. via the internet, are provided.
  • An example of this embodiment is a kit that includes a web address where the instructions can be viewed and/or from which the instructions can be downloaded. As with the instructions, this means for obtaining the instructions is recorded on a suitable substrate.
  • Standard abbreviations may be used, e.g., bp, base pair(s); kb, kilobase(s); pi, picoliter(s); s or sec, second(s); min, minute(s); h or hr, hour(s); aa, amino acid(s); kb, kilobase(s); bp, base pair(s); nt, nucleotide(s); i.m.,
  • mice were kept in accordance with welfare guidelines and project license restrictions under controlled light (12 hr light and 12 hr dark cycle, dark 6:30 pm-6:30 am), temperature (22 ⁇ 4°C) and humidity (50 +20 ) conditions.
  • the mice had free access to water (autoclaved distilled water) and were fed ad libitum on a commercial diet (Harlan laboratories, Irradiated 2018 Teklad Global 18% Protein Rodent Diet) containing 17 kcal% fat, 23 kcal% protein and 60 kcal% carbohydrate.
  • Three-day old neonates were injected with adeno-associated virus (AAV). The injected mice were weaned 3 weeks later and were maintained on 2018 Teklad Global diet containing
  • AAV adeno-associated virus
  • mice 2000mg/kg of doxycyline to induce gene expression (Harlan Laboratories).
  • CX direct cardiotoxin
  • Mdx mice were purchased from Jackson Laboratory (Bar Harbor, ME). Mdx mice were kept and maintained in similar conditions and diet as non-injected C57BL6 mice. All animal studies were approved by the NGM Institutional Animal Care and Use Committee for NGM- 12-2009 entitled
  • PCR recombinant DNA
  • cDNA recombinant DNA
  • PCR reagent kits with Phusion high-fidelity DNA polymerase purchased from New England BioLabs (F-530L, Ipswich, MA). The following primer sequences were used: forward PCR primer: 5' ATGTTCTGGAAGCTCTCGCTGACCTTGCTC and reverse PCR primer: 5'
  • amplified DNA fragment was digested with restriction enzymes Spe I and Not I (the restriction sites were included in the 5' or 3' PCR primers, respectively), to generate an amplification product.
  • the amplification product was then ligated with AAV transgene vectors that had been digested with the same restriction enzymes.
  • the vector used for expression contained a selectable marker and an expression cassette composed of tetracycline response elements flanked by minimal cytomegalovirus (CMV) promoter 5' of a site for insertion of the cloned coding sequence, followed by a 3' untranslated region and bovine growth hormone polyadenylation tail.
  • CMV minimal cytomegalovirus
  • the expression construct is also flanked by internal terminal repeats at the 5' and 3' ends.
  • another vector used for tissue- selective expression contained the same regulatory elements as noted above, and a muscle- specific promoter. Production and purification of AAV
  • AAV 293 cells obtained from Agilent Technologies, Santa Clara, CA.
  • DMEM Dulbecco's Modification of Eagle's Medium
  • Manassas, VA Manassas, VA supplemented with 10% fetal bovine serum and lx antibiotic-antimycotic solution (Mediatech, Inc. Manassas, VA).
  • the cells were plated at 50% density on day 1 in 150 mm cell culture plates and transfected on day 2, using calcium phosphate precipitation method, with the following 3 plasmids (20 ⁇ g/plate of each): AAV transgene plasmid, pHelper plasmids (Agilent Technologies) and AAV2/9 or AAV2/6 plasmid (Gao et al (2004) J. Virol. 78:6381).
  • the cells were scraped off the plates, pelleted by centrifugation at 3000xg and resuspended in buffer containing 20 mM Tris pH 8.5, 100 mM NaCl and 1 mM MgCl 2 .
  • the suspension was frozen in an alcohol dry ice bath and was then thawed in a 37°C water bath. The freeze and thaw cycles were repeated for a total of three times; benzonase (Sigma-Aldrich, St. Louis, MO) were added to 50 units/ml; deoxycholate were added to a final concentration of 0.25%.
  • Viral particles in the supernatant were purified using a discontinuous iodixanol (Sigma-Aldrich, St. Louis, MO) gradient as previously described (Zolotukhin S. et al (1999) Gene Ther. 6:973).
  • the viral stock was concentrated using Vivaspin 20 (MW cutoff 100,000 Dalton, Sartorius Stedim Biotech, Aubagne, France) and re-suspended in phosphate buffered saline (PBS) with 10% glycerol and stored at -80 °C.
  • PBS phosphate buffered saline
  • Grip strength measurements were performed in adult mice at 6, 10 and 14 weeks of age. Briefly, each mouse was held by the tail and allowed to grasp the metallic mesh of the digital grip strength meter (Columbus Instruments International Corporation, Columbus OH, USA). After the mouse grip had been established, the tail was gently pulled away from the mesh until the test animal's grip was broken. The force measured upon release was recorded as peak tension in grams. The test was repeated 10 consecutive times for the same mouse. Data are represented as the average peak tension per test animal. All test subjects were blinded prior to test administration.
  • Magnetic resonance imaging (MRI) Magnetic resonance imaging
  • Body composition measurements were performed in adult mice at 6, 10, and 14 weeks of age using the Echo MRI whole body composition analyzer (Echo Medical Systems, Houston, TX, USA). Briefly, a mouse was individually placed in a designated holder. The holder is then inserted into the MRI device for analyses. Following ⁇ 1 minute reading time, the mouse was then released and the test was complete. Each mouse in a group of 10 was analyzed. Data collected for these analyses include total body weight, lean mass and fat mass.
  • Intrinsic contractile properties of the skeletal muscle were evaluated using muscle physiology assay performed using 1305 5N In Situ Muscle Test System (Aurora Scientific Incorporated, Aurora, ON, Canada). One of the assays used was the measurement of maximum tetanic force generated by specific skeletal muscle group in live animals.
  • the mouse injected with control virus or virus expressing the target molecules was placed under inhaled isofluorane.
  • the hind leg designated for this study was shaved and disinfected.
  • the mouse was placed on a heated platform contained within a physiology apparatus that is capable of maintaining body temperature.
  • a thermometer was placed in the test mouse to closely monitor its body temperature throughout the procedure.
  • the animal was secured by keeping the knee stationary and the foot firmly fixed to a footplate.
  • the knee was secured by inserting a 25 gauge needle directly underneath the knee bone.
  • the inserted needle was firmly fixed onto a clamp ensuring the stability of the knee throughout the procedure.
  • Muscle contraction on the secured hind leg of the test animal was elicited by electrical stimulation of the common peroneal nerve.
  • Gapdh was used as an internal control gene.
  • a primer pair for Gapdh was purchased as VIC labeled Gene Expression Assay kit (Cat#: 4352339E) from Applied Biosystems. 384- well Q-PCR reactions were set-up using 2x QuantiTect Multiplex RT-PCR Master Mix (Qiagen, Valencia, CA, USA) and performed on a 7900HT Fast Real-Time PCR System from Applied Biosystems (Carlsbad, CA, USA). Data are represented as fold expression relative to Gapdh control.
  • Gremlin-2 protein expression level was determined using the ELISA kit obtained from R&D Systems (Minneapolis, MN, USA). Samples analyzed include sera collected from Gremlin-2 injected mice as well as protein lysate prepared from skeletal muscle tissue isolated.
  • shRNA short-hairpin RNA
  • GFP refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery
  • n 10 mice per group.
  • an ELISA was performed using sera obtained from GFP-injected mice and from mice with Gremlin-2 over-expression.
  • the level of Gremlin-2 in GFP-injected mice was 1-2 ng/ml.
  • GFP refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery
  • Gremlin-2 is an important factor in embryo development. To assess the overall effect of Gremlin-2 over-expression, grip strength tests were performed at varying time points in a growing mouse. Tests were performed at 3 weeks, 7 weeks, and 11 weeks after the induction of Gremlin-2 over-expression. Mice performance in the grip strength test showed a marked decrease in peak tension upon Gremlin-2 over-expression when compared to GFP injected mice.
  • GFP refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery
  • body composition measurements by magnetic resonance imaging (MRI) were also performed at 3 weeks, 7 weeks, and 11 weeks post-Gremlin-2 over-expression.
  • the parameters measured in this procedure include total body weight as well as total lean tissue and total fat tissue mass. These analyses revealed that over-expression of Gremlin-2 decreases overall lean mass when compared to GFP-injected mice. This effect was observed at 7 weeks post-Gremlin-2 induction and 11 weeks following Gremlin-2 over- expression (Figure 3). A modest increase in fat mass was observed in the same group of mice. The overall body weight between GFP controls and Gremlin-2 over-expressing mice showed no significant difference.
  • GFP refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery
  • Grem2 wild-type mice injected with lxlOEl 1 GC of rAAV expressing mouse Gremlin-2 via neonate intraperitoneal gene delivery
  • TA tibialis anterior
  • GFP wild-type 10-12 week old mice
  • Gremlin-2 expression results in decreased tetanic force (represented by N.cm) generated by TA muscle.
  • Gremlin-2 over- expression directly impacts skeletal muscle contraction and/or function.
  • GFP refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery
  • Grem2 wild-type injected with lxlOEl l GC of rAAV expressing mouse Gremlin-2 via neonate intraperitoneal gene delivery
  • GFP refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery
  • Grem2 Gremlin-2
  • Serum levels of inorganic phosphate (PHOS), calcium (CA), iron, cholesterol (CHOL), LDL, NEFA, triglyceride (TRIGL), glucose (GLU) (indications of metabolic homeostasis), liver AST, and liver ALT (indications of liver functions) were determined for sera collected from both groups. As seen in Figure 7, no significant differences were identified between Gremlin-2 and GFP groups, indicating Gremlin-2 has no significant impact on general metabolism and liver function.
  • Gremlin-2 expression levels in dystrophic muscles muscle tissues were collected from both wild type and mdx mice. mRNA levels of Gremlin-2 were determined by quantitative PCR. The data are shown in Figure 8. This analysis revealed elevated expression levels of Gremlin-2 in mdx mice.
  • Gremlin-2 protein levels dystrophic muscles serum and muscle tissue samples were collected from both wild type and mdx mice. Protein levels of Gremlin-2 were determined by ELISA. The data are shown in Figure 9. In these sets of analyses, a significant elevation in Gremlin-2 protein in both muscle and serum was observed in mdx mice.
  • Gremlin-2 protein levels injured muscles serum and muscle tissue samples were collected from both wild type and Cardiotoxin (CTX) injected mice. Protein levels of Gremlin-2 were determined by ELISA. The data are shown in Figure 11. In these sets of analyses, a significant elevation in Gremlin-2 protein in both CTX injured muscle and serum was observed.
  • GFP refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery
  • Gremlin-2 shRNA expression was determined by quantitative PCR.
  • GFP refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery
  • Grem2 shRNA "Grem2 shRNA”
  • the levels of Gremlin-2 in skeletal muscle were reduced upon expression of Gremlin-2 shRNA.
  • a grip strength test was performed at 11 weeks after the induction of Gremlin-2 shRNA expression.
  • Mouse performance in grip strength test showed a marked increase in peak tension upon Gremlin-2 shRNA expression when compared to GFP injected mice.
  • GFP refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery
  • body composition measurements by MRI were performed at 11 weeks post-shRNA expression. These measurements revealed that expression of Gremlin-2 shRNA results in an increase in the overall lean mass and body weight when compared to GFP-injected mice. No changes in fat mass were observed in both GFP controls and Gremlin-2 shRNA injected mice.

Abstract

The present disclosure provides compositions and methods for reducing levels and/or activity of Gremlin-2. The present disclosure provides compositions and methods for increasing muscle function and/or muscle mass. The present methods involve using an agent that targets Gremlin-2 polypeptide. Subject compositions and methods are useful for treating various conditions and disorders characterized by loss of muscle function and/or muscle mass.

