HAIR GROWTH METHODS USING FGFR3 EXTRACELLULAR DOMAINS

The present invention relates to a method of promoting hair growth comprising administering a fibroblast growth factor receptor 3 (FGFR3) extracellular domain (ECD), including native FGFR3 ECDs, variants, fragments, and fusion molecules, to a subject in an amount sufficient to promote hair growth.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
TECHNICAL FIELD

This application is a continuation of U.S. application Ser. No. 13/515,429, which is a national stage application of International Application No. PCT/US10/61157, filed Dec. 17, 2010, which claims priority to U.S. Provisional Application No. 61/287,690, filed Dec. 17, 2009. Each of those applications is incorporated by reference herein in its entirety for any purpose.

The present invention relates to a method of promoting hair growth comprising administering an FGFR3 extracellular domain (ECD), including native FGFR3 ECDs, variants, fragments, and fusion molecules, to a subject in an amount sufficient to promote hair growth.

BACKGROUND AND SUMMARY

Hair growth problems are wide-spread. In addition to pattern baldness, which may occur in both males and females, hair loss can be induced by drugs, such as chemotherapy drugs, or by chemical or physical damage, such as by certain hair products or styling techniques. Hair loss may also be triggered by systemic diseases, autoimmune conditions, nutritional deficiencies and physical stress, such as during pregnancy, due to surgery, or due to weight loss. It may also be induced by psychological stress.

Available treatments to promote hair growth are limited. For example, minoxidil, while relatively safe, is only moderately effective. The 5-alpha reductase inhibitor finasteride is not indicated for women or children and has negative side effects. The use of certain polypeptides to promote hair growth has been suggested. (See e.g., U.S. Pat. No. 7,335,641, U.S. Pat. No. 7,524,505, U.S. Pat. No. 7485618, and U.S. Patent Application No. 2008/0139469.) To date, the only permanent solution to hair loss is hair transplant surgery, which is both expensive and invasive. Thus, there remains a need in the art for additional agents for promoting hair growth. The present disclosure relates to a method of promoting hair growth comprising administering a fibroblast growth factor receptor 3 (FGFR3) extracellular domain (ECD) to a subject in an amount sufficient to promote hair growth.

Fibroblast growth factors (FGFs) and their receptors (FGFRs) are a highly conserved group of proteins with diverse functions. The FGFR family comprises four major types of receptors, FGFR1, FGFR2, FGFR3, and FGFR4. To date, there are 22 known FGFs, each with the capacity to bind one or more FGFRs. See, e.g., Zhang et al., J. Biol. Chem. 281:15, 694-15,700 (2006). Each FGFR binds to several FGFs, and the different FGFRs may differ from each other both in the selection of FGFs to which they bind as well as in the affinity of those interactions.

The FGFRs are transmembrane proteins having an extracellular domain (ECD), a transmembrane domain, and an intracytoplasmic tyrosine kinase domain. Extracellular FGFR activation by FGF ligand binding to an FGFR initiates a cascade of signaling events inside the cell, beginning with oligomerization of the receptor and activation of receptor tyrosine kinase activity. Each of the ECDs contains either two or three immunoglobulin-like (Ig) domains. When there are three Ig domains, they are referred to as D1, D2, and D3 domains. Receptors having two Ig domains typically lack D1. An acidic motif, called the acid box, is located in the linker region between D1 and D2 in the FGFR extracellular domain. The acid box is believed to interact with the heparin binding site in the D2 domain. Structural studies of FGFR-FGF complexes have shown that FGF ligands interact extensively with the D2 domain, the D3 domain, and the linker region connecting the D2 and D3 domains of an FGFR ECD. In FGFR1-3, an alternative splicing event leads to three versions of the D3 domain, also called Ig domain III. The splice variants of this domain are referred to as domain Δ8-10, IIIb and IIIc. Domain III or D3 is encoded by three exons, two of which are alternatively spliced. Distinct splice variants of FGFR3 have been identified in a range of tissues and cancers, such as FGFR3 IIIb, FGFR3 IIIc, and FGFR Δ8-10 (lacking exons encoding the C-terminal half of Ig domain III and the transmembrane domain). See, e.g., Tomlinson et al., Cancer Res. 65: 10,441-10,449 (2005).

In experiments to determine whether an FGFR4 ECD exhibited antitumor activity in a cancer xenograft model, the inventors discovered that an FGFR4 ECD promoted hair growth at the shaved site where the tumor cells were injected. In contrast, an FGFR1 ECD did not promote visible hair growth. In subsequent experiments, both a native FGFR4 ECD fragment fusion molecule and an FGFR4 ECD variant fusion molecule (“ABMut1”) that retained FGFR4 ECD ligand binding activity promoted hair growth when administered systemically in mice. Experiments in which ABMut1 or agarose beads bound to ABMut1 were subcutaneously injected into the flank of shaved mice showed that local delivery of ABMut1 also promoted hair growth. Further experiments demonstrated that systemic delivery of ABMut1 could also induce anagen in hair follicles, specifically elongation of the dermal papilla into the fatty layer of the dermis. The inventors conducted similar studies with a native FGFR3 ECD fragment fusion molecule and discovered that the FGFR3 ECD fragment promoted hair growth when administered systemically in mice. In contrast, an FGFR2 ECD did not promote visible hair growth. Further experiments demonstrated that the FGFR3 ECD fragment fusion molecule could also induce anagen in hair follicles, specifically elongation of the dermal papilla into the fatty layer of the dermis, while the FGFR2 ECD did not have that effect. See Example 6, FIG. 3A and FIG. 3B. Yet further experiments demonstrate that the FGFR3 ECD fragment fusion molecule stimulates hair growth in a dose dependent manner. See Example 7, FIG. 4.

In certain embodiments, the invention provides a method of promoting hair growth comprising administering an FGFR3 ECD to a subject in an amount sufficient to promote hair growth. In certain embodiments, the FGFR3 ECD is a human FGFR3 ECD. In certain embodiments, the FGFR3 ECD is a non-human FGFR3 ECD. In certain embodiments, the FGFR3 ECD is a native FGFR3 ECD. In certain embodiments, the FGFR3 ECD is an FGFR3 ECD variant. In certain embodiments, the FGFR3 ECD is an FGFR3 ECD splice variant. In certain embodiments, the FGFR3 ECD comprises an Ig domain III chosen from Δ8-10, IIIb and IIIc (the FGFR3 ECD is also referred to as FGFR3-Δ8-10 ECD, FGFR3-IIIb ECD, or FGFR3-IIIc ECD). In certain embodiments, the FGFR3 ECD is an FGFR3 ECD fragment. In certain embodiments, the FGFR3 ECD is a native FGFR3 ECD fragment. In certain embodiments, the FGFR3 ECD is a variant of an FGFR3 ECD fragment. In certain embodiments, the FGFR3 ECD is a fragment of an FGFR3 ECD splice variant. In certain embodiments, the FGFR3 ECD is an FGFR3 ECD acidic region mutein. In certain embodiments, the FGFR3 ECD may be engineered to have a decrease in the total number of acidic residues within the D1-D2 linker. In certain embodiments, the FGFR3 ECD is an FGFR3 ECD D1-D2 linker chimera. In certain embodiments, the FGFR3 ECD D1-D2 linker chimera comprises a D1-D2 linker selected from an FGFR1 D1-D2 linker, an FGFR2 D1-D2 linker, and an FGFR4 D1-D2 linker, in place of the FGFR3 D1-D2 linker. In certain embodiments, the FGFR3 ECD is an FGFR3 ECD glycosylation mutant. In certain embodiments, the amino acid sequence of the FGFR3 ECD is at least 80% identical to SEQ ID NO: 4, 5, 6, or 30. In certain embodiments, the amino acid sequence of the FGFR3 ECD is at least 85% identical to SEQ ID NO: 4, 5, 6, or 30. In certain embodiments, the amino acid sequence of the FGFR3 ECD is at least 90% identical to SEQ ID NO: 4, 5, 6, or 30. In certain embodiments, the amino acid sequence of the FGFR3 ECD is at least 95% identical to SEQ ID NO: 4, 5, 6, or 30. In certain embodiments, the amino acid sequence of the FGFR3 ECD is at least 99% identical to SEQ ID NO: 4, 5, 6, or 30. In certain embodiments, the FGFR3 ECD comprises an amino acid sequence chosen from SEQ ID NOs: 4, 5, 6, or 30. In certain embodiments, the FGFR3 ECD comprises an amino acid sequence chosen from SEQ ID NOs: 34 and 36. In certain embodiments, the FGFR3 ECD lacks a signal sequence. In certain embodiments, the FGFR3 ECD comprises a signal sequence. In certain embodiments, the signal sequence is the native signal sequence of FGFR1, FGFR2, FGFR3, or FGFR4 (SEQ ID NOs: 19-22). In certain embodiments, the signal sequence is not an FGFR signal sequence, but from a heterologous protein.

In certain embodiments, the subject is a rodent, simian, human, feline, canine, equine, bovine, porcine, ovine, caprine, mammalian laboratory animal, mammalian farm animal, mammalian sport animal, or mammalian pet. In certain embodiments, the subject is a human. In certain embodiments, the administering is intravenous, subcutaneous, intraperitoneal, topical, or transdermal.

In certain embodiments, the invention provides a method of growing hair comprising administering an FGFR3 ECD fusion molecule to a subject in an amount sufficient to promote hair growth. In certain embodiments, the FGFR3 ECD fusion molecule comprises an FGFR3 ECD polypeptide and a fusion partner. In certain embodiments, the FGFR3 ECD polypeptide is a native FGFR3 ECD. In certain embodiments, the FGFR3 ECD polypeptide is an FGFR3 ECD variant. In certain embodiments, the FGFR3 ECD polypeptide is an FGFR3 ECD splice variant. In certain embodiments, the FGFR3 ECD polypeptide is FGFR3-Δ8-10 ECD, FGFR3-IIIb ECD, or FGFR3-IIIc ECD. In certain embodiments, the FGFR3 ECD polypeptide is an FGFR3 ECD fragment. In certain embodiments, the FGFR3 ECD polypeptide is a native FGFR3 ECD fragment. In certain embodiments, the FGFR3 ECD polypeptide is a variant of an FGFR3 ECD fragment. In certain embodiments, the FGFR3 ECD polypeptide is a fragment of an FGFR3 ECD splice variant. In certain embodiments, the FGFR3 ECD polypeptide is an FGFR3 ECD acidic region mutein. In certain embodiments, the FGFR3 ECD polypeptide may be engineered to have a decrease in the total number of acidic residues within the D1-D2 linker. In certain embodiments, the FGFR3 ECD polypeptide is an FGFR3 ECD D1-D2 linker chimera. In certain embodiments, the FGFR3 ECD D1-D2 linker chimera comprises a D1-D2 linker selected from an FGFR1 D1-D2 linker, an FGFR2 D1-D2 linker, and an FGFR4 D1-D2 linker, in place of the FGFR3 D1-D2 linker. In certain embodiments, the FGFR3 ECD polypeptide is an FGFR3 ECD glycosylation mutant. In certain embodiments, the amino acid sequence of the FGFR3 ECD polypeptide is at least 80% identical to SEQ ID NO: 4, 5, 6, or 30. In certain embodiments, the amino acid sequence of the FGFR3 ECD polypeptide is at least 85% identical to SEQ ID NO: 4, 5, 6, or 30. In certain embodiments, the amino acid sequence of the FGFR3 ECD polypeptide is at least 90% identical to SEQ ID NO: 4, 5, 6, or 30. In certain embodiments, the amino acid sequence of the FGFR3 ECD polypeptide is at least 95% identical to SEQ ID NO: 4, 5, 6, or 30. In certain embodiments, the amino acid sequence of the FGFR3 ECD polypeptide is at least 99% identical to SEQ ID NO: 4, 5, 6, or 30. In certain embodiments, the FGFR3 ECD polypeptide comprises an amino acid sequence chosen from SEQ ID NOs: 4, 5, 6, and 30. In certain embodiments, the FGFR3 ECD polypeptide comprises an amino acid sequence chosen from SEQ ID NOs: 34 and 36. In certain embodiments, the FGFR3 ECD polypeptide lacks a signal sequence. In certain embodiments, the FGFR3 ECD comprises a signal sequence. In certain embodiments, the signal sequence is the native signal sequence of FGFR1, FGFR2, FGFR3, or FGFR4 (SEQ ID NOs: 19-22). In certain embodiments, the signal sequence is not an FGFR signal sequence, but from a heterologous protein.

