WO2014152090A1 - Compositions and treatments of metabolic disorders using fgf binding protein 3 and fgf 19 - Google Patents

Compositions and treatments of metabolic disorders using fgf binding protein 3 and fgf 19 Download PDF

Info

Publication number
WO2014152090A1
WO2014152090A1 PCT/US2014/026938 US2014026938W WO2014152090A1 WO 2014152090 A1 WO2014152090 A1 WO 2014152090A1 US 2014026938 W US2014026938 W US 2014026938W WO 2014152090 A1 WO2014152090 A1 WO 2014152090A1
Authority
WO
WIPO (PCT)
Prior art keywords
fgf19
fgfbp3
complex
subject
growth factor
Prior art date
Application number
PCT/US2014/026938
Other languages
French (fr)
Inventor
Anton Wellstein
Original Assignee
Georgetown University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Georgetown University filed Critical Georgetown University
Publication of WO2014152090A1 publication Critical patent/WO2014152090A1/en
Priority to US14/853,482 priority Critical patent/US9789160B2/en
Priority to US15/784,730 priority patent/US20180214514A1/en
Priority to US16/840,046 priority patent/US20200246425A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1825Fibroblast growth factor [FGF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals

Definitions

  • a computer readable text file entitled “036681-5014-WO-SequenceListing.txt,” created on or about 13 March 2014 with a file size of about 6 kb contains the sequence listing for this application and is hereby incorporated by reference in its entirety.
  • the invention relates to methods of treating a metabolic disorder in a subject, the method comprising administering a complex of fibroblast growth factor 19 (FGF19), fibroblast growth factor 21 (FGF21) or fibroblast growth factor 23 (FGF23), plus fibroblast growth factor binding protein 3 (FGFBP3) to a subject in need of treatment of a metabolic disorder.
  • FGF19 fibroblast growth factor 19
  • FGF21 fibroblast growth factor 21
  • FGF23 fibroblast growth factor 23
  • FGFBP3 fibroblast growth factor binding protein 3
  • Fibroblast growth factor 19 and other members of the FGF19 family are involved in the regulation of metabolism. FGF19 has also been recently described as a sensitizer to insulin. In addition, there some members of the FGF19 family interact with the co-receptor klotho to affect metabolism and in response to bile acid production.
  • FGFBP FGF-binding proteins
  • the invention relates to methods of treating a metabolic disorder in a subject, the method comprising administering a complex of fibroblast growth factor 19 (FGF19), fibroblast growth factor 21 (FGF21) or fibroblast growth factor 23 (FGF23), plus fibroblast growth factor binding protein 3 (FGFBP3) to a subject in need of treatment of a metabolic disorder.
  • FGF19 fibroblast growth factor 19
  • FGF21 fibroblast growth factor 21
  • FGF23 fibroblast growth factor 23
  • FGFBP3 fibroblast growth factor binding protein 3
  • the invention also relates to methods of lowering blood glucose levels in a subject, the method comprising administering a complex of FGF19, FGF21 or FGF23 and FGFBP3 to a subject in need of lowering of blood glucose levels.
  • the invention also relates to methods of lowering a subject's body weight, the method comprising administering a complex of FGF19, FGF21 or FGF23 and FGFBP3 to a subject that is in need of lowering its body weight.
  • A Five treatments with FGF19 reduced blood glucose levels at 180 minutes after the beginning of the glucose tolerance test, and the curve returned to the baseline 9 days after receiving no treatment.
  • B Five treatments with FGF19+BP3 improved the glucose tolerance at 60, 120, and 180 minutes with a sustained effect for 9 days.
  • A The glucose levels were not changed 2 days after a single dose of FGF19 alone.
  • Filled Squares Baseline, Open Squares: single dose treatment.
  • B In the FGF19+BP3 group, the glucose levels were significantly reduced at 15, 30 and 60 minutes post-injection of glucose.
  • C The area under the curve of the glucose tolerance test (AUC) was reduced 2 days after a single treatment in FGF19+BP3 group.
  • Filled Squares Baseline, Open Squares: Single treatment. * denotes P ⁇ 0.05, ** P ⁇ 0.01.
  • FIGURE 3 depicts the effects of acute single doses of BP3, FGF19, and the complex of BP3+FGF19 in ob/ob mice in response to a glucose tolerance test.
  • Figures 3A, 3B show the effect when the test was conducted two hours after treatment.
  • Figure 3C shows the effect when the test was performed 24 hours after treatment. Even 7 days of dosing of BP3 alone with a single daily dose of BP3 lacked an effect on the baseline blood glucose or on the IPGTT test ( Figure 3D).