Description

COMPOSITIONS AND METHODS FOR INCREASING MUSCLE FUNCTION AND MASS
CROSS-REFERENCE TO RELATED APPLICATIONS
[0001] This application claims the priority benefit of U.S. provisional application serial no.
61/471,616, filed April 4, 2011, which application is incorporated herein by reference in its entirety.
INTRODUCTION
[0002] Muscle wasting is associated with a number of diseases and conditions. Currently there are few effective treatments for such disorders.
[0003] There is a need in the art for methods of increasing muscle function and/or mass, in the context of disorders, diseases, and conditions that are associated with or that result in reduced muscle function and/or muscle mass.
SUMMARY
[0004] The present disclosure provides compositions and methods for reducing levels and/or activity of Gremlin-2. The present disclosure provides compositions and methods for increasing muscle function and/or muscle mass. The present methods involve using an agent that targets Gremlin-2 polypeptide. Subject compositions and methods are useful for treating various conditions and disorders characterized by loss of muscle function and/or muscle mass.
BRIEF DESCRIPTION OF THE DRAWINGS
[0005] Figure 1 shows the serum level of a protein of the present disclosure (mouse ortholog) in 14 week old mice that were injected as neonates with an adeno-associated virus (AAV) expressing this protein at day 1-3 by intraperitoneal injection, compared to those of mice injected with a control virus (n=10 per group).
[0006] Figure 2 shows forelimb grip strength of mice that were injected with an adeno-associated virus (AAV) expressing a protein of the present disclosure (mouse ortholog) at day 1-3 by intraperitoneal injection compared to those of mice injected with a control virus (n=10 per group). [0007] Figure 3 shows body weight, lean mass, and fat mass of mice that were injected with AAV expressing a protein of the present disclosure (mouse ortholog) at day 1-3 by
intraperitoneal injection compared to those of mice injected with a control virus (n=10 per group).
[0008] Figure 4 shows Tibialis Anterior (TA) muscle mass of 14 week old mice that were injected with AAV expressing a protein of the present disclosure (mouse ortholog) at day 1-3 by intraperitoneal injection compared to those of mice injected with a control virus (n=l per group).
[0009] Figure 5 shows in situ Tibialis Anterior muscle tetanic force of mice that were injected with AAV expressing a protein of the present disclosure (mouse ortholog) at week 11 by intramuscular injection compared to those of mice injected with a control virus (n=5 per group).
[0010] Figure 6 shows different tissue weights of 14 week old mice that were injected with AAV expressing a protein of the present disclosure (mouse ortholog) at day 1-3 by
intraperitoneal injection compared to those of mice injected with a control virus (n=10 per group).
[0011] Figure 7 shows serum phosphate, calcium, iron, cholesterol, low-density lipoprotein
(LDL), fatty acid, triglyceride (TRIGL), alanine transaminase (ALT), aspartate transaminase (AST), and glucose (GLU) levels of 14 week old mice that were injected with AAV expressing a protein of the present disclosure (mouse ortholog) at day 1-3 by intraperitoneal injection compared to those of mice injected with a control virus (n=10 per group).
[0012] Figure 8 shows levels of message RNA of the present disclosure (mouse ortholog) in
Tibialis Anterior muscle in a 4 week old mouse model for human Duchenne Muscular Dystrophy (DMD).
[0013] Figure 9 shows levels of a protein of the present disclosure (mouse ortholog) in Tibialis Anterior muscle and serum in a 4 week old mouse model for human Duchenne Muscular Dystrophy (DMD).
[0014] Figure 10 shows levels of the message RNA of present disclosure (mouse ortholog) at day 7 post-cardiotoxin injection in Tibialis Anterior muscle of 13 week old mice.
[0015] Figure 11 shows levels of a protein of the present disclosure (mouse ortholog) at day 7 post-cardiotoxin injection in Tibialis Anterior muscle and serum of 13 week old mice. [0016] Figure 12 shows muscle Gremlin-2 message RNA and protein levels of 14 week oldmice that were injected with AAV expressing shRNA against a nucleic acid encoding a protein of the present disclosure (mouse ortholog) at day 1-3 by intraperitoneal injection compared to those of mice injected with a control virus (n=10 per group).
[0017] Figure 13 shows forelimb grip strength of 14 week old mice that were injected with an adeno-associated virus (AAV) expressing shRNA against a nucleic acid encoding a protein of the present disclosure (mouse ortholog) at day 1-3 by intraperitoneal injection compared to those of mice injected with a control virus (n=10 per group).
[0018] Figure 14 shows body weight, lean mass and fat mass of 14 week old mice that were
injected with AAV expressing shRNA against a nucleic acid encoding a protein of the present disclosure (mouse ortholog) at day 1-3 by intraperitoneal injection compared to those of mice injected with a control virus (n=10 per group).
[0019] Figure 15 provides an alignment of Gremlin-2 amino acid sequences.
[0020] Before the present invention is further described, it is to be understood that this invention is not limited to particular embodiments described, as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to be limiting, since the scope of the present invention will be limited only by the appended claims.
[0021] Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range, is encompassed within the invention. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the invention, subject to any specifically excluded limit in the stated range. Where the stated range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the invention.
[0022] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference to disclose and describe the methods and/or materials in connection with which the publications are cited.
[0023] It must be noted that as used herein and in the appended claims, the singular forms "a,"
"an," and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "the protein" includes reference to one or more proteins, and so forth. It is further noted that the claims may be drafted to exclude any optional element. As such, this statement is intended to serve as antecedent basis for use of such exclusive terminology as "solely," "only" and the like in connection with the recitation of claim elements, or use of a "negative" limitation.
[0024] The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the present invention is not entitled to antedate such publication by virtue of prior invention. Further, the dates of publication provided may be different from the actual publication dates which may need to be independently confirmed.
DEFINITIONS
[0025] The terms "patient" or "subject" as used interchangeably herein in the context of therapy, refer to a human and non-human animal, as the recipient of a therapy or preventive care.
[0026] The phrase "in a sufficient amount to effect a change in" means that there is a detectable difference between a level of an indicator measured before and after administration of a particular therapy. Indicators include, but are not limited to, Gremlin-2 circulating levels, Gremlin-2 tissue levels, Gremlin-2 activity, muscle function, and muscle mass.
[0027] "Gremlin-2" (also known as "cysteine knot superfamily 1 bone morphogenic protein
antagonist" (CKTSF1B2); "differential-screening-selected gene Aberrative in
Neuroblastoma- 1 domain family member 3" (DAND3); "protein related to DAN and Cerberus" (PRDC); FLJ21195; and DCR5) encompasses murine and human proteins that are encoded by a GREM2 gene or a gene homologue of GREM2. Gremlin-2 is found in many mammals (e.g. human, non-human primates, and mouse). See Figure 15 for an alignment of amino acid sequences of various Gremlin-2 polypeptides.
[0028] As used herein, "homologues" or "variants" refers to protein or DNA sequences that are similar based on their amino acid or nucleic acid sequences, respectively. Homologues or variants encompass naturally occurring DNA sequences and proteins encoded thereby and their isoforms. The homologues also include known allelic or splice variants of a protein/gene. Homologues and variants also encompass nucleic acid sequences that vary in one or more bases from a naturally-occurring DNA sequence but still translate into an amino acid sequence that correspond to the naturally-occurring protein due to degeneracy of the genetic code. Homologues and variants may also refer to those that differ from the naturally-occurring sequences by one or more conservative substitutions and/or tags and/or conjugates.
[0029] The terms "polypeptide," "peptide," and "protein," used interchangeably herein, refer to a polymeric form of amino acids of any length, which can include genetically coded and non-genetically coded amino acids, chemically or biochemically modified or derivatized amino acids, and polypeptides having modified peptide backbones. The term includes fusion proteins, including, but not limited to, fusion proteins with a heterologous amino acid sequence, fusions with heterologous and homologous leader sequences, with or without N-terminal methionine residues; immunologically tagged proteins; and the like.
[0030] It will be appreciated that throughout this present disclosure reference is made to amino acids according to the single letter or three letter codes. For the reader's convenience, the single and three letter amino acid codes are provided below:
G Glycine Gly P Proline Pro
A Alanine Ala V Valine Val
L Leucine Leu I Isoleucine He
M Methionine Met c Cysteine Cys
F Phenylalanine Phe Y Tyrosine Tyr
W Tryptophan Trp H Histidine His
K Lysine Lys R Arginine Arg
Q Glutamine Gin N Asparagine Asn
E Glutamic Acid Glu D Aspartic Acid Asp
S Serine Ser T Threonine Thr
[0031] The terms "nucleic acid molecule" and "polynucleotide" are used interchangeably and refer to a polymeric form of nucleotides of any length, either deoxyribonucleotides or ribonucleotides, or analogs thereof. Non-limiting examples of polynucleotides include linear and circular nucleic acids, messenger RNA (mRNA), cDNA, recombinant polynucleotides, vectors, probes, and primers.
[0032] The term "heterologous" refers to two components that are defined by structures derived from different sources. For example, where "heterologous" is used in the context of a polypeptide, where the polypeptide includes operably linked amino acid sequences that can be derived from different polypeptides (e.g., a first component consisting of a recombinant peptide and a second component derived from a native Gremlin-2 polypeptide). Similarly, "heterologous" in the context of a polynucleotide encoding a chimeric polypeptide includes operably linked nucleic acid sequence that can be derived from different genes (e.g., a first component from a nucleic acid encoding a peptide according to an
embodiment disclosed herein and a second component from a nucleic acid encoding a carrier polypeptide). Other exemplary "heterologous" nucleic acids include expression constructs in which a nucleic acid comprising a coding sequence is operably linked to a regulatory element (e.g., a promoter) that is from a genetic origin different from that of the coding sequence (e.g., to provide for expression in a host cell of interest, which may be of different genetic origin relative to the promoter, the coding sequence or both). For example, a T7 promoter operably linked to a polynucleotide encoding a Gremlin-2 polypeptide or domain thereof is said to be a heterologous nucleic acid. "Heterologous" in the context of recombinant cells can refer to the presence of a nucleic acid (or gene product, such as a polypeptide) that is of a different genetic origin than the host cell in which it is present.
[0033] The term "operably linked" refers to functional linkage between molecules to provide a desired function. For example, "operably linked" in the context of nucleic acids refers to a functional linkage between nucleic acids to provide a desired function such as
transcription, translation, and the like, e.g., a functional linkage between a nucleic acid expression control sequence (such as a promoter, signal sequence, or array of transcription factor binding sites) and a second polynucleotide, wherein the expression control sequence affects transcription and/or translation of the second polynucleotide. "Operably linked" in the context of a polypeptide refers to a functional linkage between amino acid sequences (e.g., of different domains) to provide for a described activity of the polypeptide. [0034] As used herein in the context of the structure of a polypeptide, "N-terminus" and "C- terminus" refer to the extreme amino and carboxyl ends of the polypeptide, respectively, while "N-terminal" and "C-terminal" refer to relative positions in the amino acid sequence of the polypeptide toward the N-terminus and the C-terminus, respectively, and can include the residues at the N-terminus and C-terminus, respectively. "Immediately N- terminal" or "immediately C-terminal" refers to a position of a first amino acid residue relative to a second amino acid residue where the first and second amino acid residues are covalently bound to provide a contiguous amino acid sequence.
[0035] "Derived from" in the context of an amino acid sequence or polynucleotide sequence (e.g., an amino acid sequence "derived from" a Gremlin-2 polypeptide) is meant to indicate that the polypeptide or nucleic acid has a sequence that is based on that of a reference polypeptide or nucleic acid (e.g., a naturally occurring Gremlin-2 polypeptide or Gremlin- 2-encoding nucleic acid), and is not meant to be limiting as to the source or method in which the protein or nucleic acid is made.
[0036] "Isolated" refers to a protein of interest (e.g., a Gremlin-2 polypeptide; an anti-Gremlin-2 antibody) that, if naturally occurring, is in an environment different from that in which it may naturally occur. "Isolated" is meant to include proteins that are within samples that are substantially enriched for the protein of interest and/or in which the protein of interest is partially or substantially purified. Where the protein is not naturally occurring, "isolated" indicates the protein has been separated from an environment in which it was made by either synthetic or recombinant means.
[0037] "Enriched" means that a sample is non-naturally manipulated (e.g., by an experimentalist or a clinician) so that a protein of interest (e.g., a Gremlin-2 polypeptide; an anti-Gremlin-2 antibody) is present in a greater concentration (e.g., at least a three-fold greater, at least 4- fold greater, at least 8-fold greater, at least 64-fold greater, or more) than the concentration of the protein in the starting sample, such as a biological sample (e.g., a sample in which the protein naturally occurs or in which it is present after administration), or in which the protein was made (e.g., as in a bacterial protein and the like).
[0038] "Substantially pure" indicates that an entity (e.g., a Gremlin-2 polypeptide; an anti- Gremlin-2 antibody) makes up greater than about 50% of the total content of the composition (e.g., total protein of the composition), or greater than about 60% of the total protein content. For example, a "substantially pure" refers to compositions in which at least 75%, at least 85%, at least 90% or more of the total composition is the entity of interest (e.g. 95%, 98%, 99%, greater than 99%), of the total protein. The protein can make up greater than about 90%, or greater than about 95% of the total protein in the
composition.
[0039] The term "antibody" (also used interchangeably with "immunoglobulin") encompasses polyclonal and monoclonal antibody preparations where the antibody may be of any class of interest (e.g., IgM, IgG, and subclasses thereof), as well as preparations including hybrid antibodies, altered antibodies, F(ab')2 fragments, F(ab) molecules, Fv fragments, single chain Fv fragments (scFv), single chain antibodies, single domain antibodies (VRH), chimeric antibodies, humanized antibodies, and antigen-binding fragments thereof which exhibit immunological binding properties of the parent antibody molecule. Antibodies that have inhibitory functions for their targets are of particular interest. The antibodies described herein may be detectably labeled, e.g., with a radioisotope, an enzyme which generates a detectable product, a fluorescent protein, and the like. The antibodies may be further conjugated to other moieties, such as members of specific binding pairs, e.g., biotin (member of biotin-avidin specific binding pair), and the like. The antibodies may also be bound to a support (e.g., a solid support), such as a polystyrene plate or bead, test strip, and the like.
[0040] The term "binding" refers to a direct association between two molecules, due to, for
example, covalent, electrostatic, hydrophobic, and ionic and/or hydrogen-bond
interactions, including interactions such as salt bridges and water bridges. A subject anti- Gremlin-2 antibody binds specifically to an epitope within a Gremlin-2 polypeptide, e.g., with an affinity of at least about 10"7 M, at least about 10~8 M, at least about 10"9 M, at least about 10"10 M, at least about 10"11 M, or at least about 10"12 M, or greater than 10"12 M. A subject antibody binds to an epitope present on a Gremlin-2 polypeptide with an affinity of from about 10"7 M to about 10~8 M, from about 10"8 M to about 10"9 M, from about 10"9 M to about 10"10 M, from about 10"10 M to about 10"11 M, or from about 10"11 M to about 10"12
-12
M, or greater than 10" M.. Non-specific binding would refer to binding with an affinity of less than about 10"7 M, e.g., binding with an affinity of 10"6 M, 10"5 M, 10"4 M, etc.
[0041] Immunoglobulin polypeptides include the kappa and lambda light chains and the alpha, gamma (IgG1 ; IgG2, IgG3, IgG4), delta, epsilon and mu heavy chains or equivalents in other species. Full-length immunoglobulin "light chains" comprise a variable region at the NH2-terminus and a kappa or lambda constant region at the COOH-terminus. Full-length immunoglobulin "heavy chains" similarly comprise a variable region and one of the aforementioned heavy chain constant regions, e.g., gamma.
[0042] An immunoglobulin light or heavy chain variable region is composed of a "framework" region (FR) interrupted by three hypervariable regions, also called "complementarity determining regions" or "CDRs". CDRs have been described by Kabat et al., J. Biol.
Chem. 252:6609-6616 (1977); Kabat et al., U.S. Dept. of Health and Human Services, "Sequences of proteins of immunological interest" (1991); by Chothia et al., J. Mol. Biol. 196:901-917 (1987); and MacCallum et al., J. Mol. Biol. 262:732-745 (1996). Methods to define CDRs are available in the art and routinely preformed. For example, framework regions and CDRs may be defined by IMGT (see, "Sequences of Proteins of
Immunological Interest," E. Kabat et al., U.S. Department of Health and Human Services, (1991 and Lefranc et al. IMGT, the international ImMunoGeneTics information system®. Nucl. Acids Res., 2005, 33, D593-D597)). A detailed discussion of the IMGTS system, including how the IMGTS system was formulated and how it compares to other systems, is provided on the World Wide Web at imgt.cines.fr/ textes/ IMGTS cientificChart/
Numbering/ IMGTnumberingsTable.html. The framework region of an antibody, that is the combined framework regions of the constituent light and heavy chains, serves to position and align the CDRs. The CDRs are primarily responsible for binding to an epitope of an antigen.
[0043] The term "monoclonal antibody" refers to an antibody composition having a homogeneous antibody population. The term is not limited by the manner in which it is made. The term encompasses whole immunoglobulin molecules, as well as Fab molecules, F(ab')2 fragments, Fv fragments, scFv, fusion proteins comprising an antigen-binding portion of an antibody and a non-antibody protein, and other molecules that exhibit immunological binding properties of the parent monoclonal antibody molecule. Methods of making polyclonal and monoclonal antibodies are known in the art.
[0044] "Antibody fragments" comprise a portion of an intact antibody, for example, the antigen binding or variable region of the intact antibody. Examples of antibody fragments include Fab, Fab', F(ab')2, and Fv fragments; diabodies; linear antibodies (Zapata et al., Protein Eng. 8(10): 1057-1062 (1995)); single-chain antibody molecules; and multispecific antibodies formed from antibody fragments. Papain digestion of antibodies produces two identical antigen-binding fragments, called "Fab" fragments, each with a single antigen- binding site, and a residual "Fc" fragment, a designation reflecting the ability to crystallize readily. Pepsin treatment yields an F(ab')2 fragment that has two antigen combining sites and is still capable of cross-linking antigen.
[0045] "Fv" is the minimum antibody fragment which contains a complete antigen-recognition and -binding site. This region consists of a dimer of one heavy- and one light-chain variable domain in tight, non-covalent association. It is in this configuration that the three CDRS of each variable domain interact to define an antigen-binding site on the surface of the VR-VL dimer. Collectively, the six CDRs confer antigen-binding specificity to the antibody. However, even a single variable domain (or half of an Fv comprising only three CDRs specific for an antigen) has the ability to recognize and bind antigen, although at a lower affinity than the entire binding site.
[0046] The "Fab" fragment also contains the constant domain of the light chain and the first
constant domain (CH of the heavy chain. Fab fragments differ from Fab' fragments by the addition of a few residues at the carboxyl terminus of the heavy chain CHi domain including one or more cysteines from the antibody hinge region. Fab'-SH is the designation herein for Fab' in which the cysteine residue(s) of the constant domains bear a free thiol group. F(ab')2 antibody fragments originally were produced as pairs of Fab' fragments which have hinge cysteines between them. Other chemical couplings of antibody fragments are also known.
[0047] "Single-chain Fv" or "sFv" antibody fragments comprise the VH and VL domains of
antibody, wherein these domains are present in a single polypeptide chain. In some embodiments, the Fv polypeptide further comprises a polypeptide linker between the VH and VL domains, which enables the sFv to form the desired structure for antigen binding. For a review of sFv, see Pluckthun in The Pharmacology of Monoclonal Antibodies, vol. 113, Rosenburg and Moore eds., Springer-Verlag, New York, pp. 269-315 (1994).
[0048] The term "diabodies" refers to small antibody fragments with two antigen-binding sites, which fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) in the same polypeptide chain (VR-VL). By using a linker that is too short to allow pairing between the two domains on the same chain, the domains are forced to pair with the complementary domains of another chain and create two antigen-binding sites. Diabodies are described more fully in, for example, EP 404,097; WO 93/11161; and Hollinger et al, Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993).
DETAILED DESCRIPTION
OVERVIEW
[0049] The present disclosure provides compositions and methods for reducing levels and/or activity of Gremlin-2. The present disclosure provides compositions and methods for increasing muscle function and/or muscle mass. The present methods involve using an agent that targets a Gremlin-2 polypeptide. Subject compositions and methods are useful for treating various conditions and disorders characterized by loss of muscle function and/or muscle mass.
[0050] The proteins targeted by the methods and compositions of the present disclosure
encompass Gremlin-2, Gremlin-2 genes and/or proteins encoded thereby, and are useful for treating individuals having a deficiency in muscle function and/or having reduced muscle mass, e.g., for treating disorders, diseases, and conditions in which reduced muscle function and/or mass is a result, a sequela, or a symptom of the disorder, disease, or condition. When Gremlin-2 protein was administered (as exemplified by expression from an AAV) to wild-type mice, loss of muscle mass and reduced grip strength were observed. In the mdx mouse model of Duchenne muscular dystrophy, mice showed elevated muscle levels of Gremlin-2 mRNA, and elevated muscle and serum levels of Gremlin-2 protein. Accordingly, targeting a Gremlin-2 protein, to reduce circulating and/or tissue levels of Gremlin-2 and/or to reduce Gremlin-2 activity, can be used to increase muscle function and or muscle mass in an individual. Targeting a Gremlin-2 protein can be used to treat disorders, diseases, and conditions in which reduced muscle function (e.g., muscle weakness) and/or reduced muscle mass is a result, a sequela, or a symptom of the disorder, disease, or condition.
METHODS
[0051] The present disclosure provides methods of reducing levels and/or activity of Gremlin-2 in an individual. The present disclosure also provides methods for increasing muscle function and/or muscle mass in an individual having a deficiency in muscle function and/or having reduced muscle mass, e.g., in an individual having a condition, disease, or disorder in which reduced muscle function and/or reduced muscle mass is a result, a sequela, or a symptom of the disorder, disease, or condition. A subject method generally involves administering to an individual an effective amount of an agent that reduces Gremlin-2 levels and/or activity. Suitable agents include, e.g., an antibody that specifically binds Gremlin-2; and a short interfering nucleic acid (siNA) that specifically reduces the level of Gremlin-2.
Reducing Gremlin-2 levels and/or activity
[0052] Agents that reduce levels and/or activity of Gremlin-2 include agents that reduce Gremlin- 2 circulating and/or tissue levels of Gremlin-2; and agents that reduce one or more biological activities of Gremlin-2. Circulating levels of Gremlin-2 include levels of Gremlin-2 in the blood or any extracellular fluid.
[0053] Agents that reduce circulating and/or tissue levels of Gremlin-2 polypeptide, and that are suitable for use in a subject method, includes agents that, when administered to an individual, reduce circulating levels of Gremlin-2 polypeptide in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or more than 70%, compared to the circulating level of Gremlin-2 polypeptide in the individual not treated with the agent. In some cases, a suitable agent is one that, when administered to an individual, reduces circulating levels of Gremlin-2 polypeptide in the individual to a normal control level. Circulating levels of Gremlin-2 include serum levels. Circulating levels of Gremlin-2 polypeptide can be readily determined, using any known method, e.g., an immunological method employing anti-Gremlin-2 antibody. Suitable immunological methods include, e.g., an enzyme-linked immunosorbent assay (ELISA), a
radioimmunoassay (RIA), and the like.
[0054] Agents that reduce circulating and/or tissue levels of Gremlin-2 polypeptide, and that is suitable for use in a subject method, includes agents that, when administered to an individual, reduce tissue levels of Gremlin-2 polypeptide in the individual by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or more than 70%, compared to the tissue level of Gremlin-2 polypeptide in the individual not treated with the agent. For example, an agent suitable for use in a subject method includes an agent that reduces muscle levels (e.g., skeletal muscle levels) of Gremlin-2 polypeptide in an individual. In some cases, a suitable agent is one that, when administered to an individual, reduces tissue levels of Gremlin-2 polypeptide in the individual to a normal control level.
[0055] Agents that reduce one or more Gremlin-2 activities in an individual include, e.g., agents that reduce (e.g., inhibit) binding of Gremlin-2 to a Gremlin-2 binding partner, e.g., a Gremlin-2 binding partner present in a muscle cell, a Gremlin-2 binding partner present in a cell membrane, or an extracellular Gremlin-2 binding partner. For example, an agent suitable for use in a subject method can include an agent that inhibits binding of a