In certain embodiments, a method of growing hair comprising administering an FGFR3 ECD fusion molecule to a subject in an amount sufficient to promote hair growth is provided, wherein the fusion partner in the FGFR3 ECD fusion molecule is selected from an Fc, albumin, and polyethylene glycol. In certain embodiments, the fusion partner is an Fc. In certain embodiments, the FGFR3 ECD fusion molecule has an amino acid sequence chosen from SEQ ID NOs: 7-10. In certain embodiments, the FGFR3 ECD fusion molecule has an amino acid sequence chosen from SEQ ID NOs: 11-15, 28, 31-33, 35 and 37. In certain embodiments, the FGFR3 ECD fusion molecule has an amino acid sequence chosen from SEQ ID NO.: 9, 10 and 33. In certain embodiments, the FGFR3 ECD fusion molecule lacks a signal sequence. In certain embodiments, the FGFR3 ECD fusion molecule comprises a signal sequence. In certain embodiments, the signal sequence is the native signal sequence of FGFR1, FGFR2, FGFR3, or FGFR4. In certain embodiments, the signal sequence is not an FGFR signal sequence.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 shows an extracellular domain (ECD) amino acid sequence of an FGFR3 (SEQ ID NO: 29). The amino acid sequence in FIG. 1 includes the signal peptide, which is cleaved in the mature fusion protein. The numbers refer to the amino acid position, and certain domains within the ECD are illustrated in gray above the amino acid numbers. The amino acid positions within the signal peptide are given negative values because they are cleaved in the mature fusion protein. The first amino acid residue of the mature fusion protein is designated as amino acid position 1. The signal peptide and domains D1, D2, and D3 are noted in gray shading. The linker between the first and second Ig domains (referred to herein interchangeably as the “D1-D2 linker,” “FGFR3 ECD D1-D2 linker” and “FGFR3 ECD D1-D2 linker region”) and the linker between the second and third Ig domains (referred to herein interchangeably as the “D2-D3 linker,” “FGFR3 ECD D2-D3 linker,” and “FGFR3 ECD D2-D3 linker region”) are illustrated in a darker gray.

FIG. 2 shows an amino acid sequence alignment of a portion of the native extracellular domains (ECDs) of the FGFR isoforms FGFR3-Δ8-10, FGFR3-IIIb, FGFR3-IIIc, FGFR1-IIIb, FGFR1-IIIc, FGFR2-IIIb, FGFR2-IIIc, and FGFR4, denoting the immunoglobulin (Ig) domain III.

FIG. 3A shows that systemic delivery of an FGFR3 ECD-Fc (SEQ ID NO: 33) and ABMut1 (SEQ ID NO: 31, an FGFR4 ECD variant-Fc), promotes a substantial amount of clearly visible hair growth by 14 days post-dose, compared to vehicle-treated mice and mice treated with an FGFR2 ECD-Fc (SEQ ID NO: 32) by 14 days post-dose.

FIG. 3B shows that systemic delivery of an FGFR3 ECD-Fc (SEQ ID NO: 33) and ABMut1 (SEQ ID NO: 31, an FGFR4 ECD variant-Fc) induces anagen in hair follicles of eight-week-old mice. Shown are paraffin-embedded skin biopsies stained with Haematoxylin/Eosin from mice treated with vehicle, the FGFR3 ECD-Fc, an FGFR2 ECD-Fc or ABMut1 at days 14 post-dose. By day 14, mice treated with the FGFR3 ECD-Fc or ABMut1 showed an elongation of the dermal papilla (*) into the fatty layer of the dermis (), showing that a single dose of the FGFR3 ECD-Fc or ABMut1 can induce anagen (the growth phase of the hair cycle).

FIG. 4 shows that an FGFR3 ECD-Fc stimulates hair growth in mice in a dose dependent manner.

DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS

The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described.

Definitions

Unless otherwise defined, scientific and technical terms used in connection with the present invention shall have the meanings that are commonly understood by those of ordinary skill in the art. Further, unless otherwise required by context, singular terms shall include pluralities and plural terms shall include the singular.

Certain techniques used in connection with recombinant DNA, oligonucleotide synthesis, tissue culture and transformation (e.g., electroporation, lipofection), enzymatic reactions, and purification techniques are known in the art. Many such techniques and procedures are described, e.g., in Sambrook et al. Molecular Cloning: A Laboratory Manual (2d ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989)), among other places. In addition, certain techniques for chemical syntheses, chemical analyses, pharmaceutical preparation, formulation, delivery, and treatment of patients are also known in the art.

In this application, the use of “or” means “and/or” unless stated otherwise. In the context of a multiple dependent claim, the use of “or” refers back to more than one preceding independent or dependent claim in the alternative only. Also, terms such as “element” or “component” encompass both elements and components comprising one unit and elements and components that comprise more than one subunit unless specifically stated otherwise.

As utilized in accordance with the present disclosure, the following terms, unless otherwise indicated, shall be understood to have the following meanings:

The terms “nucleic acid molecule” and “polynucleotide” may be used interchangeably, and refer to a polymer of nucleotides. Such polymers of nucleotides may contain natural and/or non-natural nucleotides, and include, but are not limited to, DNA, RNA, and PNA.

The terms “polypeptide” and “protein” are used interchangeably, and refer to a polymer of amino acid residues. Such polymers of amino acid residues may contain natural and/or non-natural amino acid residues, and include, but are not limited to, peptides, oligopeptides, dimers, trimers, and multimers of amino acid residues. The terms “polypeptide” and “protein” include natural and non-natural amino acid sequences, and both full-length proteins and fragments thereof. Those terms also include post-translationally modified polypeptides and proteins, including, for example, glycosylated, sialylated, acetylated, and/or phosphorylated polypeptides and proteins.

The terms “acidic amino acid,” “acidic amino acid residue,” and “acidic residue” are used interchangeably herein and refer to an amino acid residue that is negatively charged at physiological pH. Acidic amino acids include, but are not limited to, aspartic acid (Asp, D) and glutamic acid (Glu, E).

The terms “non-acidic amino acid,” “non-acidic amino acid residue,” and “non-acidic residue” are used interchangeably and refer to an amino acid residue that is not negatively charged at physiological pH.

The terms “conservative amino acid substitutions” and “conservative substitutions” are used interchangeably herein to refer to intended amino acid swaps within a group of amino acids wherein an amino acid is exchanged with a different amino acid of similar size, structure, charge, and/or polarity. Examples include exchange of one of the aliphatic or hydrophobic amino acids Ala, Val, Leu, and Ile for one of the other amino acids in that group of four; exchange between the hydroxyl-containing residues Ser and Thr; exchange between the acidic residues Asp and Glu; exchange between the amide residues Asn and Gln, exchange between the basic residues Lys, Arg, and His; exchange between the aromatic residues Phe, Tyr, and Trp; and exchange between the small-sized amino acids Ala, Ser, Thr, Met, and Gly.

The terms “FGFR extracellular domain” and “FGFR ECD” in the context of this invention refer to the portion of an FGFR that is normally found in the extracellular space. An FGFR ECD may include the amino-terminal residues that precede the D1 domain, the D1 domain, the D1-D2 linker region, the D2 domain, the D2-D3 linker region, the D3 domain (Ig domain III), and the carboxy-terminal residues that follow the D3 domain.

The terms “FGFR3 extracellular domain” and “FGFR3 ECD” as used herein refer to a genus consisting of the following sub-genuses: native FGFR3 ECDs, FGFR3 ECD variants, FGFR3 ECD fragments, native FGFR3 ECD fragments, variants of FGFR3 ECD fragments, FGFR3 ECD acidic region muteins, FGFR3 ECDs engineered to have a decrease in the total number of acidic residues within the D1-D2 linker, FGFR3 ECD D1-D2 linker chimeras, FGFR3 ECD glycosylation mutants, and FGFR3 ECD fusion molecules, as well as non-human FGFR3 ECDs. FGFR3 ECDs can include those annotated as NP 001156685, P22607, NP000133, or NP075254, as described by the National Center of Bioinformatics Information (NCBI). The FGFR3 ECDs as defined herein bind FGF1 and/or FGF18 as tested herein. See Example 5.

As used herein, the terms “native FGFR3 ECD” and “wild-type FGFR3 ECD” are used interchangeably to refer to an FGFR3 ECD consisting of an amino acid sequence selected from SEQ ID NOs: 4,5, and 6. Native FGFR3 ECDs and wild-type FGFR3 ECDs include FGFR3 ECD splice variants or isoforms. As used herein, the terms FGFR3 ECD “splice variants” or “splice isoforms” are used interchangeably to refer to naturally occurring alternative splice forms of FGFR3 ECD, such as FGFR3-Δ8-10 ECD (SEQ ID NO: 6), FGFR3-IIIb (ECD SEQ ID NO: 4) and FGFR3-IIIc ECD (SEQ ID NO: 5), which comprise an Ig domain III chosen from Δ8-10, IIIb, and IIIc, respectively.

As used herein, the term “FGFR3-Δ8-10 ECD” refers to the FGFR3 ECD with an Ig domain III chosen from native FGFR3-Δ8-10 (see SEQ ID NO: 6) and Δ8-10 variants. The term “FGFR3-IIIb ECD” refers to the FGFR3 ECD with an Ig domain III chosen from native IIIb (see SEQ ID NO: 4) and IIIb variants. The term “FGFR3-IIIc ECD” refers to the FGFR3 ECD with an Ig domain III chosen from native IIIc (see SEQ ID NO: 5) and IIIc variants.

As used herein, the term “FGFR3 ECD variants” refers to FGFR3 ECDs containing amino acid additions, deletions, and/or substitutions in comparison to the native FGFR3 ECDs, such as those of SEQ ID NOs: 4, 5, and 6. Amino acid additions and deletions may be made at the amino-terminus, at the carboxy-terminus, and/or within SEQ ID NOs: 4, 5, and 6. An exemplary FGFR3 ECD variant that contains amino acid deletions has the amino acid sequence of SEQ ID No: 30. FGFR3 ECD variants may include amino acid substitutions within the FGFR3 ECD that inhibit N-glycosylation, referred to interchangeably herein as “FGFR3 ECD glycosylation mutants” and “FGFR3 ECD N-glycan mutants.” The FGFR3 ECD variants as defined herein retain the ability to bind FGF1 and/or FGF18 as tested herein.

As used herein, the term “native FGFR3 ECD fragment” refers to an FGFR3 ECD having an amino acid sequence selected from SEQ ID NOs: 4, 5, and 6, but modified in that amino acid residues have been deleted from the amino-terminus and/or from the carboxy-terminus of the polypeptide. A non-limiting exemplary FGFR3 ECD fragment has the amino acid sequence of SEQ ID NO: 30, which corresponds to the amino acid sequence of SEQ ID NO: 5, but with the last three carboxy-terminal amino acid residues, YAG, deleted.

As used herein, the terms “FGFR3 ECD fragment variant” and “variant of FGFR3 ECD fragment” are used interchangeably to refer to FGFR3 ECDs containing, not only amino acid deletions from the amino- and/or carboxy-terminus of SEQ ID NOs: 4, 5, and 6, but also amino acid additions, deletions, and/or substitutions within the retained portion of SEQ ID NOs: 4, 5, and 6.

Collectively, “native FGFR3 ECD fragments” and “FGFR3 ECD fragment variants” form the genus of “FGFR3 ECD fragments.” FGFR3 ECD fragmemts as defined herein retain the ability to bind FGF1 and/or FGF18 as tested herein.