  • FIGURE 4 depicts the body weight changes in ob/ob mice following single or multiple treatments with FGF19 or FGF19+BP3.
  • B The percentage changes in body weight is also different between the two groups immediately after 5 daily treatments (6.04 ⁇ 2.33% vs. -3.40 ⁇ 2.50%). Filled Squares: single treatment; Open Squares: 5 treatments (one dose/day). * denotes P ⁇ 0.05.
  • FIGURE 5 depicts the presence of BP3 enhancing the ability of FGF19 to induce phosphorylation of Erkl/2 in HepG2 cells.
  • HepG2 cells were treated with FGF19 ⁇ BP3 or the negative control protein MBP.
  • pERKl/2 was measured in cell lysates at different times after treatment.
  • the invention relates to methods of treating a metabolic disorder in a subject, the methods comprising administering a complex of fibroblast growth factor 19 (FGF19), fibroblast growth factor 21 (FGF21) or fibroblast growth factor 23 (FGF23), plus fibroblast growth factor binding protein 3 (FGFBP3) or variant thereof to a subject in need of treatment of a metabolic disorder.
  • FGF19 fibroblast growth factor 19
  • FGF21 fibroblast growth factor 21
  • FGF23 fibroblast growth factor 23
  • FGFBP3 fibroblast growth factor binding protein 3
  • Fibroblast growth factor 19 is a well-known member of the FGF family of proteins that is involved in nutrient metabolism.
  • FGF19 is a protein of 216 amino acids, with the signal peptide being amino acids 1-24.
  • fibroblast growth factor 19 or FGF19 can mean the full length FGF19 with or without the N-terminus signal sequence.
  • the mouse ortholog to human FGF19 is known as FGF15 and is 218 amino acids in length, including the 25-amino acid signal sequence at the N- terminus.
  • fibroblast growth factor 15 or “FGF15” is used to indicate any ortholog to FGF19 and can include the full length amino acid sequence, with or without the N-terminus signal sequence.
  • FGF19 herein will also include a reference to its art- accepted orthologs, such as mouse FGF15.
  • concentration of FGF19 is upregulated after feeding and binds to FGF Receptor 4 (FGFR4).
  • FGF19 is synthesized in the distal small intestine in response to uptake of bile acids via the nuclear bile receptor and controlled by food intake.
  • FGF21 expression in the liver and fat tissues is also regulated by the feeding or starving status and function in a temporal cascade with insulin, glucagon and other hormones to regulate responses to nutrition (Potthoff, et al., Genes and Development 2012).
  • FGFBP3 The fibroblast growth factor binding protein 3
  • FGFBP3 is a secreted protein that binds to FGF19 and is believed to act as a co-receptor with FGFR4.
  • FGFBP3 is not known to bind to other FGF receptors to any significant extent.
  • SEQ ID NO:l The full length amino acid sequence of human FGFBP3 is shown below as SEQ ID NO:l.
  • SEQ ID NO:2 The full length amino acid sequence of human FGFBP3, without the 26 amino- acid signal sequence, is shown below as SEQ ID NO:2.
  • SEQ ID NO:3 The C-terminus of FGFBP3 is shown below as SEQ ID NO:3.
  • FGFBP3 means a peptide that comprises the amino acid sequence of SEQ ID NO:3 or a variant thereof that still retains activity similar to the wild-type FGFBP3.
  • amino acid sequence of SEQ ID NOs:l and 2 are just two embodiments of the term FGFBP3 as it is used herein. "Variants" of FGFBP3 are discussed below.
  • the present invention is directed to methods that include administration of a complex of FGF19 and FGFBP3.
  • the term "complex" as it relates to FGF19 and FGFBP3 means the presence of both FGFBP3 and FGF19.
  • the FGF19 and FGFBP3 can, but need not, be specifically interacting, i.e., specifically binding to one another.
  • the FGF19 and FGFBP3 within the complex are specifically binding to one another.
  • the FGF19 and FGFBP3 within the complex are not necessarily specifically binding to one another.
  • full length FGFBP3 (a peptide amino acid sequence of SEQ ID NO:l) is complexed with FGF19.
  • the FGFBP3 in the complex comprises a peptide with an amino acid sequence that is 100% identical to the amino acid sequence of SEQ ID NO:l.
  • the FGFBP3 in the complex consists of a peptide with an amino acid sequence that is 100% identical to the amino acid sequence of SEQ ID NO:l.
  • full length FGFBP3 without the signal sequence (a peptide amino acid sequence of SEQ ID NO:2) is complexed with FGF19.
  • the FGFBP3 in the complex comprises a peptide with an amino acid sequence that is 100% identical to the amino acid sequence of SEQ ID NO:2.