Gremlin-2 polypeptide to a Gremlin-2 binding partner by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or more than 70%, compared to the binding of the Gremlin-2 polypeptide to the Gremlin-2 binding partner in the absence of the agent.
Increasing muscle function and/or muscle mass
[0056] Reducing Gremlin-2 levels and/or activity can provide for increasing muscle function
and/or muscle mass in an individual, and can be used to treat a disease, disorder, or condition resulting in or associated with reduced muscle function and/or muscle mass. As such, the present disclosure provides methods for increasing muscle function and/or muscle mass in an individual in need thereof, e.g., an individual having a deficiency in muscle function and/or reduced muscle mass. "Increasing muscle mass" includes: a) an increase in muscle mass that results from generation of new muscle tissue; and b) an increase in muscle mass that results from repair of existing muscle tissue that has been damaged (e.g., due to disease or injury).
[0057] In some cases, a subject method involves administering an anti-Gremlin-2 antibody to a subject who has a disease, disorder, or condition resulting in or associated with reduced muscle function and/or reduced muscle mass (e.g., a disease, disorder, or condition in which reduced muscle function and/or reduced muscle mass is a result, a sequela, or a symptom of the disorder, disease, or condition). The methods of the present disclosure can comprise administering one or more anti-Gremlin-2 antibodies in the context of a variety of diseases, disorders, and conditions resulting in or associated with reduced muscle function and/or reduced muscle mass.
[0058] In some cases, a subject method involves administering a short interfering nucleic acid (siNA) specific for Gremlin-2 to a subject who has a disease, disorder, or condition resulting in or associated with reduced muscle function and/or muscle mass (e.g., a disease, disorder, or condition in which reduced muscle function and/or reduced muscle mass is a result, a sequela, or a symptom of the disorder, disease, or condition). The methods of the present disclosure can include administering one or more Gremlin-2-specific siNA in the context of a variety of diseases, disorders, and conditions resulting in or associated with reduced muscle function and/or muscle mass.
[0059] Subjects having, suspected of having, or at risk of developing a disease, disorder, or
condition resulting in or associated with reduced muscle function and/or muscle mass are contemplated for therapy and diagnosis described herein.
[0060] By "treatment" is meant that at least an amelioration of the symptoms associated with the condition afflicting the host is achieved, where amelioration refers to at least a reduction in the magnitude of a parameter, e.g. symptom, associated with the condition being treated. As such, treatment includes situations where the condition, or at least symptoms associated therewith, are reduced or avoided. Thus treatment includes: (i) prevention, that is, reducing the risk of development of clinical symptoms, including causing the clinical symptoms not to develop, e.g., preventing disease progression to a harmful or otherwise undesired state; (ii) inhibition, that is, arresting the development or further development of clinical symptoms, e.g., mitigating or completely inhibiting an active disease (e.g., so as to increase muscle function and/or muscle mass).
[0061] In the methods of the present disclosure, antibody compositions or siNA compositions described herein can be administered to a subject (e.g. a human patient) to, for example, increase muscle function to a range found in a healthy individual. Subjects for treatment include those having a disease, disorder, or condition resulting in or associated with reduced muscle function and/or mass, as described herein.
[0062] In some embodiments, an effective amount of an anti-Gremlin-2 antibody or a Gremlins- specific siNA is an amount that is effective to reduce muscle atrophy, e.g., an effective amount of an anti-Gremlin-2 antibody or a Gremlin-2-specific siNA reduces muscle atrophy by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, or at least about 80%, or more than 80%, compared to the degree of atrophy in the absence of treatment with the anti-Gremlin-2 antibody or the Gremlin-2- specific siNA. [0063] In some embodiments, an effective amount of an anti-Gremlin-2 antibody or a Gremlins- specific siNA is an amount that is effective to increase muscle mass (e.g., skeletal muscle mass), e.g., an effective amount of an anti-Gremlin-2 antibody or a Gremlin-2- specific siNA increases muscle mass by at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, or at least about 5-fold, or more than 5-fold, compared to the muscle mass in the absence of treatment with the anti- Gremlin-2 antibody or the Gremlin-2- specific siNA. As noted above, "increasing muscle mass" includes: a) an increase in muscle mass that results from generation of new muscle tissue; and b) an increase in muscle mass that results from repair of existing muscle tissue that has been damaged (e.g., due to disease or injury).
[0064] Whether atrophy is reduced, and whether muscle mass is increased, can be determined using any known method, including, e.g., magnetic resonance imaging (MRI), dual energy x-ray absorptiometry (DEXA), and computed tomography (CT).
[0065] As noted above, a method of the present disclosure can provide for improved muscle
function, where muscle function includes, e.g., muscle endurance, muscle strength, muscle force, muscle fatigability, etc. "Improved" muscle function includes increased muscle endurance, increased muscle strength, increased muscle force, and decreased muscle fatigability. Thus, for example, in some embodiments, treatment with an agent that reduces Gremlin-2 levels and/or activity results in an increase in one or more of muscle endurance, muscle strength, and muscle force of at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, or at least about 5- fold, or more than 5-fold, compared to the muscle endurance, muscle strength, or muscle force in the absence of treatment with the agent. As another example, treatment with an agent that reduces Gremlin-2 levels and/or activity results in a decrease in muscle fatigability, e.g., results in an increase in the amount of time to reach a fatigued state, such that muscle fatigability is reduced by at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or more than 80%, compared to the muscle fatigability in the absence of treatment with the agent.
[0066] In some embodiments, an effective amount of an anti-Gremlin-2 antibody is an amount that is effective to improve function (e.g., increase one or more of muscle endurance, muscle strength, muscle force, etc.; and/or decrease muscle fatigability, etc.). For example, an effective amount of an anti-Gremlin-2 antibody improves muscle function by at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 2- fold, at least about 2.5-fold, or at least about 5-fold, or more than 5-fold, compared to the muscle function in the absence of treatment with the anti-Gremlin-2 antibody. As another example, an effective amount of a Gremlin-2-specific siNA improves muscle function by at least about 10%, at least about 25%, at least about 50%, at least about 75%, at least about 2-fold, at least about 2.5-fold, or at least about 5-fold, or more than 5-fold, compared to the muscle function in the absence of treatment with the Gremlin- specific siNA.
[0067] Muscle strength can be measured using any known method, including, e.g., a grip strength test. See, e.g., Geere et al. ((2007) BMC Musculoskelet. Disord. 8: 114), and references cited therein. A field test, such as the one -repetition maximum (1-RM) test, can also be used. The 1-RM test measures dynamic strength by determining how much weight an individual can lift during a single repetition. The amount can be divided by body weight to give 1-RM/BW. Muscle strength and function can be assessed by standard performance tests such as knee flexor and extensor strength, repeated sit-to-stand test, and timed up & go (TUG). Muscle strength can be measured as knee extensor and flexor in Newtons (kiloponds). TUG is a measure of functional mobility including muscle strength, gait speed, and balance and is assessed in seconds. The repeated sit-to-stand is a functional test and measured in seconds.
[0068] Muscle force, expressed as tetanic force, can be measured using any known method.
Various types of contractions can be measured, including isotonic contraction, concentric contraction, eccentric contraction, and isometric contraction. Methods of measuring muscle contraction are known in the art, and any such method can be used to measure muscle contraction. Suitable methods include, e.g., mechanomyography, ultrasound myography, acoustic myography, electromyography, and the like.
[0069] Muscle fatigability and muscle endurance can be measured in humans using a treadmill test, e.g., where the treadmill is inclined or is horizontal. Muscle fatigability and muscle endurance can be measured in rodents (e.g., mice, rats, etc.) using a rotarod test or a wire hang test. For example, in the rotarod test, mice (or rats) are placed on an elevated accelerating rod and the rod is rotated at a certain speed (e.g., 4 rotations per minute (rpm) to 40 rpm). The rodents are then scored for their latency (e.g., in seconds) to fall. A reduction in the time to fall is an indication of an increase in muscle endurance or a reduction in muscle fatigability.
ANTI-G EMLIN-2 ANTIBODY
[0070] Methods and compositions are provided herein to reduce levels and/or activity of Gremlin- 2 and/or to increase muscle function and/or muscle mass in a subject. The subject methods and/or compositions target Gremlin-2 so as to decrease the amount and/or the activity of the mature Gremlin-2 protein.
[0071] The present disclosure provides methods for reducing (e.g., inhibiting, or neutralizing) Gremlin-2 activity in a subject having a disease, disorder, or condition that is associated with or that results in reduced muscle function and/or mass. One way Gremlin-2 can be inhibited is to administer anti-Gremlin-2 antibodies (also referred to as "anti-Gremlin-2 antibodies"), where such antibodies include a whole antibody (e.g. IgG), an antigen- binding fragment thereof, single-chain Fab, or a synthetic anti-Gremlin-2 antibody that comprise portions of an antibody. Gremlin-2 is the target of such antibodies. Details on Gremlin-2 are provided below.
[0072] Administration of Gremlin-2 to wild- type mice leads to reduced muscle mass and reduced muscle function. In addition, elevated muscle levels of Gremlin-2 mRNA, and elevated muscle and serum levels of Gremlin-2 protein, were observed in the mdx mouse model of Duchenne muscular dystrophy. Thus, Gremlin-2 may exacerbate symptoms of a disease, disorder, or condition associated with reduced muscle function and/or mass. Accordingly, decreasing the amount and/or activity of Gremlin-2 can serve to treat such diseases, disorders, and conditions.
[0073] Targeting Gremlin-2 as provided by the present methods can encompass inhibiting other biomolecules ("binding partners") that normally interact with Gremlin-2 from binding to Gremlin-2 or sequestering Gremlin-2 from its signaling partners. For example, a subject anti-Gremlin-2 antibody can decrease Gremlin-2 activity. As another example, a subject anti-Gremlin-2 antibody can decrease the ability of Gremlin-2 to bind to a binding partner for Gremlin-2, e.g., a naturally- occurring cellular binding partner for Gremlin-2. The binding of a subject anti-Gremlin-2 antibody may be competitive or non-competitive with the endogenous binding partners of Gremlin-2. A subject anti-Gremlin-2 antibody may also modify the activity and/or structure of Gremlin-2. The methods can further encompass decreasing the level of Gremlin-2 expression and/or amount of mature, active Gremlin-2. The method can also increase turnover of Gremlin-2.
[0074] In some cases, a subject anti-Gremlin-2 antibody can reduce binding of a Gremlin-2
polypeptide to a binding partner for Gremlin-2, e.g., a naturally-occurring cellular binding partner for Gremlin-2. For example, in some embodiments, a subject antibody can reduce binding of a Gremlin-2 polypeptide to a Gremlin-2 binding partner by at least about 5%, at least about 10%, at least about 15%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, or more, compared to the degree of binding between the Gremlin-2 polypeptide and the binding partner in the absence of the antibody.
[0075] In certain embodiments, an antibody comprising: a) a variable domain comprising: i. a
CDR1 region that is identical in amino acid sequence to the heavy chain CDR1 region of an anti-Gremlin-2 antibody; ii. a CDR2 region that is identical in amino acid sequence to the heavy chain CDR2 region of the anti-Gremlin-2 antibody; and iii. a CDR3 region that is identical in amino acid sequence to the heavy chain CDR3 region of the anti-Gremlin-2 antibody; and b) a light chain variable domain comprising: i. a CDR1 region that is identical in amino acid sequence to the light chain CDR1 region of the anti-Gremlin-2 antibody; ii. a CDR2 region that is identical in amino acid sequence to the light chain CDR2 region of the anti-Gremlin-2 antibody; and iii. a CDR3 region that is identical in amino acid sequence to the light chain CDR3 region of the anti-Gremlin-2 antibody; or b) a variant of the variable domain of part a) that is otherwise identical to the variable domain of part a) except for a number of (e.g., 1, 2, 3, 4, 5, 6, 7 or 8) amino acid substitutions in the CDR regions, where the antibody binds a Gremlin-2 polypeptide.
[0076] In some embodiments, a subject antibody comprises: a) a light chain region comprising: i) one, two, or three complementarity determining regions (CDRs) from a mouse monoclonal anti-Gremlin-2 antibody light chain variable region sequence; and ii) a light chain framework region, e.g., a framework region from a human immunoglobulin light chain; and b) a heavy chain region comprising: i) one, two, or three CDRs from the mouse monoclonal anti-Gremlin-2 antibody heavy chain variable region sequence; and ii) a heavy chain framework region, e.g., a framework region from a human immunoglobulin heavy chain. [0077] A subject anti-Gremlin-2 antibody can find use in a variety of applications, including use in various methods of treating a host suffering from a disease, disorder, or condition associated with reduced muscle function and/or mass, as well as in diagnosis of various diseases and conditions associated with Gremlin-2 expression.
Recombinant antibody
[0078] A subject anti-Gremlin-2 antibody may be recombinant. The antibody may contain a light and/or heavy chain. Methods for producing recombinant antibodies are known in the art. For example, the nucleic acids encoding the antibody, or at least a complementary determining region (CDR) of a heavy chain polypeptide or at least a CDR of a light chain polypeptide, are introduced directly into a host cell, and the cell incubated under conditions sufficient to induce expression of the encoded antibody. The recombinant antibody may be glycosylated by an endogenous glycosylase in the host cells, unglycosylated, or may have an altered glycosylation pattern.
[0079] Where the antibody is recombinant, the antibody may be chimeric. Chimeric antibodies are immunoglobulin molecules comprising human and non-human portions. More specifically, the antigen combining region (or variable region) of a humanized chimeric antibody is derived from a non-human source (e.g. murine), and the constant region of the chimeric antibody (which confers biological effector function to the immunoglobulin) is derived from a human source. The chimeric antibody can have the antigen binding specificity of the non-human antibody molecule and the effector function conferred by the human antibody molecule. A large number of methods of generating chimeric antibodies are well known to those of skill in the art. An alternative approach is the generation of humanized antibodies by linking the CDR regions of non-human antibodies to human constant regions by recombinant DNA techniques.
[0080] A recombinant fusion antibody that is specific for a Gremlin-2 polypeptide is
contemplated, in which the antibody is modified to include a heterologous protein. For example, a heavy chain polypeptide and/or light chain polypeptide may be joined to a reporter protein or to a protein having a desired therapeutic effect. The reporter protein may be a fluorescent protein. The antibody may also be conjugated to a second antibody (or at least an antigen-binding portion thereof). Methods for producing a fusion protein of interest when provided a nucleic acid sequence are well known in the art. Humanized and human antibodies
[0081] A subject anti-Gremlin-2 antibody will in some embodiments be humanized. Amino acids may be substituted in the framework regions of a parent non-human (e.g., mouse monoclonal) antibody to produce a modified antibody that is less immunogenic in a human than the parent non-human antibody. Antibodies can be humanized using a variety of techniques known in the art. Framework substitutions are identified by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions.
[0082] The antibody may also be a fully human antibody. Human antibodies are primarily
composed of characteristically human polypeptide sequences. A subject human antibody can be produced by a wide variety of methods. For example, human antibodies can be produced initially in trioma cells (descended from three cells, two human and one mouse). Genes encoding the antibodies are then cloned and expressed in other cells, particularly non-human mammalian cells. The general approach for producing human antibodies by trioma technology has been described in the art.
[0083] Accordingly, the present disclosure contemplates a DNA molecule comprising a nucleic acid sequence encoding an antibody that binds to a Gremlin-2 polypeptide. The disclosure further contemplates recombinant host cells containing an exogenous polynucleotide encoding at least a CDR of a heavy chain polypeptide or at least a CDR of a light chain polypeptide of the subject antibody.
scFv
[0084] In some embodiments, a subject antibody comprises anti-Gremlin-2 antibody heavy chain CDRs and anti-Gremlin-2 antibody light chain CDRs in a single polypeptide chain, e.g., in some embodiments, a subject antibody is a scFv. In some embodiments, a subject antibody comprises, in order from N-terminus to C-terminus: a first amino acid sequence of from about 5 amino acids to about 25 amino acids in length; a heavy chain CDR1 of an anti- Gremlin-2 antibody; a second amino acid sequence of from about 5 amino acids to about 25 amino acids in length; a heavy chain CDR2 of an anti-Gremlin-2 antibody; a third amino acid sequence of from about 5 amino acids to about 25 amino acids in length; a heavy chain CDR3 of an anti-Gremlin-2 antibody; a fourth amino acid sequence of from about 5 amino acids to about 25 amino acids in length; a light chain CDR1 of an anti- Gremlin-2 antibody; a fifth amino acid sequence of from about 5 amino acids to about 25 amino acids in length; a light chain CDR2 an anti-Gremlin-2 antibody; a sixth amino acid sequence of from about 5 amino acids to about 25 amino acids in length; a light chain CDR3 an anti-Gremlin-2 antibody; and a seventh amino acid sequence of from about 5 amino acids to about 25 amino acids in length.
[0085] In some embodiments, a subject anti-Gremlin-2 antibody comprises scFv multimers. For example, in some embodiments, a subject antibody is an scFv dimer (e.g., comprises two tandem scFv (scFv2)), an scFv trimer (e.g., comprises three tandem scFv (scFv3)), an scFv tetramer (e.g., comprises four tandem scFv (scFv4)), or is a multimer of more than four scFv (e.g., in tandem). The scFv monomers can be linked in tandem via linkers of from about 2 amino acids to about 10 amino acids in length, e.g., 2 aa, 3 aa, 4 aa, 5 aa, 6 aa, 7 aa, 8 aa, 9 aa, or 10 aa in length. Suitable linkers include, e.g., (Gly)x, where x is an integer from 2 to 10. Other suitable linkers are those discussed above. In some embodiments, each of the scFv monomers in a subject scFV multimer is humanized, as described above.
Antibody modifications
[0086] A subject anti-Gremlin-2 antibody can comprises one or more modifications.
[0087] In some embodiments, a subject antibody comprises a free thiol (-SH) group at the
carboxyl terminus, where the free thiol group can be used to attach the antibody to a second polypeptide (e.g., another antibody, including a subject antibody), a scaffold, a carrier, etc.
[0088] In some embodiments, a subject antibody comprises one or more non-naturally occurring amino acids. In some embodiments, the non-naturally encoded amino acid comprises a carbonyl group, an acetyl group, an aminooxy group, a hydrazine group, a hydrazide group, a semicarbazide group, an azide group, or an alkyne group. See, e.g., U.S. Patent No. 7,632,924 for suitable non-naturally occurring amino acids. Inclusion of a non- naturally occurring amino acid can provide for linkage to a polymer, a second polypeptide, a scaffold, etc. For example, a subject antibody linked to a water-soluble polymer can be made by reacting a water-soluble polymer (e.g., poly(ethylene glycol) (PEG)) that comprises a carbonyl group to an the subject antibody that comprises a non-naturally encoded amino acid that comprises an aminooxy, hydrazine, hydrazide or semicarbazide group. As another example, a subject antibody linked to a water-soluble polymer can be made by reacting a subject antibody that comprises an alkyne-containing amino acid with a water-soluble polymer (e.g., PEG) that comprises an azide moiety; in some embodiments, the azide or alkyne group is linked to the PEG molecule through an amide linkage. A "non- naturally encoded amino acid" refers to an amino acid that is not one of the 20 common amino acids or pyrolysine or selenocysteine. Other terms that may be used synonymously with the term "non-naturally encoded amino acid" are "non-natural amino acid," "unnatural amino acid," "non-naturally-occurring amino acid," and variously hyphenated and non- hyphenated versions thereof. The term "non-naturally encoded amino acid" also includes, but is not limited to, amino acids that occur by modification (e.g. post-translational modifications) of a naturally encoded amino acid (including but not limited to, the 20 common amino acids or pyrolysine and selenocysteine) but are not themselves naturally incorporated into a growing polypeptide chain by the translation complex. Examples of such non-naturally-occurring amino acids include, but are not limited to, N- acetylglucosaminyl-L-serine, N-acetylglucosaminyl-L-threonine, and O-phosphotyrosine. In some embodiments, a subject antibody is linked (e.g., covalently linked) to a polymer (e.g., a polymer other than a polypeptide). Suitable polymers include, e.g., biocompatible polymers, and water-soluble biocompatible polymers. Suitable polymers include synthetic polymers and naturally-occurring polymers. Suitable polymers include, e.g., substituted or unsubstituted straight or branched chain polyalkylene, polyalkenylene or polyoxyalkylene polymers or branched or unbranched polysaccharides, e.g. a homo- or hetero- polysaccharide. Suitable polymers include, e.g., ethylene vinyl alcohol copolymer
(commonly known by the generic name EVOH or by the trade name EVAL);
polybutylmethacrylate; poly(hydroxyvalerate); poly(L-lactic acid); polycaprolactone; poly(lactide-co-glycolide); poly(hydroxybutyrate); poly(hydroxybutyrate-co-valerate); polydioxanone; polyorthoester; polyanhydride; poly(glycolic acid); poly(D,L-lactic acid); poly(glycolic acid-co-trimethylene carbonate); polyphosphoester; polyphosphoester urethane; poly(amino acids); cyanoacrylates; poly(trimethylene carbonate);
poly(iminocarbonate); copoly(ether-esters) (e.g., poly(ethylene oxide)-poly(lactic acid) (PEO/PLA) co-polymers); polyalkylene oxalates; polyphosphazenes; biomolecules, such as fibrin, fibrinogen, cellulose, starch, collagen and hyaluronic acid; polyurethanes;
silicones; polyesters; polyolefins; polyisobutylene and ethylene- alphaolef in copolymers; acrylic polymers and copolymers; vinyl halide polymers and copolymers, such as polyvinyl chloride; polyvinyl ethers, such as polyvinyl methyl ether; polyvinylidene halides, such as polyvinylidene fluoride and polyvinylidene chloride; polyacrylonitrile; polyvinyl ketones; polyvinyl aromatics, such as polystyrene; polyvinyl esters, such as polyvinyl acetate; copolymers of vinyl monomers with each other and olefins, such as ethylene-methyl methacrylate copolymers, acrylonitrile-styrene copolymers, ABS resins, and ethylene-vinyl acetate copolymers; polyamides, such as Nylon 66 and
polycaprolactam; alkyd resins; polycarbonates; polyoxymethylenes; polyimides;
polyethers; epoxy resins; polyurethanes; rayon; rayon-triacetate; cellulose; cellulose acetate; cellulose butyrate; cellulose acetate butyrate; cellophane; cellulose nitrate;
cellulose propionate; cellulose ethers; amorphous Teflon; poly(ethylene glycol); and carboxymethyl cellulose.
[0090] Suitable synthetic polymers include unsubstituted and substituted straight or branched chain poly(ethyleneglycol), poly(propyleneglycol) poly(vinylalcohol), and derivatives thereof, e.g., substituted poly(ethyleneglycol) such as methoxypoly(ethyleneglycol), and derivatives thereof. Suitable naturally-occurring polymers include, e.g., albumin, amylose, dextran, glycogen, and derivatives thereof.
[0091] Suitable polymers can have an average molecular weight in a range of from 500 Da to
50000 Da, e.g., from 5000 Da to 40000 Da, or from 25000 to 40000 Da. For example, in some embodiments, where a subject antibody comprises a poly(ethylene glycol) (PEG) or methoxypoly(ethyleneglycol) polymer, the PEG or methoxypoly(ethyleneglycol) polymer can have a molecular weight in a range of from about 0.5 kiloDaltons (kDa) to 1 kDa, from about 1 kDa to 5 kDa, from 5 kDa to 10 kDa, from 10 kDa to 25 kDa, from 25 kDa to 40 kDa, or from 40 kDa to 60 kDa.
[0092] As noted above, in some embodiments, a subject antibody is covalently linked to a PEG polymer. In some embodiments, a subject scFv multimer is covalently linked to a PEG polymer. See, e.g., Albrecht et al. (2006) J. Immunol. Methods 310: 100. Methods and reagents suitable for PEGylation of a protein are well known in the art and may be found in, e.g., U.S. Pat. No. 5,849,860. PEG suitable for conjugation to a protein is generally soluble in water at room temperature, and has the general formula R(0-CH2-CH2)nO-R, where R is hydrogen or a protective group such as an alkyl or an alkanol group, and where n is an integer from 1 to 1000. Where R is a protective group, it generally has from 1 to 8 carbons. [0093] The PEG conjugated to the subject antibody can be linear. The PEG conjugated to the subject protein may also be branched. Branched PEG derivatives such as those described in U.S. Pat. No. 5,643,575, "star-PEG's" and multi-armed PEG's such as those described in Shearwater Polymers, Inc. catalog "Polyethylene Glycol Derivatives 1997-1998." Star PEGs are described in the art including, e.g., in U.S. Patent No. 6,046,305.
[0094] A subject antibody can be glycosylated, e.g., a subject antibody can comprise a covalently linked carbohydrate or polysaccharide moiety. Glycosylation of antibodies is typically either N-linked or O-linked. N-linked refers to the attachment of the carbohydrate moiety to the side chain of an asparagine residue. The tripeptide sequences asparagine-X- serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of the carbohydrate moiety to the asparagine side chain. Thus, the presence of either of these tripeptide sequences in a polypeptide creates a potential glycosylation site. O-linked glycosylation refers to the attachment of one of the sugars N-acetylgalactosamine, galactose, or xylose to a hydroxyamino acid, most commonly serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
[0095] Addition of glycosylation sites to an 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 O-linked glycosylation sites). Similarly, removal of