The term “FGFR3 ECD D1 domain” refers to the first Ig domain of a native FGFR3 ECD. The native FGFR3 ECD D1 domain consists of the sequence of amino acids 31-88, inclusive, of SEQ ID NO: 4, SEQ ID NO: 5, or SEQ ID NO: 6. (See also FIG. 1.)

The term “FGFR3 ECD D2 domain” refers to the second Ig domain of a native FGFR3 ECD. The native FGFR3 ECD D2 domain consists of the sequence of amino acids 129-221, inclusive, of SEQ ID NO: 4, SEQ ID NO: 5, or SEQ ID NO: 6. (See also FIG. 1.)

The term “FGFR3 ECD D3 domain” refers to the third Ig domain (Ig domain III) of a native FGFR3 ECD. The native FGFR3 ECD D3 domain consists of the sequence of amino acids 246-317, inclusive, of SEQ ID NO: 5; or consists of the sequence of amino acids 231-327, inclusive, of SEQ ID NO: 4. (See also FIG. 1.)

The terms “FGFR3 ECD D1-D2 linker” and “FGFR3 ECD D1-D2 linker region” are used interchangeably to refer to the linker between the first and second Ig domains (the D1 and D2 domains, respectively) of the FGFR3 ECD. The FGFR3 ECD D1-D2 linker has the sequence DAPSSGDDEDGEDEAEDTGVDTG (SEQ ID NO: 23), which is amino acids 105 to 127, inclusive, of SEQ ID NO: 4, SEQ ID NO: 5, or SEQ ID NO: 6. (See also FIG. 1.)

The terms “FGFR3 ECD D2-D3 linker” and “FGFR3 ECD D2-D3 linker region” are used interchangeably to refer to the linker between the second and third Ig domains (the D2 and D3 domains, respectively) of the FGFR3 ECD. (See also FIG. 1.)

As used herein, an “FGFR3 ECD acidic region mutein” is an FGFR3 ECD variant having a greater or smaller number of acidic residues in the D1-D2 linker region than the native FGFR3 ECD D1-D2 linker region. An exemplary FGFR3 ECD acidic region has the amino acid sequence chosen from SEQ ID NOs: 24-27 and 41-43.

An “FGFR3 ECD D1-D2 linker chimera” refers to an FGFR3 ECD acidic region mutein wherein the D1-D2 linker region has been replaced with the D1-D2 linker region from FGFR1, FGFR2, or FGFR4. In certain exemplary D1-D2 linker chimeras, the D1-D2 linker of the FGFR3 ECD is exchanged for a D1-D2 linker of FGFR1: DALPSSEDDDDDDDSSSEEKETDNTKPNPV (SEQ ID NO: 39). In certain exemplary D1-D2 linker chimeras, the D1-D2 linker of the FGFR3 ECD is exchanged for a D1-D2 linker of FGFR2: DAISSGDDED DTDGAEDFVS ENSNNKR (SEQ ID NO: 40). In certain exemplary D1-D2 linker chimeras, the D1-D2 linker of the FGFR3 ECD is exchanged for a D1-D2 linker of FGFR4: DSLTSSNDDED PKSHRDPSNR HSYPQQ (SEQ ID NO: 38). The FGFR3 ECD linker chimeras as defined herein retain the ability to bind FGF1 and/or FGF18 as tested herein.

FGFR3 ECD variants may include amino acid substitutions within the FGFR3 ECD sequence that inhibit N-glycosylation, referred to interchangeably herein as “FGFR3 ECD glycosylation mutants” and “FGFR3 ECD N-glycan mutants.” In certain embodiments, one or more amino acids are mutated to prevent glycosylation at that site in the polypeptide. Non-limiting exemplary FGFR3 ECD amino acids that may be glycosylated include N76, N203, N240, N272, N293, and N306 in SEQ ID NO: 5. Accordingly, one or more of those amino acids may be substituted. Non-limiting exemplary amino acid mutations in FGFR3 ECD glycosylation mutants include N76A, N203A, N240A, N272A, N293A, and N306A in SEQ ID NO: 5. The FGFR3 ECD glycosylation mutants as defined herein retain the ability to bind FGF1 and/or FGF18 as tested herein.

The terms “FGFR3 ECD fusion molecule” and “FGFR3 ECD fusion” are used interchangeably herein to refer to an FGFR3 ECD comprising an FGFR3 ECD polypeptide and a fusion partner. FGFR3 ECD fusions may be constructed based upon any of the FGFR3 ECD genera defined above or any of the FGFR3 ECD species described elsewhere herein. The fusion partner may be linked to either the amino-terminus or the carboxy-terminus of the polypeptide. In certain embodiments, the polypeptide and the fusion partner are covalently linked. If the fusion partner is also a polypeptide (“the fusion partner polypeptide”), the polypeptide and the fusion partner polypeptide may be part of a continuous amino acid sequence. In such cases, the polypeptide and the fusion partner polypeptide may be translated as a single polypeptide from a coding sequence that encodes both the polypeptide and the fusion partner polypeptide. In certain embodiments, the polypeptide and the fusion partner are covalently linked through other means, such as, for example, a chemical linkage other than a peptide bond. Many methods of covalently linking polypeptides to other molecules (for example, fusion partners) are known in the art. The FGFR3 ECD fusion molecules as defined herein retain the ability to bind FGF1 and/or FGF18 as tested herein.

In certain embodiments, the polypeptide and the fusion partner are noncovalently linked. In certain such embodiments, they may be linked, for example, using binding pairs. Exemplary binding pairs include, but are not limited to, biotin and avidin or streptavidin, an antibody and its antigen, etc.

Certain exemplary fusion partners include, but are not limited to, an immunoglobulin Fc domain, albumin, and polyethylene glycol. The amino acid sequences of certain exemplary Fc domains are shown in SEQ ID NOs: 16-18. Exemplary FGFR3 ECD Fc fusions include those shown in Table 4 below.

In certain embodiments, the FGFR3 amino acid sequence is derived from that of a non-human mammal. Such FGFR3 are termed “non-human FGFR3.” In such embodiments, the FGFR3 sequence may be derived from mammals including, but not limited to, rodents, simians, felines, canines, equines, bovines, porcines, ovines, caprines, mammalian laboratory animals, mammalian farm animals, mammalian sport animals, and mammalian pets. Known non-human FGFR3 sequences include those with GenBank Accession Nos. NP001156687, NP001156688, NP001156689, NP445881, NP990840, and NP776743. Such FGFR3 sequences can be modified in the same way as the human FGFR3 sequences described above. In other words, non-human FGFR3 includes the corresponding native FGFR3, FGFR3 variants, FGFR3 fragments, native FGFR3 fragments, variants of FGFR3 fragments, and FGFR3 fusion molecules.

In certain embodiments, the FGFR3 ECD amino acid sequence is derived from that of a non-human mammal. Such FGFR3 ECDs are termed “non-human FGFR3 ECDs.” In such embodiments, the FGFR3 ECD sequence may be derived from mammals including, but not limited to, rodents, simians, felines, canines, equines, bovines, porcines, ovines, caprines, mammalian laboratory animals, mammalian farm animals, mammalian sport animals, and mammalian pets. Known non-human FGFR3 ECD sequences include those annaoted as such in sequences identified by GenBank Accession Nos. NP001156687, NP001156688, NP001156689, NP445881, NP990840, and NP776743. Such FGFR3 ECD sequences can be modified in the same way as the human FGFR3 ECD sequences described above. In other words, non-human FGFR3 ECDs include the corresponding native FGFR3 ECDs, FGFR3 ECD variants, FGFR3 ECD fragments, native FGFR3 ECD fragments, variants of FGFR3 ECD fragments, FGFR3-Δ8-10 ECDs, FGFR3-IIIb ECDs, and FGFR3-IIIc ECDs, FGFR3 ECD acidic region muteins, FGFR3 ECD D1-D2 linker chimeras, FGFR3 ECD glycosylation mutants, and FGFR3 ECD fusion molecules. The non-human FGFR3 ECD as defined herein are able to bind the corresponding non-human FGF1 and/or FGF18 as tested herein.

The terms “signal peptide” and “signal sequence” are used interchangeably herein to refer to a sequence of amino acid residues that facilitates secretion of a polypeptide from a mammalian cell. A signal peptide is typically cleaved upon export of the polypeptide from the mammalian cell. Certain exemplary signal peptides include, but are not limited to, the native signal peptides of FGFR1, FGFR2, FGFR3, and FGFR4, such as, for example, the amino acid sequences of SEQ ID NOs: 19-22. Certain exemplary signal peptides also include signal peptides from heterologous proteins. Other exemplary signal peptides also include signal peptides from non-human proteins such as non-human FGFR1, FGFR2, FGFR3, and FGFR4. A “signal sequence” refers to a polynucleotide sequence that encodes a signal peptide.

A “vector” refers to a polynucleotide that is used to express a polypeptide of interest in a host cell. A vector may include one or more of the following elements: an origin of replication, one or more regulatory sequences (such as, for example, promoters and/or enhancers) that regulate the expression of the polypeptide of interest, and/or one or more selectable marker genes (such as, for example, antibiotic resistance genes and genes that can be used in colorimetric assays, e.g., β-galactosidase).

A “host cell” refers to a cell that can be or has been a recipient of a vector or isolated polynucleotide. Host cells may be prokaryotic cells or eukaryotic cells. Exemplary eukaryotic cells include mammalian cells, such as primate or non-primate animal cells; fungal cells; plant cells; and insect cells. Certain exemplary mammalian cells include, but are not limited to, 293 and CHO cells.

The term “isolated” as used herein refers to a molecule that has been separated from at least some of the components with which it is typically found in nature. For example, a polypeptide is referred to as “isolated” when it is separated from at least some of the components of the cell in which it was produced. Where a polypeptide is secreted by a cell after expression, physically separating the supernatant containing the polypeptide from the cell that produced it is considered to be “isolating” the polypeptide. Similarly, a polynucleotide is referred to as “isolated” when it is not part of the larger polynucleotide (such as, for example, genomic DNA or mitochondrial DNA, in the case of a DNA polynucleotide) in which it is typically found in nature, or is separated from at least some of the components of the cell in which it was produced, e.g., in the case of an RNA polynucleotide. Thus, a DNA polynucleotide that is contained in a vector inside a host cell may be referred to as “isolated” so long as that polynucleotide is not found in that vector in nature.

The term “subject” is used herein to refer to mammals, including, but not limited to, rodents, simians, humans, felines, canines, equines, bovines, porcines, ovines, caprines, mammalian laboratory animals, mammalian farm animals, mammalian sport animals, and mammalian pets.

“Treatment,” as used herein, covers any administration or application of a therapeutic for disease in a mammal, including a human, and includes inhibiting the disease or progression of the disease, partially inhibiting or slowing the disease or its progression, arresting its development, partially or fully relieving the disease, or curing the disease, for example, by causing regression, or restoring or repairing a lost, missing, or defective function; or stimulating an inefficient process.

“Administration,” as used herein, includes both self-administration by the subject as well as administration by another individual, such as a physician, nurse, or veterinarian.

A “pharmaceutically acceptable carrier” refers to a non-toxic solid, semisolid, or liquid filler, diluent, encapsulating material, formulation auxiliary, or carrier conventional in the art for use with a therapeutic agent for administration to a subject. A pharmaceutically acceptable carrier is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation. The pharmaceutically acceptable carrier is appropriate for the formulation employed. For example, if the composition is to be administered orally, the carrier may be a gel capsule. If the composition is to be administered subcutaneously, the carrier ideally is not irritable to the skin and does not cause injection site reaction.

FGFR3 ECDs

As defined above, an FGFR3 ECD is a genus consisting of the following sub-genuses: native FGFR3 ECDs, FGFR3 ECD variants, FGFR3 ECD fragments, native FGFR3 ECD fragments, variants of FGFR3 ECD fragments, FGFR3 ECD acidic region muteins, FGFR3 ECDs engineered to have a decrease in the total number of acidic residues within the D1-D2 linker, FGFR3 ECD D1-D2 linker chimeras, FGFR3 ECD glycosylation mutants, and FGFR3 ECD fusion molecules, as well as non-human FGFR3 ECDs. FGFR3 ECDs can include those annotated as NP001156685, P22607, NP000133, or NP075254, as described by the National Center of Bioinformatics Information (NCBI). The FGFR3 ECDs as defined herein bind FGF1 and/or FGF18 as tested herein. (See Example 5).