  • the FGFBP3 in the complex consists of a peptide with an amino acid sequence that is 100% identical to the amino acid sequence of SEQ ID NO:2.
  • the C-terminal FGFBP3 (a peptide amino acid sequence of SEQ ID NO:3) is complexed with FGF19.
  • the FGFBP3 in the complex comprises a peptide with an amino acid sequence that is 100% identical to the amino acid sequence of SEQ ID NO:3.
  • the FGFBP3 in the complex consists of a peptide with an amino acid sequence that is 100% identical to the amino acid sequence of SEQ ID NO:3.
  • polypeptide and “protein” are used interchangeably herein.
  • an "isolated polypeptide” is intended to mean a polypeptide that has been completely or partially removed from its native environment. For example, polypeptides that have been removed or purified from cells are considered isolated. In addition, recombinantly produced polypeptides molecules contained in host cells are considered isolated for the purposes of the present invention. Moreover, a peptide that is found in a cell, tissue or matrix in which it is not normally expressed or found is also considered as “isolated” for the purposes of the present invention. Similarly, polypeptides that have been synthesized are considered to be isolated polypeptides. "Purified,” on the other hand is well understood in the art and generally means that the peptides are substantially free of cellular material, cellular components, chemical precursors or other chemicals beyond, perhaps, buffer or solvent.
  • Substantially free is not intended to mean that other components beyond the novel peptides are undetectable.
  • FGFBP3 variants comprise an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequences of SEQ ID NO: 3.
  • the FGFBP3 variant consists of a peptide with an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequences of SEQ ID NO: 3.
  • FGFBP3 variants comprise an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequences of SEQ ID NO: 1.
  • the FGFBP3 variant consists of a peptide with an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequences of SEQ ID NO: 1.
  • FGFBP3 variants comprise an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequences of SEQ ID NO: 2.
  • the FGFBP3 variant consists of a peptide with an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequences of SEQ ID NO: 2.
  • a metabolic disorder can be any disorder associated with metabolism, and examples include but are not limited to, obesity, central obesity, insulin resistance, glucose intolerance, abnormal glycogen metabolism, type 2 diabetes, hyperlipidemia, hypoalbuminemia,
  • the methods are directed towards treating at least one component of postprandial metabolism, such as, but not limited to hepatic glycogen synthesis, protein synthesis and clearance of plasma glucose.
  • twin and “phenotype” are used interchangeably herein and refer to any visible, detectable or otherwise measurable property of an organism such as symptoms of or susceptibility to a disorder.
  • phenotype are used herein to refer to symptoms of a metabolic disorder, or a susceptibility to an metabolic disorder.
  • traits of metabolic disorders include but are not limited to high total cholesterol, low high-density lipoprotein (HDL) cholesterol, impaired fasting glucose levels, insulin resistance, hyperproinsulinemia, central obesity, elevated triglyceride levels, postprandial glucose levels, elevated uric acid levels, thyroid dysfunction, increased body-mass index (BMI), hypertension, impaired glucose tolerance, alterations in hormone and peptide levels (e.g., leptin, ghrelin, obstatin, adiponectin, perilipin, omentin), interactions with substances involved in insulin signaling, lipid, amino acid and glucose metabolism, life expectancy, increased systemic inflammatory state (e.g., as reflected in levels of C-reactive protein, interleukin-6, and TNF-alpha), depression, and sleep disordered breathing.
  • HDL high-density lipoprotein
  • the peptide variants described herein are functional and capable of altering a subject's response in a glucose tolerance test when complexed with FGF19.
  • the FGFBP3 variants of the present invention when complexed with FGF19, have enhanced ability to alter a subject's response in a glucose tolerance test compared to FGF19 complexed with the wild-type FGFBP3.
  • the FGFBP3 variants of the present invention also have enhanced stability compared to the wild-type FGFBP3 regardless of their association with FGF19.
  • a polypeptide having an amino acid sequence at least, for example, about 95% "identical" to a reference an amino acid sequence is understood to mean that the amino acid sequence of the polypeptide is identical to the reference sequence except that the amino acid sequence may include up to about five modifications per each 100 amino acids of the reference amino acid sequence.
  • up to about 5% of the amino acid residues of the reference sequence may be deleted or substituted with another amino acid or a number of amino acids up to about 5% of the total amino acids in the reference sequence may be inserted into the reference sequence.
  • These modifications of the reference sequence may occur at the N- terminus or C-terminus positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among amino acids in the reference sequence or in one or more contiguous groups within the reference sequence.
  • identity is a measure of the identity of nucleotide sequences or amino acid sequences compared to a reference nucleotide or amino acid sequence. In general, the sequences are aligned so that the highest order match is obtained. "Identity” per se has an art-recognized meaning and can be calculated using well known techniques. While there are several methods to measure identity between two polynucleotide or polypeptide sequences, the term “identity” is well known to skilled artisans (Carillo (1988) J. Applied Math. 48, 1073).