glycosylation sites can be accomplished by amino acid alteration within the native glycosylation sites of an antibody.
[0096] A subject antibody will in some embodiments comprise a "radiopaque" label, e.g. a label that can be easily visualized using for example x-rays. Radiopaque materials are well known to those of skill in the art. The most common radiopaque materials include iodide, bromide or barium salts. Other radiopaque materials are also known and include, but are not limited to organic bismuth derivatives (see, e.g., U.S. Pat. No. 5,939,045), radiopaque multiurethanes (see U.S. Pat. No. 5,346,981), organobismuth composites (see, e.g., U.S. Pat. No. 5,256,334), radiopaque barium multimer complexes (see, e.g., U.S. Pat. No. 4,866,132), and the like. [0097] A subject antibody can be covalently linked to a second moiety (e.g., a lipid, a polypeptide other than a subject antibody, a synthetic polymer, a carbohydrate, and the like) using for example, glutaraldehyde, a homobifunctional cross-linker, or a heterobifunctional cross- linker. Glutaraldehyde cross-links polypeptides via their amino moieties.
Homobifunctional cross-linkers (e.g., a homobifunctional imidoester, a homobifunctional N-hydroxysuccinimidyl (NHS) ester, or a homobifunctional sulfhydryl reactive cross- linker) contain two or more identical reactive moieties and can be used in a one- step reaction procedure in which the cross-linker is added to a solution containing a mixture of the polypeptides to be linked. Homobifunctional NHS ester and imido esters cross-link amine containing polypeptides. In a mild alkaline pH, imido esters react only with primary amines to form imidoamides, and overall charge of the cross-linked polypeptides is not affected. Homobifunctional sulfhydryl reactive cross-linkers includes bismaleimidhexane (BMH), l,5-difluoro-2,4-dinitrobenzene (DFDNB), and l,4-di-(3',2'-pyridyldithio) propinoamido butane (DPDPB).
[0098] Heterobifunctional cross-linkers have two or more different reactive moieties (e.g., amine reactive moiety and a sulfhydryl-reactive moiety) and are cross-linked with one of the polypeptides via the amine or sulfhydryl reactive moiety, then reacted with the other polypeptide via the non-reacted moiety. Multiple heterobifunctional haloacetyl cross- linkers are available, as are pyridyl disulfide cross -linkers. Carbodiimides are a classic example of heterobifunctional cross-linking reagents for coupling carboxyls to amines, which results in an amide bond.
[0099] A subject antibody can be immobilized on a solid support. Suitable supports are well known in the art and comprise, inter alia, commercially available column materials, polystyrene beads, latex beads, magnetic beads, colloid metal particles, glass and/or silicon chips and surfaces, nitrocellulose strips, nylon membranes, sheets, duracytes, wells of reaction trays (e.g., multi-well plates), plastic tubes, etc. A solid support can comprise any of a variety of substances, including, e.g., glass, polystyrene, polyvinyl chloride, polypropylene, polyethylene, polycarbonate, dextran, nylon, amylose, natural and modified celluloses, polyacrylamides, agaroses, and magnetite. Suitable methods for immobilizing a subject antibody onto a solid support are well known and include, but are not limited to ionic, hydrophobic, covalent interactions and the like. Solid supports can be soluble or insoluble, e.g., in aqueous solution. In some embodiments, a suitable solid support is generally insoluble in an aqueous solution.
[00100] A subject antibody will in some embodiments comprise a detectable label. Suitable detectable labels include any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means. Suitable include, but are not limited to, magnetic beads (e.g. Dynabeads™), fluorescent dyes (e.g., fluorescein isothiocyanate, texas red, rhodamine, a green fluorescent protein, a red fluorescent protein, a yellow fluorescent protein, and the like), radiolabels (e.g., 3 H, 125 I, 35 S, 14 C, or 32 P), enzymes (e.g., horse radish peroxidase, alkaline phosphatase, luciferase, and others commonly used in an enzyme-linked immunosorbent assay (ELISA)), and colorimetric labels such as colloidal gold or colored glass or plastic (e.g. polystyrene, polypropylene, latex, etc.) beads.
[00101] In some embodiments, a subject antibody comprises a contrast agent or a
radioisotope, where the contrast agent or radioisotope is one that is suitable for use in imaging, e.g., imaging procedures carried out on humans. Non-limiting examples of labels include radioisotope such as 131I (iodine), 18F (fluorine), 99Tc (technetium), U1ln (indium), and 67Ga (gallium), and contrast agent such as gadolinium (Gd), dysprosium, and iron.
Radioactive Gd isotopes ( 153 Gd) also are available and suitable for imaging procedures in non-human mammals. A subject antibody can be labeled using standard techniques. For example, a subject antibody can be iodinated using chloramine T or 1,3,4,6-tetrachloro- 3a,6a-dephenylglycouril. For fluorination, fluorine is added to a subject antibody during the synthesis by a fluoride ion displacement reaction. See, Muller-Gartner, H., TIB Tech., 16: 122-130 (1998) and Saji, H., Crit. Rev. Ther. Drug Carrier Syst, 16(2):209-244 (1999) for a review of synthesis of proteins with such radioisotopes. A subject antibody can also be labeled with a contrast agent through standard techniques. For example, a subject antibody can be labeled with Gd by conjugating low molecular Gd chelates such as Gd diethylene triamine pentaacetic acid (GdDTPA) or Gd tetraazacyclododecanetetraacetic (GdDOTA) to the antibody. See, Caravan et al., Chem. Rev. 99:2293-2352 (1999) and Lauffer et al., J. Magn. Reson. Imaging, 3: 11-16 (1985). A subject antibody can be labeled with Gd by, for example, conjugating polylysine-Gd chelates to the antibody. See, for example, Curtet et al., Invest. Radiol., 33(10):752-761 (1998). Alternatively, a subject antibody can be labeled with Gd by incubating paramagnetic polymerized liposomes that include Gd chelator lipid with avidin and biotinylated antibody. See, for example, Sipkins et al., Nature Med., 4:623-626 (1998).
[00102] Suitable fluorescent proteins that can be linked to a subject antibody include, but are not limited to, a green fluorescent protein from Aequoria victoria or a mutant or derivative thereof e.g., as described in U.S. Patent No. 6,066,476; 6,020,192; 5,985,577; 5,976,796; 5,968,750; 5,968,738; 5,958,713; 5,919,445; 5,874,304; e.g., Enhanced GFP, many such GFP which are available commercially, e.g., from Clontech, Inc.; a red fluorescent protein; a yellow fluorescent protein; any of a variety of fluorescent and colored proteins from Anthozoan species, as described in, e.g., Matz et al. (1999) Nature Biotechnol. 17:969-973; and the like.
[00103] A subject antibody will in some embodiments be linked to (e.g., covalently or non- covalently linked) a fusion partner, e.g., a ligand; an epitope tag; a peptide; a protein other than an antibody; and the like. Suitable fusion partners include peptides and polypeptides that confer enhanced stability in vivo (e.g., enhanced serum half-life); provide ease of purification, e.g., (His)n, e.g., 6His, and the like; provide for secretion of the fusion protein from a cell; provide an epitope tag, e.g., GST, hemagglutinin (HA; e.g., CYPYDVPDYA; SEQ ID NO://), FLAG (e.g., DYKDDDDK; SEQ ID NO://), c-myc (e.g.,
CEQKLISEEDL; SEQ ID NO://), and the like; provide a detectable signal, e.g., an enzyme that generates a detectable product (e.g., β-galactosidase, luciferase), or a protein that is itself detectable, e.g., a green fluorescent protein, a red fluorescent protein, a yellow fluorescent protein, etc.; provides for multimerization, e.g., a multimerization domain such as an Fc portion of an immunoglobulin; and the like.
[00104] The fusion may also include an affinity domain, including peptide sequences that can interact with a binding partner, e.g., such as one immobilized on a solid support, useful for identification or purification. Consecutive single amino acids, such as histidine, when fused to a protein, can be used for one-step purification of the fusion protein by high affinity binding to a resin column, such as nickel sepharose. Exemplary affinity domains include His5 (HHHHH) (SEQ ID NO://), HisX6 (HHHHHH) (SEQ ID NO://), C-myc (EQKLISEEDL) (SEQ ID NO://), Flag (DYKDDDDK) (SEQ ID NO://), StrepTag (WSHPQFEK) (SEQ ID NO://), hemagglutinin, e.g., HA Tag (YPYDVPDYA; SEQ ID NO://), glutathinone-S-transferase (GST), thioredoxin, cellulose binding domain, RYIRS (SEQ ID NO://), Phe-His-His-Thr (SEQ ID NO://), chitin binding domain, S-peptide, T7 peptide, SH2 domain, C-end RNA tag, WEAAAREACCRECCARA (SEQ ID NO://), metal binding domains, e.g., zinc binding domains or calcium binding domains such as those from calcium-binding proteins, e.g., calmodulin, troponin C, calcineurin B, myosin light chain, recoverin, S-modulin, visinin, VILIP, neurocalcin, hippocalcin, frequenin, caltractin, calpain large- subunit, S 100 proteins, parvalbumin, calbindin D9K, calbindin D28K, and calretinin, inteins, biotin, streptavidin, MyoD, leucine zipper sequences, and maltose binding protein.
[00105] In some embodiments, a subject antibody is modified to include a carbohydrate moiety, where the carbohydrate moiety can be covalently linked to the antibody. In some embodiments, a subject antibody is modified to include a lipid moiety, where the lipid moiety can be covalently linked to the antibody. Suitable lipid moieties include, e.g., an N- fatty acyl group such as N-lauroyl, N-oleoyl, etc.; a fatty amine such as dodecyl amine, oleoyl amine, etc.; a C3-C16 long-chain aliphatic lipid; and the like. See, e.g., U.S. Pat. No. 6,638,513). In some embodiments, a subject antibody is incorporated into a liposome.
[00106] A subject anti-Gremlin-2 antibody can be modified to include a moiety that
modifies cellular uptake relative to unconjugated material. The modified antibody may exhibit increased cellular uptake relative to unconjugated material. In alternative embodiments, the modified antibody exhibits decreased cellular uptake relative to unmodified antibody. In this aspect, the efficiency of cellular uptake can be increased or decreased by linking to peptides or proteins that facilitate endocytosis. For example, a given antibody can be linked to a ligand for a target receptor or large molecule that is more easily engulfed by endocytotic mechanisms, such as another antibody. The antibody or other ligand can then be internalized by endocytosis and the payload released by acid hydrolysis or enzymatic activity when the endocytotic vesicle fuses with lysosomes. As such, the conjugate may be one that increases endocytosis relative to unconjugated antibody. To decrease cellular uptake, the modified antibody can include a ligand that retains the antibody on the surface of a cell, which can be useful as a control for cellular uptake, or in some instances decrease uptake in one cell type while increasing it in others.
[00107] A subject anti-Gremlin-2 antibody can comprise one or more moieties, which
moieties may be linked (e.g., covalently or non-covalently linked) to the anti-Gremlin-2 antibody, either directly or via a linker, e.g. a flexible linker. For example, where a subject anti-Gremlin-2 antibody is a fusion protein comprising an anti-Gremlin-2 antibody and a heterologous fusion partner polypeptide, the heterologous fusion partner can be linked to the anti-Gremlin-2 antibody via a linker.
[00108] Linkers suitable for use in attaching a moiety to a subject anti-Gremlin-2 antibody include "flexible linkers". If present, the linker molecules are generally of sufficient length to permit the anti-Gremlin-2 antibody and a linked carrier to allow some flexible movement between the anti-Gremlin-2 antibody and the carrier. The linker molecules are generally about 6-50 atoms long. The linker molecules may also be, for example, aryl acetylene, ethylene glycol oligomers containing 2-10 monomer units, diamines, diacids, amino acids, or combinations thereof. Other linker molecules which can bind to polypeptides may be used in light of this disclosure.
[00109] Suitable linkers can be readily selected and can be of any of a suitable of different lengths, such as from 1 amino acid (e.g., Gly) to 20 amino acids, from 2 amino acids to 15 amino acids, from 3 amino acids to 12 amino acids, including 4 amino acids to 10 amino acids, 5 amino acids to 9 amino acids, 6 amino acids to 8 amino acids, or 7 amino acids to 8 amino acids, and may be 1, 2, 3, 4, 5, 6, or 7 amino acids.
[00110] Exemplary flexible linkers include glycine polymers (G)n, glycine-serine polymers
(including, for example, (GS)n, GSGGSn (SEQ ID NO: 1) and GGGSn (SEQ ID NO: 2), where n is an integer of at least one), glycine- alanine polymers, alanine- serine polymers, and other flexible linkers known in the art. Glycine and glycine-serine polymers are of interest since both of these amino acids are relatively unstructured, and therefore may serve as a neutral tether between components. Glycine polymers are of particular interest since glycine accesses significantly more phi-psi space than even alanine, and is much less restricted than residues with longer side chains (see Scheraga, Rev. Computational Chem. 11173-142 (1992)). Exemplary flexible linkers include, but are not limited GGSG (SEQ ID NO:3), GGSGG (SEQ ID NO:4), GSGSG (SEQ ID NO: 5), GSGGG (SEQ ID NO: 6), GGGSG (SEQ ID NO: 7), GSSSG (SEQ ID NO: 8), and the like. The ordinarily skilled artisan will recognize that design of a peptide conjugated to any elements described above can include linkers that are all or partially flexible, such that the linker can include a flexible linker as well as one or more portions that confer less flexible structure.
NUCLEIC ACID AGENTS
[00111] As noted above, suitable agents for reducing circulating and/or tissue levels of
Gremlin-2 include nucleic acid agents, e.g., a short interfering nucleic acid (siNA) agent that specifically targets Gremlin-2, reduces expression of Gremlin-2, and reduces the level of Gremlin-2.
[00112] Short interfering nucleic acids include small nucleic acid molecules, such as a short interfering RNA (siRNA), a double- stranded RNA (dsRNA), a micro-RNA (miRNA), and a short hairpin RNA (shRNA).
[00113] The terms "short interfering nucleic acid," "siNA," "short interfering RNA,"
"siRNA," "short interfering nucleic acid molecule," "short interfering oligonucleotide molecule," and "chemically- modified short interfering nucleic acid molecule" as used herein refer to any nucleic acid molecule capable of inhibiting or down regulating gene expression, for example by mediating RNA interference "RNAi" or gene silencing in a sequence- specific manner. Design of RNAi molecules, given a target gene, is routine in the art. See also US 2005/0282188 (which is incorporated herein by reference) as well as references cited therein. See, e.g., Pushparaj et al. Clin Exp Pharmacol Physiol. 2006 May- Jun;33(5-6):504-10; Lutzelberger et al. Handbook Exp Pharmacol. 2006;(173):243-59; Aronin et al. Gene Ther. 2006 Mar;13(6):509-16; Xie et al. Drug Discov Today. 2006 Jan;l l(l-2):67-73; Grunweller et al. Curr Med Chem. 2005;12(26):3143-61; and Pekaraik et al. Brain Res Bull. 2005 Dec 15;68(l-2): 115-20.
[00114] Methods for design and production of siNAs (including siRNAs) to a desired target are known in the art, and their application to a Gremlin-2 gene for the purposes disclosed herein will be readily apparent to the ordinarily skilled artisan, as are methods of production of siNAs having modifications (e.g., chemical modifications) to provide for, e.g., enhanced stability, bioavailability, and other properties to enhance use as therapeutics. In addition, methods for formulation and delivery of siNAs to a subject are also well known in the art. See, e.g., US 2005/0282188; US 2005/0239731; US 2005/0234232; US 2005/0176018; US 2005/0059817; US 2005/0020525; US 2004/0192626;
US 2003/0073640; US 2002/0150936; US 2002/0142980; and US2002/0120129, each of which are incorporated herein by reference.
[00115] Publicly available tools to facilitate design of siRNAs are available in the art. See, e.g., DEQOR: Design and Quality Control of RNAi (available on the internet at cluster- 1. mpi-cbg.de/Deqor/deqor.html). See also, Henschel et al. Nucleic Acids Res. 2004 Jul l;32(Web Server issue):Wl 13-20. DEQOR is a web-based program which uses a scoring system based on state-of-the-art parameters for siRNA design to evaluate the inhibitory potency of siRNAs. DEQOR, therefore, can help to predict (i) regions in a gene that show high silencing capacity based on the base pair composition and (ii) siRNAs with high silencing potential for chemical synthesis. In addition, each siRNA arising from the input query is evaluated for possible cross-silencing activities by performing BLAST searches against the transcriptome or genome of a selected organism. DEQOR can therefore predict the probability that an mRNA fragment will cross-react with other genes in the cell and helps researchers to design experiments to test the specificity of siRNAs or chemically designed siRNAs.
[00116] Suitable Gremlin-2 gene targets include, e.g., a contiguous stretch of from about 10 nucleotides (nt) to about 15 nt, from about 15 nt to about 20 nt, from about 20 nt to about 25 nt, from about 25 nt to about 30 nt, from about 30 nt to about 35 nt, from about 35 nt to about 40 nt, from about 40 nt to about 50 nt, from about 50 nt to about 60 nt, from about 60 nt to about 70 nt, from about 70 nt to about 80 nt, from about 80 nt to about 90 nt, or from about 90 nt to about 100 nt, of a Gremlin-2 mRNA, e.g., of nucleotides 1-4199 of a Gremlin-2 mRNA provided in GenBank NM_022469 {Homo sapiens Gremlin-2 mRNA), or of nucleotides 1-3745 of a Gremlin-2 mRNA provided in GenBank NM_011825 (Mus mus cuius Gremlin-2 mRNA).
[00117] Other suitable target sequences will be readily apparent upon inspection of a
sequence alignment of the nucleotide sequences provided in GenBank Accession Nos. NM_022469, NM_011825, XM_222923, XM_525111, and XM_547488.
[00118] It should be understood that the siNA (e.g., siRNA, e.g., shRNA. e.g. dsRNA) oligonucleotides used would comprise a sequence complementary to the target.
[00119] The following are non-limiting examples of suitable shRNA sequences:
[00120] 5'-
TGCTGTTGACAGTGAGCGCGGAGTCACTAGGAAGCTGTAATAGTGAAGCCACA GATGTATTAC AGCTTCCTAGTG ACTCCTTGCCTACTGCCTCGG A- 3 ' ;
[00121] 5 -
TGCTGTTGACAGTGAGCGCCGGGTTTCAACATTTCTTAATTAGTGAAGCCACA GATGTAATTAAGAA ATGTTGAA ACCCGTTGCCTACTGCCTCGGA-3 ' ; and
[00122] 5'-
TGCTGTTGACAGTGAGCGAGCTACTTCTTGGCTTATTGTATAGTGAAGCCACAG ATGTATACAATAAGCC AAGAAGTAGCCTGCCTACTGCCTCGGA-3 ' . [00123] siNA molecules can be of any of a variety of forms. For example the siNA can be a double- stranded polynucleotide molecule comprising self-complementary sense and antisense regions, wherein the antisense region comprises nucleotide sequence that is complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region having nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof. siNA can also be assembled from two separate oligonucleotides, where one strand is the sense strand and the other is the antisense strand, wherein the antisense and sense strands are self-complementary. In this embodiment, each strand comprises nucleotide sequence that is complementary to nucleotide sequence in the other strand; such as where the antisense strand and sense strand form a duplex or double stranded structure, for example wherein the double stranded region is about 15 to about 30, e.g., about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 base pairs; the antisense strand comprises nucleotide sequence that is complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense strand comprises nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof (e.g., about 15 to about 25 or more nucleotides of the siNA molecule are complementary to the target nucleic acid or a portion thereof).
[00124] Alternatively, the siNA can be assembled from a single oligonucleotide, where the self-complementary sense and antisense regions of the siNA are linked by a nucleic acid- based or non-nucleic acid-based linker(s). The siNA can be a polynucleotide with a duplex, asymmetric duplex, hairpin or asymmetric hairpin secondary structure, having self- complementary sense and antisense regions, wherein the antisense region comprises nucleotide sequence that is complementary to nucleotide sequence in a separate target nucleic acid molecule or a portion thereof and the sense region having nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof.
[00125] The siNA can be a circular single- stranded polynucleotide having two or more loop structures and a stem comprising self-complementary sense and antisense regions, wherein the antisense region comprises nucleotide sequence that is complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof and the sense region having nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof, and wherein the circular polynucleotide can be processed either in vivo or in vitro to generate an active siNA molecule capable of mediating RNAi. The siNA can also comprise a single stranded polynucleotide having nucleotide sequence complementary to nucleotide sequence in a target nucleic acid molecule or a portion thereof (e.g., where such siNA molecule does not require the presence within the siNA molecule of nucleotide sequence corresponding to the target nucleic acid sequence or a portion thereof), wherein the single stranded polynucleotide can further comprise a terminal phosphate group, such as a 5'- phosphate (see for example Martinez et al., 2002, Cell., 110, 563-574 and Schwarz et al., 2002, Molecular Cell, 10, 537-568), or 5',3'-diphosphate.
[00126] In certain embodiments, the siNA molecule contains separate sense and antisense sequences or regions, wherein the sense and antisense regions are covalently linked by nucleotide or non-nucleotide linkers molecules as is known in the art, or are alternately non-covalently linked by ionic interactions, hydrogen bonding, van der Waals interactions, hydrophobic interactions, and/or stacking interactions. In certain embodiments, the siNA molecules comprise nucleotide sequence that is complementary to nucleotide sequence of a target gene. In another embodiment, the siNA molecule interacts with nucleotide sequence of a target gene in a manner that causes inhibition of expression of the target gene.
[00127] As used herein, siNA molecules need not be limited to those molecules containing only RNA, but further encompasses chemically-modified nucleotides and non-nucleotides. In certain embodiments, the short interfering nucleic acid molecules of the invention lack 2' -hydroxy (2'-OH) containing nucleotides. siNAs do not necessarily require the presence of nucleotides having a 2'-hydroxy group for mediating RNAi and as such, siNA molecules of the invention optionally do not include any ribonucleotides (e.g., nucleotides having a 2'-OH group). Such siNA molecules that do not require the presence of ribonucleotides within the siNA molecule to support RNAi can however have an attached linker or linkers or other attached or associated groups, moieties, or chains containing one or more nucleotides with 2'-OH groups. Optionally, siNA molecules can comprise ribonucleotides at about 5, 10, 20, 30, 40, or 50% of the nucleotide positions. The modified short interfering nucleic acid molecules of the invention can also be referred to as short interfering modified oligonucleotides "siMON."
[00128] As used herein, the term siNA is meant to be equivalent to other terms used to
describe nucleic acid molecules that are capable of mediating sequence specific RNA interference (RNAi), for example short interfering RNA (siRNA), double- stranded RNA (dsRNA), micro-RNA (miRNA), short hairpin RNA (shRNA), short interfering oligonucleotide, short interfering nucleic acid, short interfering modified oligonucleotide, chemically-modified siRNA, post-transcriptional gene silencing RNA (ptgsRNA), and others. In addition, as used herein, the term RNAi is meant to be equivalent to other terms used to describe sequence specific RNA interference, such as post transcriptional gene silencing, translational inhibition, or epigenetics. For example, siNA molecules of the invention can be used to epigenetically silence a target gene at the post-transcriptional level or the pre-transcriptional level. In a non-limiting example, epigenetic regulation of gene expression by siNA molecules of the invention can result from siNA mediated modification of chromatin structure or methylation pattern to alter gene expression (see, for example, Verdel et al., 2004, Science, 303, 672-676; Pal-Bhadra et al., 2004, Science, 303, 669-672; Allshire, 2002, Science, 297, 1818-1819; Volpe et al., 2002, Science, 297, 1833-1837; Jenuwein, 2002, Science, 297, 2215-2218; and Hall et al., 2002, Science, 297, 2232-2237).
[00129] siNA molecules contemplated herein can comprise a duplex forming
oligonucleotide (DFO) see, e.g., WO 05/019453; and US 2005/0233329, which are incorporated herein by reference). siNA molecules also contemplated herein include multifunctional siNA, (see, e.g., WO 05/019453 and US 2004/0249178). The
multifunctional siNA can comprise sequence targeting, for example, two regions of PKDl.
[00130] siNA molecules contemplated herein can comprise an asymmetric hairpin or
asymmetric duplex. By "asymmetric hairpin" as used herein is meant a linear siNA molecule comprising an antisense region, a loop portion that can comprise nucleotides or non-nucleotides, and a sense region that comprises fewer nucleotides than the antisense region to the extent that the sense region has enough complementary nucleotides to base pair with the antisense region and form a duplex with loop. For example, an asymmetric hairpin siNA molecule can comprise an antisense region having length sufficient to mediate RNAi in a cell or in vitro system (e.g. about 15 to about 30, or about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides) and a loop region comprising about 4 to about 12 (e.g., about 4, 5, 6, 7, 8, 9, 10, 11, or 12) nucleotides, and a sense region having about 3 to about 25 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) nucleotides that are complementary to the antisense region. The asymmetric hairpin siNA molecule can also comprise a 5'-terminal phosphate group that can be chemically modified. The loop portion of the asymmetric hairpin siNA molecule can comprise nucleotides, non-nucleotides, linker molecules, or conjugate molecules as described herein.
[00131] By "asymmetric duplex" as used herein is meant a siNA molecule having two
separate strands comprising a sense region and an antisense region, wherein the sense region comprises fewer nucleotides than the antisense region to the extent that the sense region has enough complementary nucleotides to base pair with the antisense region and form a duplex. For example, an asymmetric duplex siNA molecule of the invention can comprise an antisense region having length sufficient to mediate RNAi in a cell or in vitro system (e.g. about 15 to about 30, or about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleotides) and a sense region having about 3 to about 25 (e.g., about 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25) nucleotides that are complementary to the antisense region.
[00132] Stability and/or half-life of siNAs (e.g., siRNAs, shRNAs, etc.) can be improved through chemically synthesizing nucleic acid molecules with modifications (base, sugar and/or phosphate) can prevent their degradation by serum ribonucleases, which can increase their potency (see e.g., Eckstein et al., International Publication No. WO
92/07065; Perrault et al., 1990 Nature 344, 565; Pieken et al., 1991, Science 253, 314; Usman and Cedergren, 1992, Trends in Biochem. Sci. 17, 334; Usman et al., International Publication No. WO 93/15187; and Rossi et al., International Publication No. WO
91/03162; Sproat, U.S. Pat. No. 5,334,711; Gold et al., U.S. Pat. No. 6,300,074; and Burgin et al., supra; all of which are incorporated by reference herein, describing various chemical modifications that can be made to the base, phosphate and/or sugar moieties of the nucleic acid molecules described herein. Modifications that enhance their efficacy in cells, and removal of bases from nucleic acid molecules to shorten oligonucleotide synthesis times and reduce chemical requirements are desired.