Description of the method used herein to test FGF1 and FGF18 binding by FGFR3 ECD is provided in Example 5. A Biacore® T100 surface plasmon resonance (SPR) technology-based assay was used to measure binding of FGF ligands to FGFR3 ECD. In certain embodiments, an FGFR3 ECD binds to FGF1 with an equilibrium dissociation constant (KD) value no more than 100 nM, or with a KD value no more than 10 nM, or with a KD value no more than 1 nM, or with a KD value no more than 0.1 nM, in a Biacore® T100 ligand binding assay. (See Example 5.) In certain embodiments, an FGFR3 ECD binds to FGF18 with a KD value no more than 100 nM, or with a KD value no more than 10 nM, or with a KD value no more than 1 nM, with a KD value no more than 0.1 nM, or with a KD value no more than 0.01 nM, in a Biacore® T100 ligand binding assay. (Id.)

Signal Peptides

Typically, the signal peptide is cleaved from the mature FGFR3 ECD polypeptide. Thus, in many embodiments, the FGFR3 ECD lacks a signal peptide. Nonetheless, in certain embodiments, an FGFR3 ECD includes at least one signal peptide, which may be selected from a native FGFR3 signal peptide and/or a heterologous signal peptide. In some embodiments, the FGFR3 ECD comprises a signal sequence at its amino terminus. Any one of the above genuses of polypeptides defined above or the polypeptide species described herein may further include a signal peptide. Exemplary signal peptides include, but are not limited to, the signal peptides of FGFR1, FGFR2, FGFR3, and FGFR4, such as, for example, the amino acid sequences of SEQ ID NOs: 19 to 22. In other embodiments, the signal peptide may be a signal peptide from a heterologous protein.

FGFR3 ECD Fusion Molecules and Their Construction

In some embodiments, the FGFR3 ECD is a fusion molecule. Accordingly, any one of the genuses of polypeptides defined above or the polypeptide species described herein may further include a fusion partner. FGFR3 ECD fusion molecules comprising an FGFR3 ECD polypeptide and a fusion partner may be used in the methods herein.

Certain exemplary FGFR3 ECD fusion molecules are provided in Table 4. For example, an exemplary FGFR3-IIIb ECD Fc fusion (SEQ ID NO: 7) is a native FGFR3-IIIb ECD fused to an Fc. An exemplary FGFR3-IIIb ECD Fc fusion with a GS linker has the amino acid sequence of SEQ ID NO: 8. An exemplary FGFR3-IIIc ECD Fc fusion (SEQ ID NO: 9) is a native FGFR3-IIIc ECD fused to an Fc. An exemplary FGFR3-IIIc ECD Fc fusion with a GS linker has the amino acid sequence of SEQ ID NO: 10. FGFR3-IIIc ECD Δ3Fc fusion with a GS linker (SEQ ID NO: 33) is a native FGFR3-IIIc ECD fragment with a 3 amino acid C-terminal deletion fused to an Fc through the linker glycine-serine (“GS”). Certain exemplary FGFR3 ECD fusion molecules also are shown in SEQ ID NO: 11 (a native FGFR3-IIIc ECD fragment with a 4 amino acid C-terminal deletion fused to an Fc), SEQ ID NO: 12 (a native FGFR3-IIIc ECD fragment with a 8 amino acid C-terminal deletion fused to an Fc), SEQ ID NO: 13 (a native FGFR3-IIIc ECD fragment with a 9 amino acid C-terminal deletion fused to an Fc), SEQ ID NO: 14 (a native FGFR3-IIIc ECD fragment with a 13 amino acid C-terminal deletion fused to an Fc), SEQ ID NO: 15 (a native FGFR3-IIIc ECD fragment with a 20 amino acid C-terminal deletion fused to an Fc), SEQ ID NO: 28, SEQ ID NO: 35, and SEQ ID NO: 37.

Fusion Partners and Conjugates

In certain embodiments, a fusion partner is selected that imparts favorable pharmacokinetics and/or pharmacodynamics on the FGFR3 ECD fusion molecule.

Non-limiting exemplary fusion partners include polymers, polypeptides, lipophilic moieties, and succinyl groups. Exemplary polypeptide fusion partners include serum albumin and an antibody Fc domain. Exemplary polymer fusion partners include, but are not limited to, polyethylene glycol (PEG), including polyethylene glycols having branched and/or linear chains. Some embodiments may include more than one fusion partner, such as an Fc and a polymer fusion partner such as PEG.

Oligomerization Domain Fusion Partners

In various embodiments, oligomerization offers certain functional advantages to a fusion protein, including, but not limited to, multivalency, increased binding strength, and the combined function of different domains. Accordingly, in certain embodiments, a fusion partner comprises an oligomerization domain, for example, a dimerization domain. Exemplary oligomerization domains include, but are not limited to, coiled-coil domains, including alpha-helical coiled-coil domains; collagen domains; collagen-like domains, and certain immunoglobulin domains. Certain exemplary coiled-coil polypeptide fusion partners include the tetranectin coiled-coil domain; the coiled-coil domain of cartilage oligomeric matrix protein; angiopoietin coiled-coil domains; and leucine zipper domains. Certain exemplary collagen or collagen-like oligomerization domains include, but are not limited to, those found in collagens, mannose binding lectin, lung surfactant proteins A and D, adiponectin, ficolin, conglutinin, macrophage scavenger receptor, and emilin.

Antibody Fc Immunoglobulin Domain Fusion Partners

Many Fc domains that could be used as fusion partners are known in the art. In certain embodiments, a fusion partner is an Fc immunoglobulin domain. An Fc fusion partner may be a wild-type Fc found in a naturally occurring antibody, a variant thereof, or a fragment thereof. Non-limiting exemplary Fc fusion partners include Fcs comprising a hinge and the CH2 and CH3 constant domains of a human IgG, for example, human IgG1, IgG2, IgG3, or IgG4. Certain additional Fc fusion partners include, but are not limited to, those from human IgA and IgM. In certain embodiments, an Fc fusion partner is that from a human IgG1 In certain embodiments, an Fc fusion partner is from a human IgG1 and comprises a C237S mutation. In certain embodiments, an Fc fusion partner comprises a hinge, CH2, and CH3 domains of human IgG2 with a P331S mutation, as described in U.S. Pat. No. 6,900,292. Certain exemplary Fc domain fusion partners are shown in SEQ ID NOs: 16-18.

Certain exemplary FGFR3 ECD fusion molecules comprise, but are not limited to, polypeptides having the amino acid sequences of SEQ ID NOs: 7-15, 28, 35 and 37.

Albumin Fusion Partners and Albumin-binding Molecule Fusion Partners

In certain embodiments, a fusion partner is an albumin. Certain exemplary albumins include, but are not limited to, human serum album (HSA) and fragments of HSA that are capable of increasing the serum half-life and/or bioavailability of the polypeptide to which they are fused. In certain embodiments, a fusion partner is an albumin-binding molecule, such as, for example, a peptide that binds albumin or a molecule that conjugates with a lipid or other molecule that binds albumin. In certain embodiments, a fusion molecule comprising HSA is prepared as described, e.g., in U.S. Pat. No. 6,686,179.

Polymer Fusion Partners

In certain embodiments, a fusion partner is a polymer, for example, polyethylene glycol (PEG). PEG may comprise branched and/or linear chains. In certain embodiments, a fusion partner comprises a chemically-derivatized polypeptide having at least one PEG moiety attached. Pegylation of a polypeptide may be carried out by any method known in the art. Certain exemplary PEG attachment methods include, for example, EP 0 401 384; Malik et al., Exp. Hematol., 20:1028-1035 (1992); Francis, Focus on Growth Factors, 3:4-10 (1992); EP 0 154 316; EP 0 401 384; WO 92/16221; and WO 95/34326. As non-limiting examples, pegylation may be performed via an acylation reaction or an alkylation reaction, resulting in attachment of one or more PEG moieties via acyl or alkyl groups. In certain embodiments, PEG moieties are attached to a polypeptide through the α- or ε-amino group of one or more amino acids, although any other points of attachment known in the art are also contemplated.

Pegylation by acylation typically involves reacting an activated ester derivative of a PEG moiety with a polypeptide. A non-limiting exemplary activated PEG ester is PEG esterified to N-hydroxysuccinimide (NHS). As used herein, acylation is contemplated to include, without limitation, the following types of linkages between a polypeptide and PEG: amide, carbamate, and urethane. See, e.g., Chamow, Bioconjugate Chem., 5:133-140 (1994). Pegylation by alkylation typically involves reacting a terminal aldehyde derivative of a PEG moiety with a polypeptide in the presence of a reducing agent. Non-limiting exemplary reactive PEG aldehydes include PEG propionaldehyde, which is water stable, and mono C1-C10 alkoxy or aryloxy derivatives thereof. See, e.g., U.S. Pat. No. 5,252,714.

In certain embodiments, a pegylation reaction results in poly-pegylated polypeptides. In certain embodiments, a pegylation reaction results in mono-, di-, and/or tri-pegylated polypeptides. Further, desired pegylated species may be separated from a mixture containing other pegylated species and/or unreacted starting materials using various purification techniques known in the art, including among others, dialysis, salting-out, ultrafiltration, ion-exchange chromatography, gel filtration chromatography, and electrophoresis.

Exemplary Attachment of Fusion Partners

The fusion partner may be attached, either covalently or non-covalently, to the amino-terminus or the carboxy-terminus of an FGFR3 ECD. The attachment may also occur at a location within the FGFR3 ECD other than the amino-terminus or the carboxy-terminus, for example, through an amino acid side chain (such as, for example, the side chain of cysteine, lysine, histidine, serine, or threonine).

In either covalent or non-covalent attachment embodiments, a linker may be included between the fusion partner and the FGFR3 ECD. Such linkers may be comprised of amino acids and/or chemical moieties.

Exemplary methods of covalently attaching a fusion partner to an FGFR3 ECD include, but are not limited to, translation of the fusion partner and the FGFR3 ECD as a single amino acid sequence and chemical attachment of the fusion partner to the FGFR3 ECD. When the fusion partner and the FGFR3 ECD are translated as single amino acid sequence, additional amino acids may be included between the fusion partner and the FGFR3 ECD as a linker. In certain embodiments, the linker is glycine-serine (“GS”). In certain embodiments, the linker is selected based on the polynucleotide sequence that encodes it, to facilitate cloning the fusion partner and/or FGFR3 ECD into a single expression construct (for example, a polynucleotide containing a particular restriction site may be placed between the polynucleotide encoding the fusion partner and the polynucleotide encoding the FGFR3 ECD, wherein the polynucleotide containing the restriction site encodes a short amino acid linker sequence).

When the fusion partner and the FGFR3 ECD are covalently coupled by chemical means, linkers of various sizes can typically be included during the coupling reaction.

Exemplary methods of non-covalently attaching a fusion partner to an FGFR3 ECD include, but are not limited to, attachment through a binding pair. Exemplary binding pairs include, but are not limited to, biotin and avidin or streptavidin, an antibody and its antigen, etc. The selected non-covalent attachment method should be suitable for the conditions under which the FGFR3 ECD fusion molecule will be used, taking into account, for example, the pH, salt concentrations, and temperature.

Non-Human FGFR3 ECDs

As described above, in certain embodiments, the FGFR3 ECD amino acid sequence is that of a non-human mammal. In such embodiments, the FGFR3 ECD sequence may be derived from mammals including, but not limited to, rodents, simians, felines, canines, equines, bovines, porcines, ovines, caprines, mammalian laboratory animals, mammalian farm animals, mammalian sport animals, and mammalian pets. Known non-human FGFR3 ECD sequences include those with GenBank Accession Nos. NP001156687, NP001156688, NP001156689, NP445881, NP990840, and NP776743. As set forth above, such FGFR3 ECD sequences can be modified in the same way as the human FGFR3 ECD sequences described above. In other words, non-human FGFR3 ECDs include the corresponding native FGFR3 ECDs, FGFR3 ECD variants, FGFR3 ECD fragments, native FGFR3 ECD fragments, variants of FGFR3 ECD fragments, FGFR3 ECD acidic region muteins, FGFR3 ECD D1-D2 linker chimeras, FGFR3 ECD glycosylation mutants, and FGFR3 ECD fusion molecules.