[00133] For example, oligonucleotides can be modified to enhance stability and/or enhance biological activity by modification with nuclease resistant groups, for example, 2'-amino, 2'-C-allyl, 2'-fluoro, 2'-0-methyl, 2'-0-allyl, 2'-H, nucleotide base modifications (for a review see Usman and Cedergren, 1992, TIBS. 17, 34; Usman et al., 1994, Nucleic Acids Symp. Ser. 31, 163; Burgin et al., 1996, Biochemistry, 35, 14090). Sugar modification of nucleic acid molecules have been extensively described in the art (see Eckstein et al., International Publication PCT No. WO 92/07065; Perrault et al. Nature, 1990, 344, 565- 568; Pieken et al. Science, 1991, 253, 314-317; Usman and Cedergren, Trends in Biochem. Sci., 1992, 17, 334-339; Usman et al. International Publication PCT No. WO 93/15187; Sproat, U.S. Pat. No. 5,334,711 and Beigelman et al., 1995, J. Biol. Chem., 270, 25702; Beigelman et al., International PCT publication No. WO 97/26270; Beigelman et al., U.S. Pat. No. 5,716,824; Usman et al., U.S. Pat. No. 5,627,053; Woolf et al., International PCT Publication No. WO 98/13526; Thompson et al., U.S. Ser. No. 60/082,404 which was filed on Apr. 20, 1998; Karpeisky et al., 1998, Tetrahedron Lett., 39, 1131; Eamshaw and Gait, 1998, Biopolymers (Nucleic Acid Sciences), 48, 39-55; Verma and Eckstein, 1998, Annu. Rev. Biochem., 67, 99-134; and Burlina et al., 1997, Bioorg. Med. Chem., 5, 1999-2010; each of which are hereby incorporated in their totality by reference herein). In view of such teachings, similar modifications can be used as described herein to modify the siNA nucleic acid molecules of disclosed herein so long as the ability of siNA to promote RNAi in cells is not significantly inhibited.
[00134] Short interfering nucleic acid (siNA) molecules having chemical modifications that maintain or enhance activity are contemplated herein. Such a nucleic acid is also generally more resistant to nucleases than an unmodified nucleic acid. Accordingly, the in vitro and/or in vivo activity should not be significantly lowered. Nucleic acid molecules delivered exogenously are generally selected to be stable within cells at least for a period sufficient for transcription and/or translation of the target RNA to occur and to provide for modulation of production of the encoded mRNA and/or polypeptide so as to facilitate reduction of the level of the target gene product.
[00135] Production of RNA and DNA molecules can be accomplished synthetically and can provide for introduction of nucleotide modifications to provide for enhanced nuclease stability, (see, e.g., Wincott et al., 1995, Nucleic Acids Res. 23, 2677; Caruthers et al., 1992, Methods in Enzymology 211, 3-19, incorporated by reference herein. In one embodiment, nucleic acids of the present disclosure include one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) G-clamp nucleotides, which are modified cytosine analogs which confer the ability to hydrogen bond both Watson-Crick and Hoogsteen faces of a complementary guanine within a duplex, and can provide for enhanced affinity and specificity to nucleic acid targets (see, e.g., Lin et al. 1998, J. Am. Chem. Soc, 120, 8531- 8532). In another example, nucleic acid molecules can include one or more (e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more) LNA "locked nucleic acid" nucleotides such as a 2',4'-C methylene bicyclo nucleotide (see, e.g., Wengel et al., WO 00/66604 and WO 99/14226).
[00136] siNA molecules can be provided as conjugates and/or complexes, e.g., to facilitate delivery of siNA molecules into a cell. Exemplary conjugates and/or complexes include those composed of an siNA and a small molecule, lipid, cholesterol, phospholipid, nucleoside, antibody, toxin, negatively charged polymer (e.g., protein, peptide, hormone, carbohydrate, polyethylene glycol, or polyamine). In general, the transporters described are designed to be used either individually or as part of a multi-component system, with or without degradable linkers. These compounds can improve delivery and/or localization of nucleic acid molecules into cells in the presence or absence of serum (see, e.g., US 5,854,038). Conjugates of the molecules described herein can be attached to biologically active molecules via linkers that are biodegradable, such as biodegradable nucleic acid linker molecules.
[00137] Interfering RNAs may be generated exogenously by chemical synthesis, by in vitro transcription, or by cleavage of longer double- stranded RNA with dicer or another appropriate nuclease with similar activity. Chemically synthesized interfering RNAs, produced from protected ribonucleoside phosphoramidites using a conventional
DNA/RNA synthesizer, may be obtained from commercial suppliers such as Ambion Inc. (Austin, Tex.), Invitrogen (Carlsbad, Calif.), or Dharmacon (Lafayette, Colo.). Interfering RNAs are purified by extraction with a solvent or resin, precipitation, electrophoresis, chromatography, or a combination thereof, for example. Alternatively, interfering RNA may be used with little if any purification to avoid losses due to sample processing.
[00138] Interfering RNAs can also be expressed endogenously from plasmid or viral
expression vectors or from minimal expression cassettes, for example, polymerase chain reaction (PCR)-generated fragments comprising one or more promoters and an appropriate template or templates for the interfering RNA. The present disclosure provides a recombinant expression vector comprising a nucleotide sequence encoding an interfering RNA. The nucleotide sequence encoding the interfering RNA can be operably linked to a promoter, e.g., an inducible promoter, a muscle- specific promoter, a constitutive promoter, etc.
[00139] Examples of commercially available plasmid-based expression vectors for shRNA include members of the pSilencer series (Ambion, Austin, Tex.) and pCpG-siRNA (InvivoGen, San Diego, Calif.). Viral vectors for expression of interfering RNA may be derived from a variety of viruses including adenovirus, adeno-associated virus, lenti virus (e.g., human immunodeficiency virus-based vectors, feline immunodeficiency virus-based vectors, and EIAV), and herpes virus. Examples of commercially available viral vectors for shRNA expression include pSilencer adeno (Ambion, Austin, Tex.) and
pLenti6/BLOCK-iT™-DEST (Invitrogen, Carlsbad, Calif.). Selection of viral vectors, methods for expressing the interfering RNA from the vector and methods of delivering the viral vector are within the ordinary skill of one in the art. Examples of kits for production of PCR- generated shRNA expression cassettes include Silencer Express (Ambion, Austin, Tex.) and siXpress (Minis, Madison, Wis.).
[00140] In some embodiments, an interfering RNA-encoding nucleotide sequence is
operably linked to a muscle-specific control element (e.g., a promoter, an enhancer).
Muscle- specific promoters and other control elements (e.g., enhancers) are known in the art. Suitable muscle-specific control sequences include, but are not limited to, a Pitx3 promoter (see, e.g., Coulon et al. (2007) J. Biol. Chem. 282:33192); a desmin promoter (see, e.g., Talbot et al. (2010) Mol. Ther. 18:601); a muscle creatine kinase promoter (see, e.g., Wang et al. (2008) Gene Ther. 15: 1489); a smooth muscle a-actin promoter; an a- myosin heavy chain promoter (see, e.g., Franz et al. (1997) Cardiovasc. Res. 35:560); a myosin light chain 2 promoter; a mef2c promoter; a fast skeletal troponin T promoter (see, e.g., Stefancsik et al. (2003) Comp. Funct. Genomics 4:609); a SM22a promoter (see, e.g., Akyurek et al. (2000) Mol. Med. 6:983; and U.S. Patent No. 7,169,874); a smoothelin promoter (see, e.g., WO 2001/018048); an a-smooth muscle actin promoter; and the like. See also, e.g., Pacak et al. (2008) Genetic Vaccines and Therapy 6: 13; Kim, et al. (1997) Mol. Cell. Biol. 17, 2266-2278; Li, et al., (1996) J. Cell Biol. 132, 849-859; and Moessler, et al. (1996) Development 122, 2415-2425.
[00141] An interfering RNA can be delivered in a delivery system that provides tissue
targetable delivery. In addition, a suitable formulation for an interfering nucleic acid can include one or more additional properties: 1) nucleic acid binding into a core that can release the siRNA into the cytoplasm; 2) protection from non-specific interactions; 3) and tissue targeting that provides cell uptake. In some embodiments, the composition comprises a modular polymer conjugate targeting muscle cells by coupling a peptide ligand specific for those cells to one end of a protective polymer, coupled at its other end to a cationic carrier for nucleic acids. For example, a suitable polymer conjugate can have three functional domains: peptide ligand specific for a target cell; protective polymer; and cationic carrier for nucleic acids. Another suitable formulation includes surface coatings attached to a preformed nanoparticle.
[00142] Suitable formulations for delivery of an interfering nucleic acid include polymers, polymer conjugates, lipids, micelles, self-assembly colloids, nanoparticles, sterically stabilized nanoparticles, and ligand-directed nanoparticles.
[00143] The present disclosure provides a composition comprising a subject interfering nucleic acid. A subject interfering nucleic acid composition can comprise, in addition to a subject interfering nucleic acid, one or more of: a salt, e.g., NaCl, MgCl, KC1, MgS04, etc.; a buffering agent, e.g., a Tris buffer, N-(2-Hydroxyethyl)piperazine-N'-(2- ethanesulfonic acid) (HEPES), 2-(N-Morpholino)ethanesulfonic acid (MES), 2-(N- Morpholino)ethanesulfonic acid sodium salt (MES), 3-(N-Morpholino)propanesulfonic acid (MOPS), N-tris[Hydroxymethyl]methyl-3-aminopropanesulfonic acid (TAPS), etc.; a solubilizing agent; a detergent, e.g., a non-ionic detergent such as Tween-20, etc.; a nuclease inhibitor; glycerol; and the like.
[00144] The present disclosure further provides pharmaceutical compositions comprising a subject interfering nucleic acid. Thus, the present disclosure provides a pharmaceutical composition comprising a subject interfering nucleic acid and a pharmaceutically acceptable excipient. Pharmaceutically acceptable excipients are described below.
FORMULATIONS, ROUTES OF ADMINISTRATION, AND DOSAGES
[00145] An agent (e.g., an anti-Gremlin-2 antibody; a Gremlin-2-specific siNA) that
reduces circulating and/or tissue levels of Gremlin-2 and/or that reduces Gremlin-2 activity can be provided in a pharmaceutical composition, for administration to an individual in need thereof.
[00146] A composition comprising an agent that reduces circulating and/or tissue levels of
Gremlin-2 and/or that reduces Gremlin-2 activity (where such agents include, e.g., an anti- Gremlin-2 antibody; a Gremlin-2- specific siNA) can comprise a pharmaceutically acceptable excipient, a variety of which are known in the art and need not be discussed in detail herein. Pharmaceutically acceptable excipients have been amply described in a variety of publications, including, for example, "Remington: The Science and Practice of Pharmacy", 19th Ed. (1995), or latest edition, Mack Publishing Co; A. Gennaro (2000) "Remington: The Science and Practice of Pharmacy", 20th edition, Lippincott, Williams, & Wilkins; Pharmaceutical Dosage Forms and Drug Delivery Systems (1999) H.C. Ansel et al., eds 7th ed., Lippincott, Williams, & Wilkins; and Handbook of Pharmaceutical
Excipients (2000) A.H. Kibbe et al., eds., 3 rd ed. Amer. Pharmaceutical Assoc.
[00147] A subject pharmaceutical composition can comprise an agent that reduces
circulating and/or tissue levels of Gremlin-2 and/or that reduces Gremlin-2 activity, and a pharmaceutically acceptable excipient. In some cases, a subject pharmaceutical
composition will be suitable for injection into a subject, e.g., will be sterile. For example, in some embodiments, a subject pharmaceutical composition will be suitable for injection into a human subject, e.g., where the composition is sterile and is free of detectable pyrogens and/or other toxins.
[00148] A subject pharmaceutical composition may comprise other components, such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium, carbonate, and the like. The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, toxicity adjusting agents and the like, for example, sodium acetate, sodium chloride, potassium chloride, calcium chloride, sodium lactate, hydrochloride, sulfate salts, solvates (e.g., mixed ionic salts, water, organics), hydrates (e.g., water), and the like.
Routes of administration
[00149] In practicing the methods, routes of administration may vary. An agent that reduces circulating and/or tissue levels of Gremlin-2 and/or that reduces Gremlin-2 activity (where agents include, e.g., an anti-Gremlin-2 antibody; a Gremlin-2-specific siNA) can be delivered by a route that provides for delivery of the agent to the bloodstream (e.g., by parenteral administration, such as intravenous administration, intramuscular
administration, and/or subcutaneous administration) or to a specific tissue (e.g., muscle tissue). Injection can be used to accomplish parenteral administration. In some
embodiments, an anti-Gremlin-2 antibody is delivered by a route that provides for delivery of the antibody directly into affected muscle tissue, e.g., by intramuscular injection.
Dosages
[00150] In the methods, a therapeutically effective amount of an agent that reduces the level and/or activity of Gremlin-2 is administered to a subject in need thereof. For example, an agent that reduces levels and/or activity of Gremlin-2 can increase muscle function and/or muscle mass, and can in some cases cause a return to a normal level of muscle function and/or muscle mass relative to a healthy individual when the agent is delivered to the bloodstream or directly into muscle tissue in an effective amount to a patient who previously did not have a normal level of muscle function and/or muscle mass relative to a healthy individual prior to being treated.
[00151] The amount administered varies depending upon the goal of the administration, the health and physical condition of the individual to be treated, age, the degree of resolution desired, the formulation of a subject antibody, the activity of the subject antibody employed, the treating clinician's assessment of the medical situation, the condition of the subject, and the body weight of the subject, as well as the severity of the disease, disorder, or condition, and other relevant factors. The size of the dose will also be determined by the existence, nature, and extent of any adverse side-effects that might accompany the administration of a particular antibody.
[00152] It is expected that the amount will fall in a relatively broad range that can be
determined through routine trials. For example, the amount of an anti-Gremlin-2 antibody employed to increase muscle mass and/or muscle strength or other muscle function is not more than about the amount that could otherwise be irreversibly toxic to the subject (i.e., maximum tolerated dose). In other cases, the amount is around or even well below the toxic threshold, but still in an effective concentration range, or even as low as threshold dose.
[00153] Also, suitable doses and dosage regimens can be determined by comparisons to indicators of normal muscle mass and/or function. Such dosages include dosages which result in increased muscle mass and/or function, for example, comparable to a healthy individual, without significant side effects. Dosage treatment may be a single dose schedule or a multiple dose schedule (e.g., including ramp and maintenance doses). As indicated below, a subject composition may be administered in conjunction with other agents, and thus doses and regimens can vary in this context as well to suit the needs of the subject.
[00154] Individual doses are typically not less than an amount required to produce a
measurable effect on the subject, and may be determined based on the pharmacokinetics and pharmacology for absorption, distribution, metabolism, and excretion ("ADME") of the subject antibody or its by-products, and thus based on the disposition of the composition within the subject. This includes consideration of the route of administration as well as dosage amount, which can be adjusted for enteral (applied via digestive tract for systemic or local effects when retained in part of the digestive tract) or parenteral (applied by routes other than the digestive tract for systemic or local effects) applications. For instance, administration of a subject antibody can be via injection, e.g., via intravenous injection, intramuscular injection, or a combination thereof.
[00155] By "therapeutically effective amount" is meant that the administration of that
amount to an individual, either in a single dose, as part of a series of the same or different antibody compositions, is effective to increase muscle mass and/or muscle function in a subject. As noted above, the therapeutically effective amount can be adjusted in connection with dosing regimen and diagnostic analysis of the subject's condition (e.g., monitoring muscle mass) and the like.
[00156] As an example, the effective amount of a dose or dosing regimen can be gauged from the ED50 of an agent for inducing an action that leads to an increase in muscle mass by a certain amount and/or an increase in muscle function by a certain degree. By "ED50" (effective dosage) is the intended dosage which induces a response halfway between the baseline and maximum after some specified exposure time. The ED50 of a graded dose response curve therefore represents the concentration of an agent (e.g., a subject antibody) where 50% of its maximal effect is observed. ED50 may be determined by in vivo studies (e.g. animal models) using methods known in the art.
[00157] An effective amount may not be more than 100X the calculated ED50. For instance, the amount of an agent (e.g., an anti-Gremlin-2 antibody) that is administered is less than about 100X, less than about 50X, less than about 40X, 35X, 30X, or 25X and many embodiments less than about 20X, less than about 15X and even less than about 10X, 9X, 9X, 7X, 6X, 5X, 4X, 3X, 2X or IX than the calculated ED50. In one embodiment, the effective amount is about IX to 30X of the calculated ED50, and sometimes about IX to 20X, or about IX to 10X of the calculated ED50. In other embodiments, the effective amount is the same as the calculated ED50, and in certain embodiments the effective amount is an amount that is more than the calculated ED50.
[00158] An effective amount of an agent (e.g., a subject antibody) may also an amount that is effective, when administered in one or more doses, to increase muscle function and/or muscle mass by at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, or more than 80%, compared to the level of muscle function and/or the muscle mass in the individual not treated with the agent.
[00159] Further examples of dose per administration may be at less than 10 μg, less than 2 μg, or less than 1 μg. Dose per administration may also be more than 50 μg, more 100 μg, more than 300 μg up to 600 μg or more. An example of a range of dosage per weight is about 0.1 μg/kg to about 1 μg/kg, up to about 1 mg/kg or more. Effective amounts and dosage regimen can readily be determined empirically from assays, from safety and escalation and dose range trials, individual clinician-patient relationships, as well as in vitro and in vivo assays known in the art.
[00160] The term "unit dosage form," as used herein, refers to physically discrete units suitable as unitary dosages for human and animal subjects, each unit containing a predetermined quantity of an active agent (e.g., an agent that reduces circulating and/or tissue levels of Gremlin-2) calculated in an amount sufficient to produce the desired effect in association with a pharmaceutically acceptable diluent, carrier or vehicle. The specifications for the novel unit dosage forms depend on the particular protein employed and the effect to be achieved, and the pharmacodynamics associated with each active agent in the host.
COMBINATION THERAPY
[00161] Any of a variety of therapies directed to increasing muscle function and/or muscle mass can be combined in a composition or therapeutic method with an anti-Gremlin-2 antibody. A subject antibody can also be administered in combination with a modified diet and/or exercise regimen to promote muscle strength and/or muscle mass.
[00162] "Combination" as used herein is meant to include therapies that can be
administered separately, e.g. formulated separately for separate administration (e.g., as may be provided in a kit), or undertaken as a separate regime (as in exercise and diet modifications), as well as for administration in a single formulation (i.e., "co-formulated").
[00163] Second therapeutic agents that can be administered in combination therapy with an anti-Gremlin-2 antibody include, but are not limited to, follistatin (see, e.g., Kota et al. (2009) Sci. Transl. Med. I:6ral5; and U.S. Patent Publication No. 2010/0178348); a follistatin domain-containing protein other than follistatin (see, e.g., U.S. Patent Publication No. 2011/0020372); a corticosteroid; a myostatin inhibitor (see, e.g., U.S. Patent Publication No. 2010/0330072); an anti-activin receptor IIB antibody (see, e.g., U.S. Patent Publication No. 2010/0272734); a truncated activin receptor IIB (see, e.g., U.S. Patent Publication No. 2011/0034372); and the like.
[00164] Where a subject antibody is administered in combination with one or more other therapies, the combination can be administered anywhere from simultaneously to up to 5 hours or more, e.g., 10 hours, 15 hours, 20 hours or more, prior to or after administration of a subject protein. In certain embodiments, a subject antibody and other therapeutic intervention are administered or applied sequentially, e.g., where a subject antibody is administered before or after another therapeutic treatment. In yet other embodiments, a subject antibody and other therapy are administered simultaneously, e.g., where a subject antibody and a second therapy are administered at the same time, e.g., when the second therapy is a drug it can be administered along with a subject antibody as two separate formulations or combined into a single composition that is administered to the subject. Regardless of whether administered sequentially or simultaneously, as illustrated above, the treatments are considered to be administered together or in combination for purposes of the present disclosure.
PATIENT POPULATIONS
[00165] Individuals suitable for treatment with a subject method of increasing muscle
function and/or muscle mass include individuals having a deficiency in muscle function and/or having reduced muscle mass. Individuals suitable for treatment with a subject method of increasing muscle function and/or muscle mass include individuals having a disease, disorder, or condition associated with or resulting in reduced muscle function and/or muscle mass, e.g., a disease, disorder, or condition in which reduced muscle function and/or muscle mass is a symptom or a sequela of the disease, disorder, or condition. Such diseases, disorders, or conditions include immobilization, chronic disease, cancer, and injury.
[00166] Individuals suitable for treatment with a subject method for reducing levels and/or activity of Gremlin-2 include individuals having a disease, disorder, or condition characterized, at least in part, by elevated circulating and/or tissue levels of Gremlin-2. Thus, an individual to be treated using a method of the present disclosure can have elevated Gremlin-2 levels, e.g., elevated circulating and/or tissue levels of Gremlin-2, compared to a normal control level of Gremlin-2. Such individuals can include individuals having a disease, disorder, or condition associated with or resulting in reduced muscle function and/or muscle mass, e.g., a disease, disorder, or condition in which reduced muscle function and/or muscle mass is a symptom or a sequela of the disease, disorder, or condition.
GREMLIN-2
[00167] A Gremlin-2 polypeptide can be targeted in order to reduce levels and/or activity of
Gremlin-2 polypeptide, and/or to increase muscle function and/or muscle mass in a subject. Methods and compositions targeting a Gremlin-2 polypeptide find use in reducing circulating and/or tissue levels and/or reducing activity of Gremlin-2 polypeptide, and/or increasing muscle function and/or muscle mass in an individual having a disorder, disease, or condition associated with or resulting in reduced muscle function and/or mass.
[00168] "Gremlin-2 polypeptide" encompasses naturally-occurring Gremlin-2 polypeptides and homologues from different species. "Gremlin-2 polypeptide" encompasses naturally- occurring variants. Gremlin-2 encompasses murine and human variants that are encoded by the GREM2 gene or a gene homologous to GREM2.
[00169] Gremlin-2 refers to Gremlin-2 proteins or Gremlin-2 DNA sequences, which
encompass their naturally occurring isoforms and/or allelic/splice variants. A Gremlin-2 protein also refers to proteins that have one or more alteration in the amino acid residues (e.g. at locations that are not conserved across variants and/or species) while retaining the conserved domains and having the same biological activity as the naturally- occurring Gremlin-2. Gremlin-2 also encompasses nucleic acid sequences that vary in one or more bases from a naturally-occurring DNA sequence but still translate into an amino acid sequence that correspond to the a naturally- occurring protein due to degeneracy of the genetic code. For example, Gremlin-2 may also refer to those that differ from the naturally- occurring sequences of Gremlin-2 by one or more conservative substitutions and/or tags and/or conjugates.
[00170] Proteins targeted by the method of the present disclosure can contain a contiguous amino acid residues of a length derived from Gremlin-2. A sufficient length of contiguous amino acid residues may vary depending on the specific naturally-occurring amino acid sequence from which the protein is derived. For example, the protein may be at least 100 amino acids to 125 amino acid residues in length, at least 125 amino acids to 150 amino acid residues in length, or at least 165 amino acids up to the full-length protein (e.g., 165 amino acids, 166 amino acids, 167 amino acids, 168 amino acids). For example, the protein may be of about 168 amino acid residues in length when derived from a human Gremlin-2 protein or from a mouse Gremlin-2 protein.
[00171] A protein containing an amino acid sequence that is substantially similar to the amino acid sequence of a Gremlin-2 polypeptide includes a polypeptide comprising an amino acid sequence having at least about 90%, at least about 94%, at least about 95%, at least about 98%, or at least about 99%, or 100%, amino acid sequence identity to a contiguous stretch of from about 100 amino acids (aa) to about 125 aa, from about 125 aa to about 150 aa, from about 150 aa to about 165 aa, or from about 165 aa up to the full length of a naturally occurring Gremlin-2 polypeptide. For example, a Gremlin-2 polypeptide suitable as a target in a subject method can comprise an amino acid sequence having at least about 90%, at least about 94%, at least about 95%, at least about 98%, or at least about 99%, or 100%, amino acid sequence identity to a contiguous stretch of from about 100 amino acids (aa) to about 125 aa, from about 125 aa to about 150 aa, from about 150 aa to about 165 aa, or from 165 aa up to the full length of the human Gremlin-2 polypeptide amino acid sequence depicted in Figure 15.
[00172] The protein may lack at least 5, at least 10, up to at least 50 or more aa relative to a naturally-occurring full-length Gremlin-2 polypeptide. For example, the protein may not contain the signal sequence of based on the amino acid sequence of a naturally-occurring Gremlin-2 polypeptide. The Gremlin-2 protein may also contain the same or similar glycosylation pattern as that of a naturally- occurring Gremlin-2 polypeptide, may contain no glycosylation, or may contain the glycosylation pattern of host cells used to produce the protein.
[00173] Many DNA and protein sequences of Gremlin-2 are known in the art and certain sequences are discussed below.
[00174] The proteins targeted in the method of the present disclosure include those
containing contiguous amino acid sequences of any naturally-occurring Gremlin-2 polypeptide, as well as those having 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 usually no more than 20, 10, or 5 amino acid substitutions, where the substitution is usually a conservative amino acid substitution. By "conservative amino acid substitution" generally refers to substitution of amino acid residues within the following groups:
1) L, I, M, V, F;
2) R, K;
3) F, Y, H, W, R;
4) G, A, T, S;
5) Q, N; and
6) D, E.
[00175] Conservative amino acid substitutions in the context of a peptide or a protein
disclosed herein are selected so as to preserve putative activity of the protein. Such presentation may be preserved by substituting with an amino acid with a side chain of similar acidity, basicity, charge, polarity, or size to the side chain of the amino acid being replaced. Guidance for substitutions, insertion, or deletion may be based on alignments of amino acid sequences of different variant proteins or proteins from different species. For example, at certain residue positions that are fully conserved (*), substitution, deletion or insertion may not be allowed while at other positions where one or more residues are not conserved, an amino acid change can be tolerated. Residues that are semi-conserved (. or :) may tolerate changes that preserve charge, polarity, and/or size.
[00176] The present disclosure provides methods and compositions for targeting any of the
Gremlin-2 polypeptides described above. For screening and characterization purposes, the protein may be isolated from a natural source, e.g., is in an environment other than its naturally-occurring environment. The subject protein may also be recombinantly made, e.g., in a genetically modified host cell (e.g., bacteria; yeast; Pichia; insect cells; and the like), where the genetically modified host cell is genetically modified with a nucleic acid comprising a nucleotide sequence encoding the subject protein. The subject protein encompasses synthetic polypeptides, e.g., a subject synthetic polypeptide is synthesized chemically in a laboratory (e.g., by cell-free chemical synthesis). Methods of productions are described in more detail below.
Nucleic acid and protein sequences
[00177] The polypeptide to be targeted in the subject methods may be generated using
recombinant techniques to manipulate various Gremlin-2 nucleic acids known in the art to provide constructs encoding a protein of interest. It will be appreciated that, provided an amino acid sequence, the ordinarily skilled artisan will immediately recognize a variety of different nucleic acids encoding such amino acid sequence in view of the knowledge of the genetic code.
[00178] For production of a Gremlin-2 protein derived from naturally- occurring
polypeptides, it is noted that nucleic acids encoding a variety of different Gremlin-2 polypeptides are known and available in the art. Nucleotide sequences encoding Gremlin-2 polypeptides are also provided in GenBank. Several sequences and further information on the nucleic acid and protein sequences can also be found below and in the Examples section.
[00179] Nucleic acid (and amino acid sequences) for various Gremlin-2 are provided in
GenBank as accession nos.: 1) Homo sapiens: amino acid sequence NP_071914;
nucleotide sequence: NM_022469) Mus musculus: amino acid sequence NP_035955; nucleotide sequence NM_011825), Rattus norvegicus: amino acid sequence XP_222923; nucleotide sequence XM_222923, Pan troglodytes: amino acid sequence XP_525111; nucleotide sequence XM_525111; and Canis familiaris: amino acid sequence XP_547488; nucleotide sequence XM_547488. Exemplary Gremlin-2 amino acid sequences are depicted in Figure 15.
[00180] It will be appreciated that the nucleotide sequences encoding the protein may be modified so as to optimize the codon usage to facilitate expression in a host cell of interest (e.g., Escherichia coli, and the like). Methods for production of codon optimized sequences are known in the art.
METHODS OF PRODUCTION
[00181] Gremlin-2 polypeptides and anti-Gremlin-2 antibodies can be produced by any suitable method, including recombinant and non-recombinant methods (e.g., chemical synthesis).
[00182] Where a polypeptide is chemically synthesized, the synthesis may proceed via liquid-phase or solid-phase. Solid-phase synthesis (SPPS) allows the incorporation of unnatural amino acids, peptide/protein backbone modification. Various forms of SPPS, such as Fmoc and Boc, are available for synthesizing peptides of the present invention. Details of the chemical synthesis are known in the art (e.g. Ganesan A. 2006 Mini Rev. Med Chem. 6:3-10 and Camarero JA et al. 2005 Protein Pept Lett. 12:723-8). Briefly, small insoluble, porous beads are treated with functional units on which peptide chains are built. After repeated cycling of coupling/deprotection, the free N-terminal amine of a solid- phase attached is coupled to a single N-protected amino acid unit. This unit is then deprotected, revealing a new N-terminal amine to which a further amino acid may be attached. The peptide remains immobilized on the solid-phase and undergoes a filtration process before being cleaved off.
[00183] Where the Gremlin-2 protein and/or anti-Gremlin-2 antibody is produced using recombinant techniques, the proteins and/or antibody may be produced as an intracellular protein or as an secreted protein, using any suitable construct and any suitable host cell, which can be a prokaryotic or eukaryotic cell, such as a bacterial (e.g. E. coli) or a yeast host cell, respectively.
[00184] Other examples of eukaryotic cells that may be used as host cells include insect cells, mammalian cells, and/or plant cells. Where mammalian host cells are used, the cells may include one or more of the following: human cells (e.g. HeLa, 293, H9 and Jurkat cells); mouse cells (e.g., NIH3T3, L cells, and C127 cells); primate cells (e.g. Cos 1, Cos 7 and CV1) and hamster cells (e.g., Chinese hamster ovary (CHO) cells).
[00185] A wide range of host- vector systems suitable for the expression of the subject
protein and/or antibody may be employed according standard procedures known in the art. See for example, Sambrook et al. 1989 Current Protocols in Molecular Biology Cold Spring Harbor Press, New York and Ausubel et al. 1995 Current Protocols in Molecular Biology, Eds. Wiley and Sons.
[00186] Methods for introduction of genetic material into host cells include, for example, transformation, electroporation, conjugation, calcium phosphate methods and the like. The method for transfer can be selected so as to provide for stable expression of the introduced Gremlin-2-encoding nucleic acid or the introduced anti-Gremlin-2 antibody-encoding nucleic acid. The polypeptide-encoding nucleic acid can be provided as an inheritable episomal element (e.g., plasmid) or can be genomically integrated. A variety of appropriate vectors for use in production of a polypeptide of interest are available commercially.
[00187] Vectors can provide for extrachromosomal maintenance in a host cell or can
provide for integration into the host cell genome. The expression vector provides transcriptional and translational regulatory sequences, and may provide for inducible or constitutive expression, where the coding region is operably linked under the
transcriptional control of the transcriptional initiation region, and a transcriptional and translational termination region. In general, the transcriptional and translational regulatory sequences may include, but are not limited to, promoter sequences, ribosomal binding sites, transcriptional start and stop sequences, translational start and stop sequences, and enhancer or activator sequences. Promoters can be either constitutive or inducible, and can be a strong constitutive promoter (e.g., T7, and the like).
[00188] Expression constructs generally have convenient restriction sites located near the promoter sequence to provide for the insertion of nucleic acid sequences encoding proteins of interest. A selectable marker operative in the expression host may be present to facilitate selection of cells containing the vector. In addition, the expression construct may include additional elements. For example, the expression vector may have one or two replication systems, thus allowing it to be maintained in organisms, for example in mammalian or insect cells for expression and in a prokaryotic host for cloning and amplification. In addition the expression construct may contain a selectable marker gene to allow the selection of transformed host cells. Selectable genes are well known in the art and will vary with the host cell used.
[00189] Isolation and purification of a protein and/or antibody can be accomplished
according to methods known in the art. For example, a protein can be isolated from a lysate of cells genetically modified to express the protein constitutively and/or upon induction, or from a synthetic reaction mixture, by immunoaffinity purification, which generally involves contacting the sample with an anti- protein antibody, washing to remove non- specifically bound material, and eluting the specifically bound protein. The isolated protein can be further purified by dialysis and other methods normally employed in protein purification methods. In one embodiment, the protein may be isolated using metal chelate chromatography methods. Protein of the present disclosure may contain modifications to facilitate isolation.
[00190] The subject proteins and/or antibody may be prepared in substantially pure or
isolated form (e.g., free from other polypeptides). The protein can present in a composition that is enriched for the polypeptide relative to other components that may be present (e.g., other polypeptides or other host cell components). Purified protein may be provided such that the protein is present in a composition that is substantially free of other expressed proteins, e.g., less than 98%, less than 95%, less than 90%, less than 80%, less than 60%, or less than 50%, of the composition is made up of other expressed proteins. Antibody production
[00191] A subject anti-Gremlin-2 antibody can be prepared using a wide variety of
techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof. For example, a subject anti-Gremlin-2 antibody may be made and isolated using methods of phage display. A subject anti- Gremlin-2 antibody may also be isolated from sera of an animal host immunized with an immunogenic composition containing a Gremlin-2 protein, which encompasses whole proteins and fragments thereof.
[00192] The antigen that coats the wells for phage display panning or the immunogenic composition used to elicit the antibody of the present disclosure may contain an aggregate of one or more Gremlin-2 polypeptides, as described above. The method may involve exposing antigens to an aggregating condition so as to form an aggregate. Thus the methods of production described may further include a step of forming an aggregate of the isolated antigens. Examples of the aggregating conditions include heating, addition of an excipient that facilitates aggregation, and the like.
[00193] Antigens used to coat the wells for phage panning or to elicit a subject anti-
Gremlin-2 antibody may be conjugated to another molecule. For example, the antigen can be conjugated to a second molecule such as a peptide, polypeptide, lipid, carbohydrate and the like that aids in solubility, storage or other handling properties, cell permeability, half- life, controls release and/or distribution such as by targeting a particular cell (e.g., muscle cells, etc.) or cellular location (e.g., lysosome, endosome, mitochondria etc.), tissue or other bodily location (e.g., blood, muscle tissue, particular organs etc.).
[00194] A particular embodiment of an antigen conjugated to a second molecule is where the second molecule is an immunomodulator. "Immunomodulator" is a molecule that directly or indirectly modifies an immune response. A specific class of immunomodulators includes those that stimulate or aid in the stimulation of an immunological response. Non- limiting examples include antigens and antigen carriers such as a toxin or derivative thereof, including tetanus toxoid.
Phage display
[00195] Phage display can be used for the high-throughput screening of protein interactions.
Phages may be utilized to display antigen-binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine). Phage expressing an antigen binding domain that binds Gremlin-2 can be selected or identified with Gremlin-2, e.g., using labeled Gremlin-2 or Gremlin-2 bound or captured to a solid surface or bead. Phage used in these methods are typically filamentous phage including fd and Ml 3 binding domains expressed from phage with Fab, Fv (individual Fv region from light or heavy chains) or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene III or gene VIII protein. Details of the methods are set forth, for example, in Nieri P et al. (2009) Curr Med Chem 16:753:79. In another example, ribosomal display can be used to replace bacteriophage as the display platform. Cell surface libraries may be screened for antibodies. Such procedures provide alternatives to traditional hybridoma techniques for the isolation and subsequent cloning of monoclonal antibodies.
[00196] After phage selection, the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any other desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria. For example, techniques to recombinantly produce Fab, Fab' and F(ab')2 fragments can also be employed using methods known in the art.
Immunization and antibody production
[00197] The method of eliciting antibodies in a host animal involves administering an
effective amount of Gremlin-2 as antigens described above to the host animal (i.e., a suitable mammal such as a mouse, rabbit or guinea pig, or a suitable avian, such as a chicken) to elicit production of an antibody that specifically binds and inhibits Gremlin-2. Methods of immunizing animal, including the adjuvants used, booster schedules, sites of injection, suitable animals, etc. are well understood in the art, e.g., Harlow et al.
(Antibodies: A Laboratory Manual, First Edition (1988) Cold spring Harbor, N.Y.), and administration of living cells to animals has been described for several mammals and birds. Next, a population of antibody producing cells is generated. The population of cells is produced using hybridoma methods that well known to one of skill in the art (see, e.g., Harlow Antibodies: A Laboratory Manual, First Edition (1988) Cold Spring Harbor, N.Y.). Cells are fused to immortalized cells, such as myeloma cells or transformed cells, which are capable of replicating indefinitely in cell culture, thereby producing an immortal, immunoglobulin-secreting cell line. The immortal cell line utilized can be selected to be deficient in enzymes necessary for the utilization of certain nutrients. Many such cell lines (such as myelomas) are known to those skilled in the art, and include, for example:
thymidine kinase (TK) or hypoxanthine-guanine phosphoribosyl transferase (HGPRT). These deficiencies allow selection for fused cells according to their ability to grow on, for example, hypoxanthine-aminopterin-thymidine (HAT) medium. In alterative embodiments, populations of cells expressing monoclonal antibodies may be made using phage display methods.
[00198] Anti-Gremlin-2 antibodies, including antigen binding fragments of anti-Gremlin-2 antibodies, may also be produced by genetic engineering. In this technique, as with the standard hybridoma procedure, antibody-producing cells are sensitized to the desired antigen or immunogen. The messenger RNA isolated from the immune spleen cells or hybridomas is used as a template to make cDNA using polymerase chain reaction (PCR) amplification. A library of vectors, each containing one heavy chain gene and one light chain gene retaining the initial antigen specificity, is produced by insertion of appropriate sections of the amplified immunoglobulin cDNA into the expression vectors. A
combinatorial library can be constructed by combining the heavy chain gene library with the light chain gene library. This results in a library of clones which co-express a heavy and light chain (resembling the Fab fragment or antigen binding fragment of an antibody molecule). The vectors that carry these genes are co-transfected into a host (e.g. bacteria, insect cells, mammalian cells, or other suitable protein production host cell.). When antibody gene synthesis is induced in the transfected host, the heavy and light chain proteins self-assemble to produce active antibodies that can be detected by screening with the antigen or immunogen.
Phage panning and screening
[00199] Once the population of antibody-producing cells or phages is produced, the
antibodies are screened using one or a combination of a variety of assays. In general, these assays are functional assays, and may be grouped as follows: assays that detect an antibody's binding affinity or specificity, and assays that detect the ability of an antibody to initialize or inhibit a process.
[00200] For example, the antigen (e.g. Gremlin-2 polypeptide) is coupled to beads or wells or other solid support and incubated with phage displaying the antibody of interest. After washings, bound phage is then recovered by inoculation of log phase E. coli cells. The cells are grown and expanded with helper phage. Steps are repeated for the amplification of tightly bound phages. The phage-infected E. coli colonies after several round of enrichment are harvested and Fab antibodies are purified from the periplasmic fractions. The purified antibodies are then analyzed in accordance with methods known in the art. Certain exemplary examples are detailed below.
[00201] The population of antibody isolated from phage-infected cells or hybridomas is further analyzed and/or screened for binding to a single antigen (i.e., antigens that are not mixed with other antigens of the plurality of antigens) of the plurality of antigens in vitro or in situ (e.g. on cells). Immuno specific binding may be carried out according to methods routine and known in the art. The immunoassays which can be used include, but are not limited to, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), "sandwich" immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, and protein A
immunoassays, to name but a few. See, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York, which is incorporated by reference herein in its entirety.
[00202] In addition to binding assays, the cells and antibodies may be screened based on the ability of the antibody in the supernatant to perform a specific function (e.g. modulation of Gremlin-2).
[00203] A subject anti-Gremlin-2 antibody may also be screened in vivo. The method
involves administering a subject antibody to an animal model for a disease or condition and determining the effect of the antibody on the disease or condition of the model animal. In vivo assays of the invention include controls, where suitable controls include a sample in the absence of the antibody. Generally, a plurality of assay mixtures is run in parallel with different antibody concentrations to obtain a differential response to the various concentrations. Typically, one of these concentrations serves as a negative control, i.e., at zero concentration or below the level of detection.
[00204] In some cases, a monoclonal antibody of interest is one that modulates, e.g.,
reduces, an undesired symptom in a non-human animal model disease or condition by at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 80%, at least about 90%, or more, when compared to a control in the absence of the antibody. In some cases, a monoclonal antibody of interest is one that increases muscle function in a non-human animal model disease by at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 80%, at least about 90%, at least about 2-fold, at least about 2.5-fold, at least about 5-fold, or more than 5-fold, when compared to a control in the absence of the antibody. In general, a monoclonal antibody of interest will cause an animal to be more similar to an equivalent animal that is not suffering from the disease or condition. Antibodies that have therapeutic value that have been identified using the methods and compositions of the invention are termed "therapeutic" antibodies. The effect of an anti-Gremlin-2 antibody can be determined by in vivo studies (e.g. non-human animal models) using methods known in the art.
[00205] Suitable non-human animal models include, e.g., the mdx mouse model for human
Duchenne Muscular Dystrophy; the mdx/utnr" mouse model of Duchenne Muscular Dystrophy; the BIO 14.6 cardiomyopathic hamster; a dysferlin mouse model of myopathy; a sarcoglycan-disrupted mouse model; a laminin a2-deficicient mouse model of congenital muscular dystrophy; other dystrophic mouse models; a non-human animal model (e.g., a mouse model or a rat model) for muscle disuse immobilization; a non-human animal model of muscle injury such as cardiotoxin-induced injury and regeneration model); and the like. See, e.g., Allamand and Campbell (2000) Hum. Mol. Genet. 9:2459; Grounds et al. (2006) Neurobiol. Dis. 31: 1; Powers et al. (2005) Am. J. Physiol. Regul. Integr. Comp. Physiol. 288:R337; Musacchia et al. (1988) Exercise & Sport Sciences Review 16:61.
[00206] Selected monoclonal antibodies of interest can be expanded in vitro, using routine tissue culture methods, or in vivo, using mammalian subjects. For example, pristane- primed mice can be inoculated with log phase hybridoma cells in phosphate-buffered saline (PBS) for ascites production. Ascites fluid can be stored at -70° C prior to further purification.
Nucleic acid encoding the antibody
[00207] Cell expressing a monoclonal antibody of interest contains the immunoglobulin heavy and light chain-encoding expression cassettes. As such, the nucleic acids encoding the monoclonal antibody of interest may be identified. Accordingly, the subject nucleic acids may be identified by a variety of methods known to one of skill in the art. Similar methods are used to identify host cell cultures in monoclonal antibody production using hybridoma technology (Harlow et al., Antibodies: A Laboratory Manual, First Edition (1988) Cold spring Harbor, N.Y.), and rely on an "addressable" host cell and an
"addressable" monoclonal antibody, such that once a monoclonal antibody of interest is identified, a host cell address may be determined and the nucleic acid encoding the antibody of interested isolated from the cell.
[00208] The nucleic acids encoding a monoclonal antibody of interest may be recovered, characterized and manipulated from a cell expressing the antibody using techniques familiar to one of skill in the art (Ausubel, et al, Short Protocols in Molecular Biology, 3rd ed., Wiley & Sons, (1995) and Sambrook, et al, Molecular Cloning: A Laboratory
Manual, Third Edition, (2001) Cold Spring Harbor, N.Y.).
COMPOSITIONS
[00209] The present disclosure provides compositions comprising an anti-Gremlin-2
antibody, which may be administered to a subject in need of an increase in muscle function and/or muscle mass.
[00210] A subject antibody composition can contain, in addition to a subject antibody, one or more of: a salt, e.g., NaCl, MgCl, KC1, MgS04, etc.; a buffering agent, e.g., a Tris buffer, N-(2-Hydroxyethyl)piperazine-N'-(2-ethanesulfonic acid) (HEPES), 2-(N- Morpholino)ethanesulfonic acid (MES), 2-(N-Morpholino)ethanesulfonic acid sodium salt (MES), 3-(N-Morpholino)propanesulfonic acid (MOPS), N-tris[Hydroxymethyl]methyl-3- aminopropanesulfonic acid (TAPS), etc.; a solubilizing agent; a detergent, e.g., a non-ionic detergent such as Tween-20, etc.; a protease inhibitor; glycerol; and the like.
[00211] Compositions comprising a subject antibody may include a buffer, which is
selected according to the desired use of the protein, and may also include other substances appropriate to the intended use. Those skilled in the art can readily select an appropriate buffer, a wide variety of which are known in the art, suitable for an intended use.
[00212] The composition may comprise a pharmaceutically acceptable excipient, a variety of which are known in the art and need not be discussed in detail herein. Pharmaceutically acceptable excipients have been amply described in a variety of publications, including, for example, "Remington: The Science and Practice of Pharmacy", 19 Ed. (1995), or latest edition, Mack Publishing Co; A. Gennaro (2000) "Remington: The Science and Practice of Pharmacy", 20th edition, Lippincott, Williams, & Wilkins; Pharmaceutical Dosage Forms and Drug Delivery Systems (1999) H.C. Ansel et al., eds 7th ed., Lippincott, Williams, &
Wilkins; and Handbook of Pharmaceutical Excipients (2000) A.H. Kibbe et al., eds., 3 rd ed. Amer. Pharmaceutical Assoc.
[00213] A subject pharmaceutical composition can comprise an anti-Gremlin-2 antibody, and a pharmaceutically acceptable excipient. In some cases, a subject pharmaceutical composition will be suitable for injection into a subject, e.g., will be sterile. For example, in some embodiments, a subject pharmaceutical composition will be suitable for injection into a human subject, e.g., where the composition is sterile and is free of detectable pyrogens and/or other toxins.
[00214] A subject antibody composition may comprise other components, such as
pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium, carbonate, and the like. The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions such as pH adjusting and buffering agents, toxicity adjusting agents and the like, for example, sodium acetate, sodium chloride, potassium chloride, calcium chloride, sodium lactate, hydrochloride, sulfate salts, solvates (e.g., mixed ionic salts, water, organics), hydrates (e.g., water), and the like.
[00215] For example, compositions may include aqueous solution, powder form, granules, tablets, pills, suppositories, capsules, suspensions, sprays, suppositories, and the like. The composition may be formulated according to the different routes of administration described later below.
[00216] Where the antibody is administered as an injectable (e.g. subcutaneously,
intraperitoneally, and/or intravenous) directly into a tissue, a formulation can be provided as a ready-to-use dosage form, or as non-aqueous form (e.g. a reconstitutable storage- stable powder) or aqueous form, such as liquid composed of pharmaceutically acceptable carriers and excipients. The antibody-containing formulations may also be provided so as to enhance serum half-life of the subject protein following administration. For example, the antibody may be provided in a liposome formulation, prepared as a colloid, or other conventional techniques for extending serum half-life. A variety of methods are available for preparing liposomes, as described in, e.g., Szoka et al. 1980 Ann. Rev. Biophys. Bioeng. 9:467, U.S. Pat. Nos. 4,235,871, 4,501,728 and 4,837,028. The preparations may also be provided in controlled release or slow-release forms.
[00217] Other examples of formulations suitable for parenteral administration include
isotonic sterile injection solutions, anti- oxidants, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. For example, a subject pharmaceutical composition can be present in a container, e.g., a sterile container, such as a syringe. The formulations can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described.
[00218] The concentration of a subject antibody in a formulation can vary widely (e.g., from less than about 0.1%, usually at or at least about 2% to as much as 20% to 50% or more by weight) and will usually be selected primarily based on fluid volumes, viscosities, and patient-based factors in accordance with the particular mode of administration selected and the patient's needs.
METHODS OF SCREENING
[00219] A screening method of the present disclosure can be employed to screen for a