Nucleic Acid Molecules, Vectors, and Protein Expression Methods

Nucleic acid molecules that encode FGFR3 ECDs can be constructed by one skilled in the art using recombinant DNA techniques conventional in the art.

In certain embodiments, a polynucleotide encoding a polypeptide of the invention comprises a nucleotide sequence that encodes a signal peptide, which, when translated, is fused to the amino-terminus of the FGFR3 polypeptide. As discussed above, the signal peptide may be the native signal peptide, the signal peptide of FGFR1, FGFR2, FGFR3, or FGFR4, or may be another heterologous signal peptide. The amino acid sequences for certain exemplary FGFR signal peptides are shown, e.g., in SEQ ID NOs: 16 to 18. Certain exemplary signal peptides are known in the art, and are described, e.g., in the online Signal Peptide Database maintained by the Department of Biochemistry, National University of Singapore, http://proline.bic.nus.edu.sg/spdb/index.html (see also Choo et al., BMC Bioinformatics, 6: 249 (2005)); and in PCT Publication No. WO 2006/081430.

To prepare the polypeptides, the nucleic acid molecule comprising the polynucleotide encoding the FGFR3 ECD may be placed into a vector suitable for expression in a selected host cell. Such vectors include, but are not limited to, DNA vectors, phage vectors, viral vectors, retroviral vectors, etc.

In certain embodiments, a vector is selected that is optimized for expression of polypeptides in CHO-S or CHO-S-derived cells. Exemplary such vectors are described, e.g., in Running Deer et al., Biotechnol. Prog. 20:880-889 (2004).

In certain embodiments, a vector is chosen for in vivo expression of the polypeptides of the invention in animals, including humans. In certain such embodiments, expression of the polypeptide is under the control of a promoter that functions in a tissue-specific manner. For example, liver-specific promoters are described, e.g., in PCT Publication No. WO 2006/076288.

The polypeptides of the invention can be expressed, in various embodiments, in prokaryotic cells, such as bacterial cells; or eukaryotic cells, such as fungal cells, plant cells, insect cells, and mammalian cells. Such expression may be carried out, for example, according to procedures known in the art. Certain exemplary eukaryotic cells that can be used to express polypeptides include, but are not limited to, Cos cells, including Cos 7 cells; 293 cells, including 293-6E and 293-T cells; CHO cells, including CHO-S and DG44 cells; and NS0 cells. In certain embodiments, a particular eukaryotic host cell is selected based on its ability to make certain desired post-translational modifications of the polypeptide of the invention. For example, in certain embodiments, CHO cells produce FGFR3 ECDs that have a higher level of glycosylation and/or sialylation than the same polypeptide produced in 293 cells.

Introduction of a nucleic acid into a desired host cell can be accomplished by any method known in the art, including, but not limited to, calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, etc. Certain exemplary methods are described, e.g., in Sambrook et al., Molecular Cloning, A Laboratory Manual, 3rd ed. Cold Spring Harbor Laboratory Press (2001). Nucleic acids may be transiently or stably transfected in the desired host cells, according to methods known in the art.

In certain embodiments, a polypeptide can be produced in vivo in an animal that has been engineered or transfected with a nucleic acid molecule encoding the polypeptide, according to methods known in the art.

Purification of FGFR3 ECD Polypeptides

The polypeptides of the invention can be purified by various methods known in the art. Such methods include, but are not limited to, the use of affinity matrices, ion exchange chromatography, and/or hydrophobic interaction chromatography. Suitable affinity ligands include any ligands of the FGFR3 ECD, antibodies to FGFR3 ECD, or, in the case of an FGFR3 ECD fusion, a ligand of the fusion partner. For example, a Protein A, Protein G, Protein A/G, or an antibody affinity column may be used to bind to an Fc fusion partner to purify a polypeptide of the invention. Hydrophobic interactive chromatography, for example, a butyl or phenyl column, may also suitable for purifying certain polypeptides.

Methods of Administration

Routes of Administration and Carriers

The polypeptides of the invention can be administered in vivo by various routes known in the art, including, but not limited to, intravenous, subcutaneous, parenteral, intranasal, intramuscular, buccal, intraperitoneal, intradermal, topical, transdermal, and intrathecal, or otherwise by implantation or inhalation. The subject compositions can be formulated into preparations in solid, semi-solid, liquid, or gaseous forms; including, but not limited to, tablets, capsules, powders, granules, ointments, solutions, injections, inhalants, and aerosols. Nucleic acid molecules encoding the polypeptides of the invention can be coated onto gold microparticles and delivered intradermally by a particle bombardment device, or “gene gun,” as described in the literature (see, e.g., Tang et al., Nature 356:152-154 (1992)).

In some embodiments, compositions comprising the polypeptides of the invention are provided in formulation with pharmaceutically acceptable carriers, a wide variety of which are known in the art (see, e.g., Gennaro, Remington: The Science and Practice of Pharmacy with Facts and Comparisons: Drugfacts Plus, 20th ed. (2003); Ansel et al., Pharmaceutical Dosage Forms and Drug Delivery Systems, 7th ed., Lippencott Williams and Wilkins (2004); Kibbe et al., Handbook of Pharmaceutical Excipients, 3rd ed., Pharmaceutical Press (2000)). Various pharmaceutically acceptable carriers, which include vehicles, adjuvants, carriers, and diluents, are available to the public. Moreover, various pharmaceutically acceptable auxiliary substances, such as pH adjusting and buffering agents, tonicity adjusting agents, stabilizers, wetting agents and the like, are also available to the public. Certain non-limiting exemplary carriers include saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof.

In various embodiments, compositions comprising polypeptides of the invention can be formulated for injection by dissolving, suspending, or emulsifying them in an aqueous or nonaqueous solvent, such as vegetable or other oils, synthetic aliphatic acid glycerides, esters of higher aliphatic acids, or propylene glycol; and if desired, with conventional additives such as solubilizers, isotonic agents, suspending agents, emulsifying agents, stabilizers and preservatives. The compositions may also be formulated, in various embodiments, into sustained release microcapsules, such as with biodegradable or non-biodegradable polymers. A non-limiting exemplary biodegradable formulation includes poly lactic acid-glycolic acid polymer. A non-limiting exemplary non-biodegradable formulation includes a polyglycerin fatty acid ester. Certain methods of making such formulations are described, for example, in EP 1 125 584 A1.

Pharmaceutical packs and kits comprising one or more containers, each containing one or more doses of the polypeptides of the invention are also provided. In certain embodiments, a unit dosage is provided wherein the unit dosage contains a predetermined amount of a composition comprising a polypeptide of the invention, with or without one or more additional agents. In certain embodiments, such a unit dosage is supplied in single-use prefilled syringe for injection. In various embodiments, the composition contained in the unit dosage may comprise saline, sucrose, or the like; a buffer, such as phosphate, or the like; and/or be formulated within a stable and effective pH range. Alternatively, in certain embodiments, the composition may be provided as a lyophilized powder that can be reconstituted upon addition of an appropriate liquid, for example, sterile water. In certain embodiments, the composition comprises one or more substances that inhibit protein aggregation, including, but not limited to, sucrose and arginine. In certain embodiments, a composition of the invention comprises heparin and/or a proteoglycan.

The FGFR3 ECD compositions are administered in an amount effective to promote hair growth. The effective amount is typically dependent on the weight of the subject being treated, his or her physical or health condition, the extensiveness of the condition to be treated, and/or the age of the subject being treated. In general, the polypeptides of the invention can be administered subcutaneously in an amount in the range of about 10 ng to about 500 μg. Optionally, the polypeptides of the invention can be administered subcutaneously in an amount in the range of about 10 ng to about 100 μg. Further optionally, the polypeptides of the invention can be administered subcutaneously in an amount in the range of about 100 ng to about 10 μg. In general, the polypeptides of the invention can be administered intravenously in an amount in the range of about 10 μg/kg body weight to about 30 mg/kg body weight per dose. Optionally, the polypeptides of the invention can be administered intravenously in an amount in the range of about 100 μg/kg body weight to about 20 mg/kg body weight per dose. Further optionally, the polypeptides of the invention can be administered intravenously in an amount in the range of about 0.5 mg/kg body weight to about 20 mg/kg body weight per dose.

The compositions comprising the polypeptides of the invention can be administered as needed to subjects. Determination of the frequency of administration can be made by persons skilled in the art, such as an attending physician or pharmacist or hair growth specialist based on considerations of the condition being treated, age of the subject being treated, severity of the condition being treated, general state of health of the subject being treated and the like. In certain embodiments, an effective dose of the polypeptide of the invention is administered to a subject one or more times. In various embodiments, an effective dose of the polypeptide of the invention is administered to the subject no more than once a year, nor more than twice a year, no more than twice a month, no more than once a week, no more than twice a week, or no more than three times a week. In various embodiments, an effective dose of the polypeptide of the invention is administered to the subject for no more than a week, for no more than a month, for no more than three months, for no more than six months, or for no more than a year.

Combination Therapy

Polypeptides of the invention may be administered alone or with other modes of treatment. They may be provided before, substantially contemporaneous with, or after other modes of treatment. Certain exemplary combination therapies could include a combination of an FGFR3 ECD with minoxidil, finasteride, dutasteride, other 5-alpha reductase inhibitors, and/or hair transplant surgery.

EXAMPLES

The examples discussed below are intended to be purely exemplary of the invention and should not be considered to limit the invention in any way. The examples are not 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 (for example, 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 Centigrade, and pressure is at or near atmospheric.

Example 1 Construction of an FGFR3 ECD-Fc Fusion Molecule

An FGFR3 ECD-Fc having a 3 amino acid carboxy-terminal deletion from the FGFR3-IIIc ECD (“FGFR3 ECD Δ3-Fc” or “FGFR3 ECD-Fc”) fused to Fc with a GS linker (SEQ ID NO: 33) was subcloned into the pTT5 and pDEF38 vectors using PCR and conventional subcloning techniques.

The primary sequence and domain structure of the FGFR3 ECD moiety in the FGFR3 ECD Δ3-Fc construct is shown in FIG. 1. FIG. 2 shows an amino acid sequence alignment of a portion of the C-terminal region of FGFR ECDs, denoting Ig domain III.

For transient expression in 293-6E cells, the vector pTT5 (Biotechnology Research Institute, Montreal, Canada) was used. For expression of the fusion proteins in CHO-S host cells, we used the pTT5 and pDEF38 (ICOS Corporation, Bothell, Wash.) vectors. DG44 (Invitrogen, Carlsbad, Calif.) is a derivative cell line of the CHO-S cell line that we have found can give higher yields of recombinant proteins. For expression of the fusion proteins in DG44 host cells, we used the vector pDEF38.

Example 2 Purification of Expressed Protein

An FGFR3 ECD-Fc fusion protein expressed from recombinant host cells was purified from the cell culture supernatant using a first purification step of Protein-A affinity chromatography, followed by a second purification step of butyl hydrophobic interaction chromatography. For the Protein-A affinity chromatography step, the components of the media were separated on a Mabselect Protein-A Sepharose column (GE Healthcare Bio-Sciences, Piscataway, N.J.), which will bind to the Fc region of the fusion molecule. The column was equilibrated with ten column volumes of a sterile buffer of 10 mM Tris, 100 mM NaCl, pH 8.0; then the cell culture supernatant was applied to the column. The column was washed with eight column volumes of sterile 10 mM Tris, 100 mM NaCl buffer, pH 8.0. The bound material, including the fusion protein, was then eluted at a rate of 10 ml/min with a one step elution using seven column volumes of elution buffer (100 mM glycine, 100 mM NaCl, pH 2.7). Ten ml fractions were collected in tubes containing one ml 1 M Tris pH 8.0 (Ambion, Austin, Tex.) to neutralize the eluate. Fractions comprising the fusion protein were identified by gel electrophoresis and pooled.