agents that bind Gremlin-2, e.g., a binding agent that down regulates (e.g., inhibits or neutralizes) Gremlin-2 activity. The method can involve contacting a Gremlin-2 polypeptide with a candidate agent and detecting binding of the candidate agent with Gremlin-2. The method may also involve contacting a Gremlin-2 polypeptide with a candidate agent in the presence or absence of one or more known binding partners of Gremlin-2 and detecting the effect of the candidate agent on binding of Gremlin-2 with the binding partner. The method may also involve the use of libraries of constructs encoding antibodies, aptamers, and/or libraries of small molecules to screen for a Gremlin-2-binding agent. The binding agent may be selected for its potent inhibition of Gremlin-2 activities, inhibition of the expression of mature Gremlin-2, and/or inhibition of the binding affinity for Gremlin-2-interacting proteins. The method may be executed according to methods known in the art.
[00220] Briefly, Gremlin-2 (e.g. Gremlin-2 alone or Gremlin-2 complexed with its
interacting molecules) is contacted with a candidate agent. The binding of the candidate agent to Gremlin-2 is measured to see if there is a binding affinity for Gremlin-2. The ability of the candidate agent to disrupt Gremlin-2 binding to its interacting molecules is also assessed. Candidate agents that are effective in disrupting binding of Gremlin-2 to its interacting molecules are selected to be potential agents to be used in diagnostic and therapeutic compositions and methods of use. Candidate agents that can disrupt binding of Gremlin-2 to its interacting molecules encompass those that can decrease the binding affinity of Gremlin-2 to its interacting partners either competitively or noncompetitively.
[00221] Gremlin-2 that may be used to screen for potential agent include Gremlin-2 as described above. Examples of Gremlin-2 to be used in the subject screening methods include but are not limited to full-length Gremlin-2, mature Gremlin-2, fragments of Gremlin-2 (e.g., a binding partner-binding fragment of Gremlin-2), Gremlin-2 alone, or Gremlin-2 bound to one or more interacting molecules.
[00222] In an example of a screening method, Gremlin-2 may be immobilized on an ELISA plate or on beads through a covalent or non-covalent interaction, such as hydrophobic adsorption, biotin-avidin interaction, and Ni2+-6xHis interaction. A population of candidate agents is then incubated with the immobilized Gremlin-2, washed, and recovered. During selection, the bound candidate is recovered and identified. Multiple successive selection rounds ensure a selection of a candidate that acts as a specific binding agent for Gremlin-2. Other methods such as surface plasmon resonance, western blot, functional assays (e.g. phosphorylation or dephosphorylation of downstream targets), fluorescence activated cell sorting, etc. can also be used to screen and select for agents that can bind Gremlin-2, and/or inhibit its interaction to one or more interacting molecules. Other assays known in the art that involve comparing binding and/or activity of Gremlin-2 in the presence or absence of the candidate agents can be employed.
[00223] Candidate Gremlin-2-binding agents may also be engineered so that the agent
contains sites that are known to have affinity for the ligand-binding site (e.g. Gremlin-2 domain homologous to the Gremlin-2-binding site). KITS
[00224] Also provided by the present disclosure are kits for using the compositions
disclosed herein and for practicing the methods, as described above. The kits may be provided for administration of the subject protein in a subject in need of an increase in muscle function and/or muscle mass. The kit can include one or more of a Gremlin-2 polypeptide and/or an anti-Gremlin-2 antibody disclosed herein, which may be provided in a sterile container, and can be provided in formulation with a suitable a pharmaceutically acceptable excipient for administration to a subject. The proteins (e.g., an anti-Gremlin-2 antibody) can be provided with a formulation that is ready to be used as it is or can be reconstituted to have the desired concentrations. Where the proteins (e.g., an anti-Gremlin- 2 antibody) are provided to be reconstituted by a user, the kit may also provide buffers, pharmaceutically acceptable excipient, and the like, packaged separately from the subject protein. The proteins (e.g., an anti-Gremlin-2 antibody) of the present kit may be formulated separately or in combination with other drugs. Where an antibody is formulated separately with another drug, a subject kit can include: 1) a first container (e.g., a sterile container) comprising a subject pharmaceutical composition (e.g., a pharmaceutical composition comprising a subject anti-Gremlin-2 antibody); and 2) a second container (e.g., a sterile container) comprising a second agent (e.g., a second agent that treats a disease, disorder, or condition resulting in or associated with reduced muscle function and/or reduced muscle mass).
[00225] In addition to above-mentioned components, the kits can further include
instructions for using the components of the kit to practice the subject methods. The instructions for practicing the subject methods are generally recorded on a suitable recording medium. For example, the instructions may be printed on a substrate, such as paper or plastic, etc. As such, the instructions may be present in the kits as a package insert, in the labeling of the container of the kit or components thereof (i.e., associated with the packaging or subpackaging) etc. In other embodiments, the instructions are present as an electronic storage data file present on a suitable computer readable storage medium, e.g. CD-ROM, diskette, etc. In yet other embodiments, the actual instructions are not present in the kit, but means for obtaining the instructions from a remote source, e.g. via the internet, are provided. An example of this embodiment is a kit that includes a web address where the instructions can be viewed and/or from which the instructions can be downloaded. As with the instructions, this means for obtaining the instructions is recorded on a suitable substrate.
EXAMPLES
[00226] The following examples are put forth so as to provide those of ordinary skill in the art with a complete disclosure and description of how to make and use the present invention, and are not intended to limit the scope of what the inventors regard as their invention nor are they intended to represent that the experiments below are all or the only experiments performed. Efforts have been made to ensure accuracy with respect to numbers used (e.g. amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees Celsius, and pressure is at or near atmospheric. Standard abbreviations may be used, e.g., bp, base pair(s); kb, kilobase(s); pi, picoliter(s); s or sec, second(s); min, minute(s); h or hr, hour(s); aa, amino acid(s); kb, kilobase(s); bp, base pair(s); nt, nucleotide(s); i.m.,
intramuscular(ly); i.p., intraperitoneal(ly); s.c, subcutaneous (ly); and the like.
MATERIALS AND METHODS
Animals
[00227] Timed -pregnant C57BL/6 mice were purchased from the Charles River
Laboratory (Wilmington, MA). Mice were kept in accordance with welfare guidelines and project license restrictions under controlled light (12 hr light and 12 hr dark cycle, dark 6:30 pm-6:30 am), temperature (22±4°C) and humidity (50 +20 ) conditions. The mice had free access to water (autoclaved distilled water) and were fed ad libitum on a commercial diet (Harlan laboratories, Irradiated 2018 Teklad Global 18% Protein Rodent Diet) containing 17 kcal% fat, 23 kcal% protein and 60 kcal% carbohydrate. Three-day old neonates were injected with adeno-associated virus (AAV). The injected mice were weaned 3 weeks later and were maintained on 2018 Teklad Global diet containing
2000mg/kg of doxycyline to induce gene expression (Harlan Laboratories). In select AAV- injected mice, muscle-injury was induced by direct cardiotoxin (CTX) (Calbiochem) injection into one of the tibialis anterior muscles. Mdx mice were purchased from Jackson Laboratory (Bar Harbor, ME). Mdx mice were kept and maintained in similar conditions and diet as non-injected C57BL6 mice. All animal studies were approved by the NGM Institutional Animal Care and Use Committee for NGM- 12-2009 entitled
"Characterization of Biologies, Compounds and Viral Vectors for Treatment of Muscle Wasting Using Rodent Models".
DNA sequence
[00228] cDNA of ORF encoding murine Gremlin! (GenBank Accession No. NM_011825)
[00229] atgttctggaag ctctcgctga ccttgctcct ggtggctgtg ctggtaaag tagctgaaac aeggaagaac cggcctgcgg gcgccatccc ctcgccttac aaggatggta gcagcaacaa ctcagagagg tggcatcacc agatcaagga ggtgctggcc tccagccagg aggccctggt agtcaccgag cgcaagtacc tcaagagtga ctggtgcaag acgcagcctc tgcggcagac agtgagcgag gagggttgcc gcagccgcac catcctcaac cgcttctgct aeggecagtg caactccttc tacatcccgc gacacgtgaa gaaggaggag gactccttcc aatcctgcgc tttctgcaag ccccagcgtg tcacctctgt categtagag ctcgaatgcc egggtctega cccacctttc cgaatcaaga aaatccagaa ggtgaagcat tgccggtgca tgtcagtgaa cctgagtgac tccgacaagc agtga
[00230] Gremlin2 open reading frame (ORF) was amplified with polymerase chain reaction
(PCR) using recombinant DNA (cDNA) prepared from mouse testes. PCR reagent kits with Phusion high-fidelity DNA polymerase were purchased from New England BioLabs (F-530L, Ipswich, MA). The following primer sequences were used: forward PCR primer: 5' ATGTTCTGGAAGCTCTCGCTGACCTTGCTC and reverse PCR primer: 5'
TCACTGCTTGTCGGAGTCACTCAGGTTCACTG.
Polymerase chain reaction
[00231] PCR reactions were set up according to manufacturer's instructions, and the
amplified DNA fragment was digested with restriction enzymes Spe I and Not I (the restriction sites were included in the 5' or 3' PCR primers, respectively), to generate an amplification product. The amplification product was then ligated with AAV transgene vectors that had been digested with the same restriction enzymes. The vector used for expression contained a selectable marker and an expression cassette composed of tetracycline response elements flanked by minimal cytomegalovirus (CMV) promoter 5' of a site for insertion of the cloned coding sequence, followed by a 3' untranslated region and bovine growth hormone polyadenylation tail. The expression construct is also flanked by internal terminal repeats at the 5' and 3' ends. Alternatively, another vector used for tissue- selective expression contained the same regulatory elements as noted above, and a muscle- specific promoter. Production and purification of AAV
[00232] AAV 293 cells (obtained from Agilent Technologies, Santa Clara, CA) were
cultured in Dulbecco's Modification of Eagle's Medium (DMEM, Mediatech, Inc.
Manassas, VA) supplemented with 10% fetal bovine serum and lx antibiotic-antimycotic solution (Mediatech, Inc. Manassas, VA). The cells were plated at 50% density on day 1 in 150 mm cell culture plates and transfected on day 2, using calcium phosphate precipitation method, with the following 3 plasmids (20 μg/plate of each): AAV transgene plasmid, pHelper plasmids (Agilent Technologies) and AAV2/9 or AAV2/6 plasmid (Gao et al (2004) J. Virol. 78:6381). 48 hours after transfection, the cells were scraped off the plates, pelleted by centrifugation at 3000xg and resuspended in buffer containing 20 mM Tris pH 8.5, 100 mM NaCl and 1 mM MgCl2. The suspension was frozen in an alcohol dry ice bath and was then thawed in a 37°C water bath. The freeze and thaw cycles were repeated for a total of three times; benzonase (Sigma-Aldrich, St. Louis, MO) were added to 50 units/ml; deoxycholate were added to a final concentration of 0.25%. After an incubation at 37 °C for 30 min, cell debris was pelleted by centrifugation at 5000 x g for 20 min. Viral particles in the supernatant were purified using a discontinuous iodixanol (Sigma-Aldrich, St. Louis, MO) gradient as previously described (Zolotukhin S. et al (1999) Gene Ther. 6:973). The viral stock was concentrated using Vivaspin 20 (MW cutoff 100,000 Dalton, Sartorius Stedim Biotech, Aubagne, France) and re-suspended in phosphate buffered saline (PBS) with 10% glycerol and stored at -80 °C. To determine the viral genome copy number, 2 μΐ of viral stock was incubated in 6 μΐ of solution containing 50 units/ml benzonase, 50 mM Tris-HCl pH 7.5, 10 mM MgCl2 and 10 mM CaCl2 for at 37°C for 30 minutes.
[00233] Afterwards, 15 μΐ of the solution containing 2 mg/ml of Proteinase K, 0.5% sodium dodecyl sulfate (SDS) and 25 mM ethylenediaminetetraacetic acid (EDTA) were added and the mixture was incubated for additional 20 min at 55 °C to release viral DNA. Viral DNA was cleaned with mini DNeasy Kit (Qiagen, Valencia, CA) and eluted with 40 μΐ of water. Viral genome copy (GC) was determined by using quantitative PCR. Viral stock was diluted with PBS to the desired GC/ml. 50 μΐ of viral working solution was delivered into neonates via intraperitoneal injection or in adult mice via intramuscular injection. Grip Strength Test
[00234] Grip strength measurements were performed in adult mice at 6, 10 and 14 weeks of age. Briefly, each mouse was held by the tail and allowed to grasp the metallic mesh of the digital grip strength meter (Columbus Instruments International Corporation, Columbus OH, USA). After the mouse grip had been established, the tail was gently pulled away from the mesh until the test animal's grip was broken. The force measured upon release was recorded as peak tension in grams. The test was repeated 10 consecutive times for the same mouse. Data are represented as the average peak tension per test animal. All test subjects were blinded prior to test administration.
Magnetic resonance imaging (MRI)
[00235] Body composition measurements were performed in adult mice at 6, 10, and 14 weeks of age using the Echo MRI whole body composition analyzer (Echo Medical Systems, Houston, TX, USA). Briefly, a mouse was individually placed in a designated holder. The holder is then inserted into the MRI device for analyses. Following ~ 1 minute reading time, the mouse was then released and the test was complete. Each mouse in a group of 10 was analyzed. Data collected for these analyses include total body weight, lean mass and fat mass.
Muscle Physiology
[00236] Intrinsic contractile properties of the skeletal muscle were evaluated using muscle physiology assay performed using 1305 5N In Situ Muscle Test System (Aurora Scientific Incorporated, Aurora, ON, Canada). One of the assays used was the measurement of maximum tetanic force generated by specific skeletal muscle group in live animals.
Briefly, the mouse injected with control virus or virus expressing the target molecules was placed under inhaled isofluorane. The hind leg designated for this study was shaved and disinfected. The mouse was placed on a heated platform contained within a physiology apparatus that is capable of maintaining body temperature. In addition, a thermometer was placed in the test mouse to closely monitor its body temperature throughout the procedure. The animal was secured by keeping the knee stationary and the foot firmly fixed to a footplate. The knee was secured by inserting a 25 gauge needle directly underneath the knee bone. The inserted needle was firmly fixed onto a clamp ensuring the stability of the knee throughout the procedure. Muscle contraction on the secured hind leg of the test animal was elicited by electrical stimulation of the common peroneal nerve. To access the common peroneal nerve, a Teflon coated monopolar electrode was externally inserted through the skin on either side of the tibialis anterior muscle (TA). The proximal end of the wire was connected to an electrical stimulator. To determine the maximum tetanic force generated by the TA muscle, the nerve was stimulated at IHz (twitch), lOHz, 20Hz, 40Hz, 60Hz, 80Hz, 100Hz and 150Hz for 500ms with 30 second pause between tetanus. Data per n=5 are represented as force (N.cm) at various frequency from twitch (IHz) until 150Hz, the frequency where full tetanization is achieved.
Gremlin-2 Expression Level
[00237] Gene expression level for Gremlin-2 was determined by a quantitative PCR (Q-
PCR) approach. Briefly, total RNA was isolated from skeletal muscle following manufacturer's RNA protocol for using Trizol reagent (Invitrogen, Carlsbad, CA, USA). Reverse transcription reaction was performed following the protocol outlined from iScript cDNA synthesis kit from Biorad (Hercules, CA, USA). The Gremlin-2 primer pair was obtained from Applied Biosystems (Carlsbad, CA, USA) as FAM labeled Gene Expression Assay kit (Cat#: MN00501909_ml). Glyceraldehyde-3-phosphate dehydrogenase
(GAPDH) was used as an internal control gene. A primer pair for Gapdh was purchased as VIC labeled Gene Expression Assay kit (Cat#: 4352339E) from Applied Biosystems. 384- well Q-PCR reactions were set-up using 2x QuantiTect Multiplex RT-PCR Master Mix (Qiagen, Valencia, CA, USA) and performed on a 7900HT Fast Real-Time PCR System from Applied Biosystems (Carlsbad, CA, USA). Data are represented as fold expression relative to Gapdh control.
[00238] Gremlin-2 protein expression level was determined using the ELISA kit obtained from R&D Systems (Minneapolis, MN, USA). Samples analyzed include sera collected from Gremlin-2 injected mice as well as protein lysate prepared from skeletal muscle tissue isolated.
[00239] To reduce the overall gene expression level of Gremlin-2 in mice, a loss-of- function approach in vivo was adapted. To achieve a targeted inactivation of Gremlin-2 expression, short-hairpin RNA (shRNA) sequences were designed and cloned into the AAV vector. The following shRNA sequences were used to specifically target Gremlin-2:
[00240] 5'-
TGCTGTTGACAGTGAGCGCGGAGTCACTAGGAAGCTGTAATAGTGAAGCCACA GATGTATTAC AGCTTCCTAGTG ACTCCTTGCCTACTGCCTCGG A- 3 ' ;
[00241] 5'-
TGCTGTTGACAGTGAGCGCCGGGTTTCAACATTTCTTAATTAGTGAAGCCACA GATGTAATTAAGAA ATGTTGAA ACCCGTTGCCTACTGCCTCGGA-3 ' ; and [00242] 5'-
TGCTGTTGACAGTGAGCGAGCTACTTCTTGGCTTATTGTATAGTGAAGCCACAG ATGTATACAATAAGCC AAGAAGTAGCCTGCCTACTGCCTCGGA-3 ' .
Serum Analyses
[00243] Sera from mice injected with Gremlin-2 were measured for levels of various
metabolic substrates and enzyme to determine overall effect on test animals. Levels of inorganic phosphate, calcium, iron, cholesterol, low-density lipoprotein (LDL), non- esterified fatty acids (NEFA), triglycerides, and glucose were determined for sera collected from control-injected and Gremlin-2 injected animals. In addition, levels of the enzymes aspartate transaminase (AST) and alanine transaminase (ALT) were determined. All tests were measured using Cobas Integra 400 Plus Clinical Chemistry system (Roche
Diagnostics, IN, USA).
RESULTS
[00244] The results are shown in Figures 1-14.
[00245] In Figure 1, "GFP" refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery, and "Gremlin-2" to wild-type mice injected with lxlOEl l GC of rAAV expressing mouse Gremlin-2 via neonate intraperitoneal gene delivery (n = 10 mice per group). To determine the circulating level of Gremlin-2, an ELISA was performed using sera obtained from GFP-injected mice and from mice with Gremlin-2 over-expression. The level of Gremlin-2 in GFP-injected mice was 1-2 ng/ml. In contrast, a 10-20 fold increase was observed in the serum level upon Gremlin-2 over-expression (Figure 1). Thus, the endogenous Gremlin-2 serum level is relatively low and in vivo over-expression using AAV substantially increases the level of circulating Gremlin-2.
[00246] In Figure 2, "GFP" refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery, and "Gremlin-2" ("Grem2") to wild-type mice injected with lxlOEl 1 GC of rAAV expressing mouse Gremlin-2 via neonate intraperitoneal gene delivery (n = 10 mice per group).
Gremlin-2 is an important factor in embryo development. To assess the overall effect of Gremlin-2 over-expression, grip strength tests were performed at varying time points in a growing mouse. Tests were performed at 3 weeks, 7 weeks, and 11 weeks after the induction of Gremlin-2 over-expression. Mice performance in the grip strength test showed a marked decrease in peak tension upon Gremlin-2 over-expression when compared to GFP injected mice.
[00247] In Figure 3, "GFP" refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery, and "Gremlin-2" ("Grem2") to wild-type mice injected with lxlOEl l GC of rAAV expressing mouse Gremlin-2 via neonate intraperitoneal gene delivery (n = 10 mice per group). To further characterize additional phenotypes associated with Gremlin-2 over-expression, body composition measurements by magnetic resonance imaging (MRI) were also performed at 3 weeks, 7 weeks, and 11 weeks post-Gremlin-2 over-expression. The parameters measured in this procedure include total body weight as well as total lean tissue and total fat tissue mass. These analyses revealed that over-expression of Gremlin-2 decreases overall lean mass when compared to GFP-injected mice. This effect was observed at 7 weeks post-Gremlin-2 induction and 11 weeks following Gremlin-2 over- expression (Figure 3). A modest increase in fat mass was observed in the same group of mice. The overall body weight between GFP controls and Gremlin-2 over-expressing mice showed no significant difference.
[00248] In Figure 4, "GFP" refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery, and "Gremlin-2" ("Grem2") to wild-type mice injected with lxlOEl 1 GC of rAAV expressing mouse Gremlin-2 via neonate intraperitoneal gene delivery (n = 10 mice per group). To evaluate the effects of Gremlin-2 over-expression on skeletal muscle which comprise most of the lean tissue mass, tibialis anterior (TA) muscles were isolated and weighed. These analyses showed that TA muscles isolated from mice over-expressing Gremlin-2 are smaller in weight when compared to age-matched GFP- injected controls (Figure 4).
Tissue weight analyses of various organs from Gremlin-2 and GFP expressing mice showed no significant difference (Figure 6). Thus, Gremlin-2 over-expression appears to selectively affect skeletal muscle.
[00249] In Figure 5, "GFP" refers to wild-type 10-12 week old mice that were
intramuscularly injected with 5xlOE10 GC of rAAV expressing green fluorescent protein, and "Gremlin-2" ("Grem2") to wild- type intramuscularly injected with 5xlOE10 GC of rAAV expressing mouse Gremlin-2 (n = 5 mice per group). To evaluate the potential effects of Gremlin-2 on skeletal muscle, the maximum force generated by the tibialis anterior (TA) muscle was measured by tetanic force stimulation in situ. This analysis showed that Gremlin-2 directly contributes to the decrease in maximum tetanic force generated by TA muscle. No significant difference was observed between Gremlin-2 and GFP groups at low frequency stimulations. Interestingly, at a higher stimulation frequencies (60Hz, 80Hz, 100Hz and 150Hz), Gremlin-2 expression results in decreased tetanic force (represented by N.cm) generated by TA muscle. Thus, Gremlin-2 over- expression directly impacts skeletal muscle contraction and/or function.
[00250] In Figure 6, "GFP" refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery, and "Gremlin-2" ("Grem2") to wild-type injected with lxlOEl l GC of rAAV expressing mouse Gremlin-2 via neonate intraperitoneal gene delivery (n = 10 mice per group). Various tissues, including liver, intestine, kidneys, brain, heart, lung, spleen, and testis, were isolated and measured for weight comparison. No significant differences were observed between Gremlin-2 and GFP groups (Figure 6). This observation may indicate that Gremlin-2 has no significant effect on other soft tissues weights.
[00251] In Figure 7, "GFP" refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery, and "Gremlin-2" ("Grem2") to wild-type injected with lxlOEl 1 GC of rAAV expressing mouse Grem 2 via neonate intraperitoneal gene delivery (n = 10 mice per group). Serum levels of inorganic phosphate (PHOS), calcium (CA), iron, cholesterol (CHOL), LDL, NEFA, triglyceride (TRIGL), glucose (GLU) (indications of metabolic homeostasis), liver AST, and liver ALT (indications of liver functions) were determined for sera collected from both groups. As seen in Figure 7, no significant differences were identified between Gremlin-2 and GFP groups, indicating Gremlin-2 has no significant impact on general metabolism and liver function.
[00252] To determine Gremlin-2 expression levels in dystrophic muscles, muscle tissues were collected from both wild type and mdx mice. mRNA levels of Gremlin-2 were determined by quantitative PCR. The data are shown in Figure 8. This analysis revealed elevated expression levels of Gremlin-2 in mdx mice.
[00253] To determine Gremlin-2 protein levels dystrophic muscles, sera and muscle tissue samples were collected from both wild type and mdx mice. Protein levels of Gremlin-2 were determined by ELISA. The data are shown in Figure 9. In these sets of analyses, a significant elevation in Gremlin-2 protein in both muscle and serum was observed in mdx mice.
[00254] To determine Gremlin-2 expression levels in injured muscles, muscle tissues were collected from both wild type and Cardiotoxin (CTX) injected mice. mRNA levels of Gremlin-2 were determined by quantitative PCR. The data are shown in Figure 10. This analysis revealed elevated expression levels of Gremlin-2 in CTX injured muscle tissues.
[00255] To determine Gremlin-2 protein levels injured muscles, sera and muscle tissue samples were collected from both wild type and Cardiotoxin (CTX) injected mice. Protein levels of Gremlin-2 were determined by ELISA. The data are shown in Figure 11. In these sets of analyses, a significant elevation in Gremlin-2 protein in both CTX injured muscle and serum was observed.
[00256] In Figure 12, "GFP" refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery, and "Gremlin-2 shRNA" ("Grem2 shRNA") to wild-type injected with lxlOEl l GC of rAAV mixture expressing 3 mouse Gremlin-2 shRNAs via neonate intraperitoneal gene delivery (n = 10 mice per group). To determine the effect of Gremlin-2 shRNA expression on endogenous Gremlin-2, muscle tissues were collected from both GFP and Gremlin-2 shRNA injected mice. mRNA levels of Gremlin-2 were determined by quantitative PCR. Using shRNA sequences against Gremlin-2, a significant decrease in Gremlin-2 levels in skeletal muscle was observed. To determine whether this effect also results in the reduction of Gremlin-2 protein levels, an ELISA was performed. Consistent with the reduction in mRNA levels, the use of shRNA to target Gremlin-2 also results in the reduction of Gremlin-2 protein level in skeletal muscle.
[00257] In Figure 13, "GFP" refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery, and "Gremlin-2 shRNA" ("Grem2 shRNA") to wild-type injected with lxlOEl 1 GC of rAAV mixture expressing 3 mouse Gremlin-2 shRNAs via neonate intraperitoneal gene delivery (n = 10 mice per group). The levels of Gremlin-2 in skeletal muscle were reduced upon expression of Gremlin-2 shRNA. To determine the effects of reducing Gremlin-2 expression on muscle function, a grip strength test was performed at 11 weeks after the induction of Gremlin-2 shRNA expression. Mouse performance in grip strength test showed a marked increase in peak tension upon Gremlin-2 shRNA expression when compared to GFP injected mice.
[00258] In Figure 14, "GFP" refers to wild-type mice injected with lxlOEl 1 GC of rAAV expressing green fluorescent protein via neonate intraperitoneal gene delivery, and "Gremlin-2 shRNA" ("Grem2 shRNA") to wild-type injected with lxlOEl l GC of rAAV mixture expressing 3 mouse Gremlin-2 shRNAs via neonate intraperitoneal gene delivery (n = 10 mice per group). To determine additional phenotypes associated with a reduced Gremlin-2 levels in Gremlin-2 shRNA injected mice, body composition measurements by MRI were performed at 11 weeks post-shRNA expression. These measurements revealed that expression of Gremlin-2 shRNA results in an increase in the overall lean mass and body weight when compared to GFP-injected mice. No changes in fat mass were observed in both GFP controls and Gremlin-2 shRNA injected mice.
[00259] While the present invention has been described with reference to the specific embodiments thereof, it should be understood by those skilled in the art that various changes may be made and equivalents may be substituted without departing from the true spirit and scope of the invention. In addition, many modifications may be made to adapt a particular situation, material, composition of matter, process, process step or steps, to the objective, spirit and scope of the present invention. All such modifications are intended to be within the scope of the claims appended hereto.