For the second purification step of butyl hydrophobic interaction chromatography, pooled Protein-A column eluates were further purification on a butyl Sepharose column using a GE Healthcare Akta Purifier 100 (GE Healthcare Bio-Sciences, Piscataway, N.J.). The column was first equilibrated with five column volumes of sterile 10 mM Tris, 1 M ammonium sulfate, pH 8.0. A half volume of 3 M ammonium sulfate was then added to the eluate, which was then applied to the equilibrated butyl Sepharose column. The column was washed with four column volumes of the equilibration buffer and the bound material was eluted at a rate of five ml/min with a linear gradient starting at 50% equilibration buffer/50% elution buffer (10 mM Tris pH 8.0) and ending at 90% elution buffer/10% equilibration buffer over a total volume of 20 column volumes. Finally, an additional two column volumes of 100% elution buffer was used. Fourteen ml fractions were collected. The fusion protein was eluted with approximately 40-60% elution buffer. The fractions containing the bulk of the fusion protein were identified by gel electrophoresis and pooled.

After purification, endotoxin levels were checked by the limulus amoebocyte lysate (LAL) assay (Cambrex, Walkersville, Md.). Endotoxin levels were confirmed to be less than or equal to 1 endotoxin unit (EU) per mg of the fusion protein.

Example 3 Transient Expression of Fusion Protein in CHO-S Host Cells

An FGFR3 ECD-Fc fusion protein was transiently expressed in CHO-S cells. Briefly, a 500 ml culture of CHO-S cells (Invitrogen) was established by inoculating 0.5×106 cells/ml in fresh 37° C. Freestyle CHO medium containing 8 mM L-Glutamine (Invitrogen). The cells were grown in a 2 l plastic flask and were derived from a seed strain that was continuously maintained up to passage 20. The following day, the cells were counted and diluted, if necessary, to 1×106 cells/ml in 37° C. Freestyle CHO medium (Invitrogen) with a cell viability greater than 95%. The cells were transfected by transferring 10 ml of 37° C. OptiPRO SFM medium containing 8 mM L-Glutamine (dilution media) into two 50 ml tubes. To the first tube (A), 625 ul of FreestyleMax transfection reagent (Invitrogen) were added. To the second tube (B), 625 ug of DNA were added. Both tubes were gently mixed by inverting, and the contents of tube A were immediately added to tube B, followed by gentle mixing by inversion. The mixture was incubated at room temperature for between 10 to 20 min, and was then delivered drop-wise into the 500 ml cell culture in the 21 culture flask while slowly swirling the flask. The culture was then transferred to an incubator at 37° C., 5% CO2, 125 rpm. After six days, the cell viability was greater than 80%, and the culture supernatant was collected into a centrifuge bottle. The supernatant was centrifuged at 1,000×g for 10 min, transferred to a new centrifuge bottle, and centrifuged at 4,000×g for 10 min. The supernatant was collected into a new bottle and filtered through a 0.2 um filter. The supernatant was stored at 37° C. prior to the purification step. The fusion protein was purified from the culture supernatant as described in Example 2, except that Q Sepharose anion exchange chromatography was used as the second purification step. Protein-A eluates were applied to a Q Sepharose HP column (GE Healthcare 17-1014-01) equilibrated with five column volumes of sterile buffer (10 mM Tris, 50 mM NaCl, pH 8.0). The column was washed with five column volumes of the same buffer and the bound material was eluted at a rate of five ml/min with a linear gradient of 15 column volumes of elution buffer (10 mM Tris, 2 M NaCl, pH 8.0), followed by five column volumes with 100% elution buffer. Fourteen ml fractions were collected and the fractions comprising the FGFR3 ECD-Fc were identified by gel electrophoresis and pooled. The FGFR3 ECD-Fc fusion proteins eluted with approximately 10-25% elution buffer. Protein levels were determined based on absorbance measurements at 280 nm.

Example 4 Stable Production in DG44 Cells

The expression vector FGFR3-Fc/pDEF38, described in Example 1, was used to transfect DG44 host cells for stable production of FGFR ECD fusion proteins. The untransfected DHFR-negative CHO cell line, DG44, was cultured in CHO-CD serum free medium (Irvine Scientific, Irvine, Calif.) supplemented with 8 mM L-Glutamine, 1× Hypoxanthine/Thymidine (HT; Invitrogen), and 18 ml/L of Pluronic-68 (Invitrogen). About 50 ug of plasmid DNA of each of FGFR3 ECD-Fc/pDEF38, FGFR2 ECD-Fc/pDEF38, and ABmut1/pDEF38 was linearized by digestion with restriction enzyme PvuI, then precipitated by addition of ethanol, briefly air-dried, and then resuspended in 400 ul of sterile, distilled water. The DG44 cells were seeded into a shaker flask at a density of about 4×105/ml the day before transfection, and reached a density of about 0.8×106/ml on the day of transfection. The cells were harvested by centrifugation and about 1×107 cells were used per transfection.

For transfection, each cell pellet was resuspended in 0.1 ml of Nucleofector V solution and transferred to an Amaxa Nucleofector cuvette (Amaxa, Cologne, Germany). About 5 ug of the resuspended linearized plasmid DNA was added and mixed with the suspended DG44 cells in the cuvette. Cells were then electroporated with an Amaxa Nucleofector Device II using program U-024. Electroporated cells were cultured in CHO-CD medium for two days and then transferred into selective medium (CHO-CD serum free medium supplemented with 8 mM L-Glutamine and 18 ml/L Pluronic-68). The selective medium was changed once every week. After about 12 days, 1 ug/ml R3 Long IGF I growth factor (Sigma, St. Louis, Mo.) was added to the medium and the culture was continued for another week until confluent. The supernatants from pools of stably transfected cell lines were assayed by a sandwich ELISA to determine the product titer. This transfection method generated an expression level of about 30 ug/ml of the expressed fusion protein from the pools of stably transfected cells.

Example 5 Specificity and Affinity of Ligand Binding to FGFR3-IIIc ECD-Fc Measured by Biacore Analysis

The specificity of FGF ligand binding to an FGFR3-IIIc ECD-Fc (SEQ ID NO: 10) was assessed using Biacore® T100 surface plasmon resonance (SPR) technology (Biacore; Piscataway, N.J.). Expression constructs for expressing the FGFR3-IIIc ECD-Fc (SEQ ID NO: 10) fusion protein in 293-6E host cells using the pTT5 vector were made in a manner similar to that described above using cDNAs prepared internally and conventional techniques. The FGFR3-IIIc ECD-Fc fusion protein was produced from 293-6E host cells as described in WO/2007/014123 (PCT/US06/028597) (Examples 2 and 3).

Protein-A was covalently linked to a CM5 chip, according to manufacturer's instructions and then an FGFR ECD fusion protein was bound to the chip by the interaction of the Fc domain with the Protein-A. The FGF ligands were placed in contact with the FGFR ECD fusion protein, also according to manufacturer's instructions, in the presence of HBS-P buffer (Biacore; Piscataway, N.J.) supplemented with 50 ug/ml heparin (Sigma; St. Louis, Mo.).

All the recombinant FGF ligands were from R&D Systems (Minneapolis, Minn.) except for FGF-18 which was from Wako Chemicals (Richmond, Va.). FGF ligands were each tested at six to eight concentrations ranging from 4.5 ng/ml to 10 ug/ml. The FGF ligands were recombinant and of human origin, except for FGF-18, which was of recombinant mouse origin.

The binding of the FGFR3-IIIc ECD-Fc, to various FGF ligands was measured in real time. Table 1 below shows the resulting association constants (ka), dissociation constants (kd) and equilibrium dissociation constants (KD) that were determined from these studies.

As summarized in Table 1, the relative rank of FGF binding affinity to the FGFR3-IIIc-Fc was FGF-18>FGF-1>FGF-9>FGF-2, FGF-4>FGF-20>FGF-5>FGF-7>FGF-19.

TABLE 1 Real-Time Ligand Binding to FGFs FGFR3-IIIc-ECD-Fc Ligand ka (1/(M · s)) kd (1/s) KD (M) FGF-1 2.83 × 106* 3.31 × 10−4* 1.26 × 10−10* FGF-2 3.36 × 105 1.37 × 10−3 1.06 × 10−9 FGF-4 8.08 × 105 1.55 × 10−3 1.91 × 10−9 FGF-5 2.31 × 105 1.69 × 10−3 9.70 × 10−9 FGF-7 3.19 × 105 4.71 × 10−2 1.48 × 10−7 FGF-9 7.76 × 105 4.17 × 10−4 5.37 × 10−10 FGF-18 5.16 × 106* 1.80 × 10−4* 3.50 × 10−11* FGF-19 5.63 × 104 4.43 × 10−1 7.87 × 10−6 FGF-20 1.85 × 105 4.04 × 10−4 2.17 × 10−9 *= average of two independent measurements

Example 6 Systemic Delivery of an FGFR3 ECD Fusion Molecule Promotes Hair Growth in Mice

Eight-week-old female C57B1.6 mice (Charles River Labs, Wilmington, Mass.) were weighed and sorted into 4 treatment groups of 5 or 10 mice each based on body weight, as shown in Table 2. All mice were shaved on the right flank. Group I was dosed IV with 0.2 cc/mouse saline and groups 2-4 were dosed IV with 20 mg/kg of the appropriate test article in a 0.2 cc/mouse volume as indicated in Table 2.

TABLE 2 Study Design of Hair Growth in Wildtype Mice Study Group Dose Group Sequence 1 Saline 2 FGFR2-ECD-Fc SEQ ID NO: 32 3 FGFR3-ECD-Fc SEQ ID NO: 33 4 FGFR4-ECD-Fc SEQ ID NO: 31 (ABMut1)

On day 14 post initial dose, animals were observed for hair growth. As shown in FIG. 3A, marked hair growth was observed in 4 of 5 animals in group 3 (FGFR3-ECD-Fc) and 2 of 5 animals in group 4 (FGFR4-ECD-Rc; ABMut1) compared to the control group 1 (Saline) as indicated by pigmented skin and partial hair regrowth. There was no observable hair growth in group 2 (FGFR2-ECD-Fc).

The animals were euthanized on the same day and a 2 cm2 skin biopsy was harvested and fixed in neutral buffered saline for 12 hours. Samples were paraffin embedded and structural differences were visualized with Haematoxylin/Eosin staining (Gladstone Institute Histology Core, San Francisco, Calif.). Structural differences were observed in animals from groups 3 and 4 as demonstrated by elongation of the dermal papilla (*) into the fatty layer of the dermis () (FIG. 3B), suggesting re-entry into the anagen or growth phase of the hair cycle.

Example 7 An FGFR3 ECD Fusion Molecule Promotes Hair Growth in Mice in a Dose Dependent Manner

At exactly 61 days of age, female C57B1.6 mice (Charles River Labs, Wilmington, Mass.) were weighed and sorted into 7 groups of 10 mice each as demonstrated in the chart below. The entire back and belly of all mice was shaved and the animals were dosed subcutaneously with 0.1 mg/kg, 1 mg/kg or 10 mg/kg of FGFR3-ECD-Fc in a 0.05 cc volume directly in the center of the belly along the midline according to the chart below.

TABLE 3 Study Design of Hair Growth at Increasing Doses Dose Group Dose Group (mg/kg) N 1 Vehicle 0 10 2 FGFR3-ECD-Fc 0.1 10 3 FGFR3-ECD-Fc 1 10 4 FGFR3-ECD-Fc 10 10

On Day 13 post initial dose, mice were euthanized, and the area of skin that was shaved on day 0 was removed (pelt), tacked down flat and photographed. Photographs of mouse pelts were analyzed for hair growth using Image J (National Institutes of Health, Bethesda, Md.). Hair growth was calculated by measuring the percentage area of hair re-growth within a given pelt per total area of the pelt. As shown in FIG. 4, marked hair growth was observed in groups 3 and 4 compared to group 1. Additionally, significantly more hair growth was observed in groups 3 and 4 compared to group 2 and in group 4 compared to group 3, demonstrating that FGFR3-ECD-Fc stimulated hair growth in a dose dependent manner. FIG. 4 represents the average percentage of hair growth per dose group.

INDUSTRIAL APPLICABILITY

The FGFR ECDs described herein can be used to promote hair growth, which may be useful to subjects suffering from hair loss.

TABLE OF SEQUENCES

Table 4 provides certain sequences discussed herein. Solely for the sake of simplicity and not for any limiting reason, all FGFR sequences are shown without the signal peptide unless otherwise indicated.

TABLE 4 Sequences and Descriptions SEQ. ID. NO. Description Sequence  1 FGFR3 IIIc ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG NSIGFSHHSA WLVVLPAEEE LVEADEAGSV YAGILSYGVG FFLFILVVAA VTLCRLRSPP KKGLGSPTVH KISRFPLKRQ VSLESNASMS SNTPLVRIAR LSSGEGPTLA NVSELELPAD PKWELSRARL TLGKPLGEGC FGQVVMAEAI GIDKDRAAKP VTVAVKMLKD DATDKDLSDL VSEMEMMKMI GKHKNIINLL GACTQGGPLY VLVEYAAKGN LREFLRARRP PGLDYSFDTC KPPEEQLTFK DLVSCAYQVA RGMEYLASQK CIHRDLAARN VLVTEDNVMK IADFGLARDV HNLDYYKKTT NGRLPVKWMA PEALFDRVYT HQSDVWSFGV LLWEIFTLGG SPYPGIPVEE LFKLLKEGHR MDKPANCTHD LYMIMRECWH AAPSQRPTFK QLVEDLDRVL TVTSTDEYLD LSAPFEQYSP GGQDTPSSSS SGDDSVFAHD LLPPAPPSSG GSRT  2 FGFR3 Δ8-10 ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKVS LESNASMSSN TPLVRIARLS SGEGPTLANV SELELPADPK WELSRARLTL GKPLGEGCFG QVVMAEAIGI DKDRAAKPVT VAVKMLKDDA TDKDLSDLVS EMEMMKMIGK HKNIINLLGA CTQGGPLYVL VEYAAKGNLR EFLRARRPPG LDYSFDTCKP PEEQLTFKDL VSCAYQVARG MEYLASQKCI HRDLAARNVL VTEDNVMKIA DFGLARDVHN LDYYKKTTNG RLPVKWMAPE ALFDRVYTHQ SDVWSFGVLL WEIFTLGGSP YPGIPVEELF KLLKEGHRMD KPANCTHDLY MIMRECWHAA PSQRPTFKQL VEDLDRVLTV TSTDEYLDLS APFEQYSPGG QDTPSSSSSG DDSVFAHDLL PPAPPSSGGS RT  3 FGFR3 IIIb ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKSW ISESVEADVR LRLANVSERD GGEYLCRATN FIGVAEKAFW LSVHGPRAAE EELVEADEAG SVYAGILSYG VGFFLFILVV AAVTLCRLRS PPKKGLGSPT VHKISRFPLK RQVSLESNAS MSSNTPLVRI ARLSSGEGPT LANVSELELP ADPKWELSRA RLTLGKPLGE GCFGQVVMAE AIGIDKDRAA KPVTVAVKML KDDATDKDLS DLVSEMEMMK MIGKHKNIIN LLGACTQGGP LYVLVEYAAK GNLREFLRAR RPPGLDYSFD TCKPPEEQLT FKDLVSCAYQ VARGMEYLAS QKCIHRDLAA RNVLVTEDNV MKIADFGLAR DVHNLDYYKK TTNGRLPVKW MAPEALFDRV YTHQSDVWSF GVLLWEIFTL GGSPYPGIPV EELFKLLKEG HRMDKPANCT HDLYMIMREC WHAAPSQRPT FKQLVEDLDR VLTVTSTDEY LDLSAPFEQY SPGGQDTPSS SSSGDDSVFA HDLLPPAPPS SGGSRT  4 FGFR3-IIIb ECD ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKSW ISESVEADVR LRLANVSERD GGEYLCRATN FIGVAEKAFW LSVHGPRAAE EELVEADEAG SVYAG  5 FGFR3-IIIc ECD ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG NSIGFSHHSA WLVVLPAEEE LVEADEAGSV YAG  6 FGFR3-Δ8-10 ECD ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLK  7 FGFR3-IIIb ECD + ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP Fc PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKSW ISESVEADVR LRLANVSERD GGEYLCRATN FIGVAEKAFW LSVHGPRAAE EELVEADEAG SVYAGEPKSS DKTHTCPPCP APELLGGPSV FLFPPKPKDT LMISRTPEVT CVVVDVSHED PEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLH QDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYT LPPSRDELTK NQVSLTCLVK GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHE ALHNHYTQKS LSLSPGK  8 FGFR3-IIIb ECD + ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP GS linker + Fc PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKSW ISESVEADVR LRLANVSERD GGEYLCRATN FIGVAEKAFW LSVHGPRAAE EELVEADEAG SVYAGGSEPK SSDKTHTCPP CPAPELLGGP SVFLFPPKPK DTLMISRTPE VTCVVVDVSH EDPEVKFNWY VDGVEVHNAK TKPREEQYNS TYRVVSVLTV LHQDWLNGKE YKCKVSNKAL PAPIEKTISK AKGQPREPQV YTLPPSRDEL TKNQVSLTCL VKGFYPSDIA VEWESNGQPE NNYKTTPPVL DSDGSFFLYS KLTVDKSRWQ QGNVFSCSVM HEALHNHYTQ KSLSLSPGK  9 FGFR3-IIIc ECD + ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP Fc PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG NSIGFSHHSA WLVVLPAEEE LVEADEAGSV YAGEPKSSDK THTCPPCPAP ELLGGPSVFL FPPKPKDTLM ISRTPEVTCV VVDVSHEDPE VKFNWYVDGV EVHNAKTKPR EEQYNSTYRV VSVLTVLHQD WLNGKEYKCK VSNKALPAPI EKTISKAKGQ PREPQVYTLP PSRDELTKNQ VSLTCLVKGF YPSDIAVEWE SNGQPENNYK TTPPVLDSDG SFFLYSKLTV DKSRWQQGNV FSCSVMHEAL HNHYTQKSLS LSPGK 10 FGFR3-IIIc ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP ECD + GS linker + PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE Fc DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG NSIGFSHHSA WLVVLPAEEE LVEADEAGSV YAGGSEPKSS DKTHTCPPCP APELLGGPSV FLFPPKPKDT LMISRTPEVT CVVVDVSHED PEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLH QDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYT LPPSRDELTK NQVSLTCLVK GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHE ALHNHYTQKS LSLSPGK 11 FGFR3-IIIc ECD + ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP Fc(R3Mut1) PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE (FGFR3-IIIc ECD DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE with C-terminal DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP deletion of VYAG, TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD fused to Fc) RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG NSIGFSHHSA WLVVLPAEEE LVEADEAGSE PKSSDKTHTC PPCPAPELLG GPSVFLFPPK PKDTLMISRT PEVTCVVVDV SHEDPEVKFN WYVDGVEVHN AKTKPREEQY NSTYRVVSVL TVLHQDWLNG KEYKCKVSNK ALPAPIEKTI SKAKGQPREP QVYTLPPSRD ELTKNQVSLT CLVKGFYPSD IAVEWESNGQ PENNYKTTPP VLDSDGSFFL YSKLTVDKSR WQQGNVFSCS VMHEALHNHY TQKSLSLSPG K 12 FGFR3-IIIc ECD + ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP Fc (R3Mut2) PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE (FGFR3-IIIc ECD DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE with C-terminal DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP deletion of TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD EAGSVYAG, fused RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN to Fc) QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG NSIGFSHHSA WLVVLPAEEE LVEADEPKSS DKTHTCPPCP APELLGGPSV FLFPPKPKDT LMISRTPEVT CVVVDVSHED PEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLH QDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYT LPPSRDELTK NQVSLTCLVK GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHE ALHNHYTQKS LSLSPGK 13 FGFR3-IIIc ECD + ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP Fc(R3Mut3) PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE (FGFR3-IIIc ECD DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE with C-terminal DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP deletion of TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD DEAGSVYAG, RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN fused to Fc) QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG NSIGFSHHSA WLVVLPAEEE LVEAEPKSSD KTHTCPPCPA PELLGGPSVF LFPPKPKDTL MISRTPEVTC VVVDVSHEDP EVKFNWYVDG VEVHNAKTKP REEQYNSTYR VVSVLTVLHQ DWLNGKEYKC KVSNKALPAP IEKTISKAKG QPREPQVYTL PPSRDELTKN QVSLTCLVKG FYPSDIAVEW ESNGQPENNY KTTPPVLDSD GSFFLYSKLT VDKSRWQQGN VFSCSVMHEA LHNHYTQKSL SLSPGK 14 FGFR3-IIIc ECD + ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP Fc(R3Mut4) PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE (FGFR3-IIIc ECD DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE with C-terminal DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP deletion of TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD LVEADEAGSVYA RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN G, fused to Fc) QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG NSIGFSHHSA WLVVLPAEEE EPKSSDKTHT CPPCPAPELL GGPSVFLFPP KPKDTLMISR TPEVTCVVVD VSHEDPEVKF NWYVDGVEVH NAKTKPREEQ YNSTYRVVSV LTVLHQDWLN GKEYKCKVSN KALPAPIEKT ISKAKGQPRE PQVYTLPPSR DELTKNQVSL TCLVKGFYPS DIAVEWESNG QPENNYKTTP PVLDSDGSFF LYSKLTVDKS RWQQGNVFSC SVMHEALHNH YTQKSLSLSP GK 15 FGFR3-IIIc ECD + ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP Fc(R3Mut5) PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE (FGFR3-IIIc ECD DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE with C-terminal DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP deletion of TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD VLPAEEELVEADE RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN AGSVYAG, fused to QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG Fc) TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG NSIGFSHHSA WLVEPKSSDK THTCPPCPAP ELLGGPSVFL FPPKPKDTLM ISRTPEVTCV VVDVSHEDPE VKFNWYVDGV EVHNAKTKPR EEQYNSTYRV VSVLTVLHQD WLNGKEYKCK VSNKALPAPI EKTISKAKGQ PREPQVYTLP PSRDELTKNQ VSLTCLVKGF YPSDIAVEWE SNGQPENNYK TTPPVLDSDG SFFLYSKLTV DKSRWQQGNV FSCSVMHEAL HNHYTQKSLS LSPGK 16 Fc C237S EPKSSDKTHT CPPCPAPELL GGPSVFLFPP KPKDTLMISR (GI17939658_233- TPEVTCVVVD VSHEDPEVKF NWYVDGVEVH NAKTKPREEQ 464_C237S) YNSTYRVVSV LTVLHQDWLN GKEYKCKVSN KALPAPIEKT ISKAKGQPRE PQVYTLPPSR DELTKNQVSL TCLVKGFYPS DIAVEWESNG QPENNYKTTP PVLDSDGSFF LYSKLTVDKS RWQQGNVFSC SVMHEALHNH YTQKSLSLSP GK 17 Fc ERKCCVECPP CPAPPVAGPS VFLFPPKPKD TLMISRTPEV (GI34528298_241- TCVVVDVSHE DPEVQFNWYV