Claims

CLAIMS What is claimed is:
1. A method of treating a subject, the method comprising:
administering to a subject having a deficiency in muscle function and/or reduced muscle mass a therapeutically effective amount of an antibody that specifically binds to a protein comprising an amino acid sequence having at least 90% amino acid sequence identity to an amino acid sequence of human Gremlin-2, wherein said administering is effective to increase muscle function and/or muscle mass in the subject.
2. The method of claim 1, wherein the deficiency in muscle function and/or reduced muscle mass is a sequela of immobilization, chronic disease, cancer, or injury.
3. The method of claim 1, wherein said subject is human.
4. The method of claim 1, wherein said administering is by intramuscular, intravenous, or subcutaneous injection.
5. The method of claim 1, wherein said antibody is a monoclonal antibody.
6. The method of claim 1, wherein said antibody is a humanized antibody.
7. A method of treating a subject, the method comprising:
administering to a subject having a having a deficiency in muscle function and/or reduced muscle mass a therapeutically effective amount of a short interfering nucleic acid (siNA) that specifically reduces synthesis of a protein comprising an amino acid sequence having at least 90% amino acid sequence identity to an amino acid sequence of human Gremlin-2, wherein said administering is effective to increase muscle function and/or muscle mass in the subject.
8. The method of claim 7, wherein the deficiency in muscle function and/or reduced muscle mass is a sequela of immobilization, chronic disease, cancer, or injury.
9. The method of claim 7, wherein said subject is human.
10. The method of claim 7, wherein said administering is by intramuscular, intravenous, or subcutaneous injection.
11. The method of claim 7, wherein said siNA is a short hairpin RNA.
12. The method of claim 7, wherein said siNA is administered in the form of a recombinant vector comprising a nucleotide sequence encoding the siRNA.
13. A pharmaceutical composition comprising.
a) a monoclonal antibody that binds specifically to a Gremlin-2 polypeptide comprising an amino acid sequence having at least 90% amino acid sequence identity to an amino acid sequence of human Gremlin-2; and
b) a pharmaceutically acceptable excipient.
14. The pharmaceutical composition of claim 13, wherein the antibody comprises a light chain variable region and a heavy chain variable region present in separate polypeptides.
15. The pharmaceutical composition of claim 13, wherein the antibody comprises a light chain variable region and a heavy chain variable region present in a single polypeptide.
16. The pharmaceutical composition of claim 13, wherein the antibody binds the
7 -1 12 -1
Gremlin-2 polypeptide with an affinity of from about 10 M" to about 10 M" .
17. The pharmaceutical composition of claim 13, wherein the antibody comprises heavy chain constant region, and wherein the heavy chain constant region is of the isotype IgGl, IgG2, IgG3, or IgG4.
18. The pharmaceutical composition of claim 13, wherein the antibody is a Fv, scFv, Fab, F(ab')2, or Fab'.
19. The pharmaceutical composition of claim 13, wherein the antibody comprises a covalently linked non-peptide synthetic polymer.
20. The pharmaceutical composition of claim 13, wherein the antibody comprises a covalently linked moiety selected from a lipid moiety, a fatty acid moiety, a polysaccharide moiety, and a carbohydrate moiety.
21. The pharmaceutical composition of claim 13, wherein the antibody is a humanized antibody.
22. The pharmaceutical composition of claim 13, wherein the excipient is an isotonic injection solution.
23. The pharmaceutical composition of claim 13, wherein the composition is suitable for human administration.
24. A sterile container comprising the pharmaceutical composition of claim 13.
25. The container of claim 24, wherein the container is a syringe.
26. A kit comprising the sterile container of claim 24.
27. The kit of claim 26, further comprising a second sterile container comprising a second therapeutic agent.
PCT/US2012/031630 2011-04-04 2012-03-30 Compositions and methods for increasing muscle function and mass WO2012138575A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161471616P 2011-04-04 2011-04-04
US61/471,616 2011-04-04

Publications (1)

Publication Number Publication Date
WO2012138575A1 true WO2012138575A1 (en) 2012-10-11

Family

ID=46969509

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/031630 WO2012138575A1 (en) 2011-04-04 2012-03-30 Compositions and methods for increasing muscle function and mass

Country Status (1)

Country Link
WO (1) WO2012138575A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050265978A1 (en) * 1998-05-01 2005-12-01 University Of Pittsburgh Muscle-derived cells (MDCs) for treating muscle- or bone-related injury or dysfunction
US20070154563A1 (en) * 2003-12-31 2007-07-05 Keyvan Behnam Bone matrix compositions and methods
US20100267633A1 (en) * 2005-09-26 2010-10-21 Barbara Hanney N-(4-Oxo-3,4-Dihydroquinazolin-2Yl) Butanamides as Androgen Receptor Modulators

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050265978A1 (en) * 1998-05-01 2005-12-01 University Of Pittsburgh Muscle-derived cells (MDCs) for treating muscle- or bone-related injury or dysfunction
US20070154563A1 (en) * 2003-12-31 2007-07-05 Keyvan Behnam Bone matrix compositions and methods
US20100267633A1 (en) * 2005-09-26 2010-10-21 Barbara Hanney N-(4-Oxo-3,4-Dihydroquinazolin-2Yl) Butanamides as Androgen Receptor Modulators

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
SUDO ET AL.: "Protein Related to DAN and Cerberus Is a Bone Morphogenetic Protein Antagonist That Particpates in Ovarian Paracrine Regulation", JBC, vol. 279, no. 22, 28 May 2004 (2004-05-28), pages 23134 - 23141 *

Similar Documents

Publication Publication Date Title
US10633662B2 (en) Methods and compositions for modulating AAV infection
US9150647B2 (en) Biological inhibitors of ROR1 capable of inducing cell death
JP2020504076A (en) Neutralization of anti-TL1A monoclonal antibody
JP6628787B2 (en) Antagonistic anti-OX40L antibodies and methods of using them
JP2015134782A (en) Treatment of autoimmune disease by modulating annexin-1 (lipocortin 1)
JP2019514994A (en) Compositions and methods for treating spinal muscular atrophy
CN105709224B (en) Compounds for the treatment of remyelination blockade in diseases associated with HERV-W envelope protein expression
JP7330174B2 (en) Methods and pharmaceutical compositions for modulating autophagy
JPWO2007049624A1 (en) Cancer preventive / therapeutic agent
US20180230469A1 (en) Method of treating pain with an antibody against netrin-4, unc5b or neogenin
JP5704722B2 (en) Cell adhesion inhibitor and use thereof
US10722571B2 (en) Rabies virus G protein epitope, and rabies virus neutralising binding molecule that binds specifically thereto
WO2011045202A1 (en) Selective antagonist or agonist of amhrii for modulating fertility
GB2526898A (en) Biological materials and therapeutic uses thereof
RU2744909C2 (en) Method for treatment or prevention of stroke
AU2008270218B2 (en) Compounds and methods for the treatment of renal disease
WO2012138575A1 (en) Compositions and methods for increasing muscle function and mass
EP3647426A1 (en) Fusion protein
WO2018187284A1 (en) Blood test to predict endurance athletic performance
JP2016079170A (en) Prophylactic/therapeutic agent for diseases associated with cell migration regulation and disease activity determination/prognosis evaluation of pulmonary interstitial disease
WO2010112034A2 (en) Compositions and methods for treatment and diagnosis of synucleinopathies
JPWO2012157589A1 (en) Cell adhesion inhibitor, cell growth inhibitor, and cancer test method and test kit
WO2023172989A2 (en) Epo receptor agonists and antagonists
CA3230295A1 (en) Method of treatment
US20130216546A1 (en) Methods of Treating Glucose Metabolism Disorders

Legal Events

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

Ref document number: 12768119

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12768119

Country of ref document: EP

Kind code of ref document: A1