DGVEVHNAKT KPREEQFNST 468) FRVVSVLTVV HQDWLNGKEY KCKVSNKGLP APIEKTISKT KGQPREPQVY TLPPSREEMT KNQVSLTCLV KGFYPSDIAV EWESNGQPEN NYKTTPPMLD SDGSFFLYSK LTVDKSRWQQ GNVFSCSVMH EALHNHYTQK SLSLSPGK 18 Fc ESKYGPPCPS CPAPEFLGGP SVFLFPPKPK DTLMISRTPE (GI19684073_245- VTCVVVDVSQ EDPEVQFNWY VDGVEVHNAK TKPREEQFNS 473) TYRVVSVLTV LHQDWLNGKE YKCKVSNKGL PSSIEKTISK AKGQPREPQV YTLPPSQEEM TKNQVSLTCL VKGFYPSDIA VEWESNGQPE NNYKTTPPVL DSDGSFFLYS RLTVDKSRWQ EGNVFSCSVM HEALHNHYTQ KSLSLSLGK 19 FGFR1 signal MWSWKCLLFWAVLVTATLCTA peptide 20 FGFR2 signal MVSWGRFICLVVVTMATLSLA peptide 21 FGFR3 signal MGAPACALALCVAVAIVAGASS peptide 22 FGFR4 signal MRLLLALLGILLSVPGPPVLS peptide 23 FGFR3 D1-D2 linker DAPSSGDDED GEDEAEDTGV DTG 24 FGFR3 acid box DDEDGED region 1 25 FGFR3 acid box DDEDGEDE region 2 26 FGFR3 acid box DDEDGEDEAE region 3 27 FGFR3 acid box DDEDGEDEAED region4 28 FGFR3-IIIc ECD ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP R3(110-117): PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE R1(105-112) + Fc DSGAYSCRQR LTQRVLCHFS VRVTDAPSSE DDDDDDDEAE (FGFR3-IIIc ECD DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP D1-D2 linker TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD chimera fused to Fc) RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG NSIGFSHHSA WLVVLPAEEE LVEADEAGSV YAGEPKSSDK THTCPPCPAP ELLGGPSVFL FPPKPKDTLM ISRTPEVTCV VVDVSHEDPE VKFNWYVDGV EVHNAKTKPR EEQYNSTYRV VSVLTVLHQD WLNGKEYKCK VSNKALPAPI EKTISKAKGQ PREPQVYTLP PSRDELTKNQ VSLTCLVKGF YPSDIAVEWE SNGQPENNYK TTPPVLDSDG SFFLYSKLTV DKSRWQQGNV FSCSVMHEAL HNHYTQKSLS LSPGK 29 FGFR3-IIIc ECD MGAPACALAL CVAVAIVAGA SSESLGTEQR VVGRAAEVPG with signal peptide PEPGQQEQLV FGSGDAVELS CPPPGGGPMG PTVWVKDGTG LVPSERVLVG PQRLQVLNAS HEDSGAYSCR QRLTQRVLCH FSVRVTDAPS SGDDEDGEDE AEDTGVDTGA PYWTRPERMD KKLLAVPAAN TVRFRCPAAG NPTPSISWLK NGREFRGEHR IGGIKLRHQQ WSLVMESVVP SDRGNYTCVV ENKFGSIRQT YTLDVLERSP HRPILQAGLP ANQTAVLGSD VEFHCKVYSD AQPHIQWLKH VEVNGSKVGP DGTPYVTVLK TAGANTTDKE LEVLSLHNVT FEDAGEYTCL AGNSIGFSHH SAWLVVLPAE EELVEADEAG SVYAG 30 FGFR3-IIIc ECD Δ3 ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP (FGFR3-IIIc ECD PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE with a deleion of the DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE C-terminal 3 amino DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP acids YAG) TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG NSIGFSHHSA WLVVLPAEEE LVEADEAGSV 31 FGFR4 ECD Δ17 R1 LEASEEVELE PCLAPSLEQQ EQELTVALGQ PVRLCCGRAE D1-D2 linker RGGHWYKEGS RLAPAGRVRG WRGRLEIASF LPEDAGRYLC chimera + Fc LARGSMIVLQ NLTLITGDAL PSSEDDDDDD DSSSEEKETD (also called FGFR4 NTKPNPVAPY WTHPQRMEKK LHAVPAGNTV KFRCPAAGNP ECD (ABMut1: delta TPTIRWLKDG QAFHGENRIG GIRLRHQHWS LVMESVVPSD 17)-Fc and ABMut1) RGTYTCLVEN AVGSIRYNYL LDVLERSPHR PILQAGLPAN TTAVVGSDVE LLCKVYSDAQ PHIQWLKHIV INGSSFGADG FPYVQVLKTA DINSSEVEVL YLRNVSAEDA GEYTCLAGNS IGLSYQSAWL TVLPEPKSSD KTHTCPPCPA PELLGGPSVF LFPPKPKDTL MISRTPEVTC VVVDVSHEDP EVKFNWYVDG VEVHNAKTKP REEQYNSTYR VVSVLTVLHQ DWLNGKEYKC KVSNKALPAP IEKTISKAKG QPREPQVYTL PPSRDELTKN QVSLTCLVKG FYPSDIAVEW ESNGQPENNY KTTPPVLDSD GSFFLYSKLT VDKSRWQQGN VFSCSVMHEA LHNHYTQKSL SLSPGK 32 FGFR2 ECD Δ3 + RPSFSLVEDT TLEPEEPPTK YQISQPEVYV AAPGESLEVR GS linker + Fc CLLKDAAVIS WTKDGVHLGP NNRTVLIGEY LQIKGATPRD SGLYACTASR TVDSETWYFM VNVTDAISSG DDEDDTDGAE DFVSENSNNK RAPYWTNTEK MEKRLHAVPA ANTVKFRCPA GGNPMPTMRW LKNGKEFKQE HRIGGYKVRN QHWSLIMESV VPSDKGNYTC VVENEYGSIN HTYHLDVVER SPHRPILQAG LPANASTVVG GDVEFVCKVY SDAQPHIQWI KHVEKNGSKY GPDGLPYLKV LKAAGVNTTD KEIEVLYIRN VTFEDAGEYT CLAGNSIGIS FHSAWLTVLP APGREKEITA SPDGSEPKSS DKTHTCPPCP APELLGGPSV FLFPPKPKDT LMISRTPEVT CVVVDVSHED PEVKFNWYVD GVEVHNAKTK PREEQYNSTY RVVSVLTVLH QDWLNGKEYK CKVSNKALPA PIEKTISKAK GQPREPQVYT LPPSRDELTK NQVSLTCLVK GFYPSDIAVE WESNGQPENN YKTTPPVLDS DGSFFLYSKL TVDKSRWQQG NVFSCSVMHE ALHNHYTQKS LSLSPGK 33 FGFR3-IIIc ECD Δ3 + ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP GS linker + Fc PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE (FGFR3-IIIc ECD DSGAYSCRQR LTQRVLCHFS VRVTDAPSSG DDEDGEDEAE with a deletion of the DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP C-terminal 3 amino TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD acids YAG, fused to RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN Fc with a GS linker) QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG NSIGFSHHSA WLVVLPAEEE LVEADEAGSV GSEPKSSDKT HTCPPCPAPE LLGGPSVFLF PPKPKDTLMI SRTPEVTCVV VDVSHEDPEV KFNWYVDGVE VHNAKTKPRE EQYNSTYRVV SVLTVLHQDW LNGKEYKCKV SNKALPAPIE KTISKAKGQP REPQVYTLPP SRDELTKNQV SLTCLVKGFY PSDIAVEWES NGQPENNYKT TPPVLDSDGS FFLYSKLTVD KSRWQQGNVF SCSVMHEALH NHYTQKSLSL SPGK 34 FGFR3-IIIc ECD ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP R3(110-117): PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE R1(105-112) DSGAYSCRQR LTQRVLCHFS VRVTDAPSSE DDDDDDDEAE (FGFR3-IIIc ECD DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP D1-D2 linker TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD chimera) RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG NSIGFSHHSA WLVVLPAEEE LVEADEAGSV YAG 35 FGFR3-IIIc ECD Δ3 ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP R3(110-117): PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE R1(105-112) + Fc DSGAYSCRQR LTQRVLCHFS VRVTDAPSSE DDDDDDDEAE (FGFR3-IIIc ECD DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP D1-D2 linker TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD chimera with a RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN deletion of the C- QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG terminal three amino TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG acids YAG, fused to NSIGFSHHSA WLVVLPAEEE LVEADEAGSV EPKSSDKTHT Fc) CPPCPAPELL GGPSVFLFPP KPKDTLMISR TPEVTCVVVD VSHEDPEVKF NWYVDGVEVH NAKTKPREEQ YNSTYRVVSV LTVLHQDWLN GKEYKCKVSN KALPAPIEKT ISKAKGQPRE PQVYTLPPSR DELTKNQVSL TCLVKGFYPS DIAVEWESNG QPENNYKTTP PVLDSDGSFF LYSKLTVDKS RWQQGNVFSC SVMHEALHNH YTQKSLSLSP GK 36 FGFR3-IIIc ECD Δ3 ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP R3(110-117): PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE R1(105-112) DSGAYSCRQR LTQRVLCHFS VRVTDAPSSE DDDDDDDEAE (FGFR3-IIIc ECD DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP D1-D2 linker TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD chimera with a RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN deletion of the C- QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG terminal three amino TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG acids YAG) NSIGFSHHSA WLVVLPAEEE LVEADEAGSV 37 FGFR3-IIIc ECD Δ3 ESLGTEQRVV GRAAEVPGPE PGQQEQLVFG SGDAVELSCP R3(110-117): PPGGGPMGPT VWVKDGTGLV PSERVLVGPQ RLQVLNASHE R1(105-112) + GS DSGAYSCRQR LTQRVLCHFS VRVTDAPSSE DDDDDDDEAE linker + Fc DTGVDTGAPY WTRPERMDKK LLAVPAANTV RFRCPAAGNP (FGFR3-IIIc ECD TPSISWLKNG REFRGEHRIG GIKLRHQQWS LVMESVVPSD D1-D2 linker RGNYTCVVEN KFGSIRQTYT LDVLERSPHR PILQAGLPAN chimera with a QTAVLGSDVE FHCKVYSDAQ PHIQWLKHVE VNGSKVGPDG deletion of the C- TPYVTVLKTA GANTTDKELE VLSLHNVTFE DAGEYTCLAG terminal three amino NSIGFSHHSA WLVVLPAEEE LVEADEAGSV GSEPKSSDKT acids YAG, fused to HTCPPCPAPE LLGGPSVFLF PPKPKDTLMI SRTPEVTCVV Fc with a GS linker) VDVSHEDPEV KFNWYVDGVE VHNAKTKPRE EQYNSTYRVV (also called SVLTVLHQDW LNGKEYKCKV SNKALPAPIE KTISKAKGQP FGFR3ECD(FGFR3 REPQVYTLPP SRDELTKNQV SLTCLVKGFY PSDIAVEWES (110-117): NGQPENNYKT TPPVLDSDGS FFLYSKLTVD KSRWQQGNVF FGFR1(105-112): SCSVMHEALH NHYTQKSLSL SPGK delta3)-GS linker-Fc and R3(110- 117): R1(105-112)) 38 FGFR4 D1-D2 linker DSLTSSNDDED PKSHRDPSNR HSYPQQ 39 FGFR1 D1-D2 linker DALPSSEDDDD DDDSSSEEKE TDNTKPNPV 40 FGFR2 D1-D2 linker DAISSGDDED DTDGAEDFVS ENSNNKR 41 FGFR3 acid box DDEDGE 42 FGFR3 long acid box GDDEDGEDEA ED 43 FGFR3 short acid DDED box

Claims

1.-40. (canceled)

41. A method of promoting hair growth comprising administering an FGFR3 ECD and at least one additional agent selected from minoxidil, finasteride, and dutasteride to a subject in an amount sufficient to promote hair growth.

42. A kit comprising one or more containers, wherein each container contains one or more doses of an FGFR3 ECD for promoting hair growth.

43. A vector comprising a polynucleotide that encodes an FGFR3 ECD, wherein the vector is for in vivo expression of FGFR3 ECD in an animal.

Patent History
Publication number: 20140274898
Type: Application
Filed: Feb 20, 2014
Publication Date: Sep 18, 2014
Applicant: FIVE PRIME THERAPEUTICS, INC. (South San Francisco, CA)
Inventors: Thomas Brennan (Saratoga, CA), Robert Dean (Alameda, CA), W. Michael Kavanaugh (Orinda, CA), Janine Powers (Alameda, CA)
Application Number: 14/185,086