  • Examples of computer program methods to determine identity and similarity between two sequences include, but are not limited to, GCG program package (Devereux (1984) Nucleic Acids Research 12, 387), BLASTP, ExPASy, BLASTN, FASTA (Atschul (1990) J. Mol. Biol. 215, 403) and FASTDB. Examples of methods to determine identity and similarity are discussed in Michaels (2011) Current Protocols in Protein Science, Vol. 1, John Wiley & Sons.
  • the algorithm used to determine identity between two or more polypeptides is BLASTP.
  • the algorithm used to determine identity between two or more polypeptides is FASTDB, which is based upon the algorithm of Brutlag (1990) Comp. App. Biosci. 6, 237-245).
  • FASTDB sequence alignment the query and reference sequences are amino sequences. The result of sequence alignment is in percent identity.
  • the percent identity is corrected by calculating the number of residues of the query sequence that are N-and C- terminus to the reference sequence that are not matched/aligned, as a percent of the total bases of the query sequence.
  • the results of the FASTDB sequence alignment determine matching/alignment.
  • the alignment percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score. This corrected score can be used for the purposes of determining how alignments "correspond" to each other, as well as percentage identity.
  • Residues of the reference sequence that extend past the N- or C-termini of the query sequence may be considered for the purposes of manually adjusting the percent identity score. That is, residues that are not matched/aligned with the N- or C-termini of the comparison sequence may be counted when manually adjusting the percent identity score or alignment numbering.
  • a 90 amino acid residue query sequence is aligned with a 100 residue reference sequence to determine percent identity.
  • the deletion occurs at the N-terminus of the query sequence and therefore, the FASTDB alignment does not show a match/alignment of the first 10 residues at the N- terminus.
  • the 10 unpaired residues represent 10% of the reference sequence (number of residues at the N- and C-termini not matched/total number of residues in the reference sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 residues were perfectly matched (100% alignment) the final percent identity would be 90% (100% alignment - 10% unmatched overhang).
  • a 90 residue query sequence is compared with a 100 reference sequence, except that the deletions are internal deletions.
  • the percent identity calculated by FASTDB is not manually corrected, since there are no residues at the N- or C- termini of the subject sequence that are not matched/aligned with the query.
  • a 110 amino acid query sequence is aligned with a 100 residue reference sequence to determine percent identity. The addition in the query occurs at the N-terminus of the query sequence and therefore, the FASTDB alignment may not show a match/alignment of the first 10 residues at the N-terminus. If the remaining 100 amino acid residues of the query sequence have 95% identity to the entire length of the reference sequence, the N-terminal addition of the query would be ignored and the percent identity of the query to the reference sequence would be 95%.
  • the terms “correspond(s) to” and “corresponding to,” as they relate to sequence alignment, are intended to mean enumerated positions within the reference protein, e.g., wild-type FGFBP3, and those positions in the variant or ortholog FGFBP3 that align with the positions with the reference protein.
  • the amino acid sequence of a subject FGFBP3 is aligned with the amino acid sequence of a reference FGFBP3, e.g., SEQ ID NO: 2
  • the amino acids in the subject sequence that "correspond to" certain enumerated positions of the reference sequence are those that align with these positions of the reference sequence, e.g., SEQ ID NO: 2, but are not necessarily in these exact numerical positions of the reference sequence.
  • the invention further embraces other species, preferably mammalian, homologs with amino acid sequences that correspond to FGFBP3.
  • Species homologs sometimes referred to as "orthologs,” in general, share at least 35%, 40%, 45%, 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the human version of the full length binding proteins or the full length binding proteins without the signal sequence.
  • Such corresponding sequences account for FGFBP3 from across a variety of species, such as canine, feline, mouse, rat, rabbit, monkey, etc.
  • FGFBP3 with an additional methionine residue at position -1 are contemplated, as are variants with additional methionine and lysine residues at positions -2 and -1 (Met ⁇ -Lys 1 - peptide).
  • Variants of FGFBP3 with additional Met, Met-Lys, or Lys residues (or one or more basic residues in general) are particularly useful for enhanced recombinant protein production in bacterial host cells.
  • variants resulting from insertion of the polynucleotide encoding FGFBP3 into an expression vector system are also contemplated.
  • variants usually insertions
  • the invention provides deletion variants wherein one or more amino acid residues in FGFBP3 are removed.
  • Deletions can be effected at one or both termini of the FGFBP3 peptide, or with removal of one or more non-terminal amino acid residues of the FGFBP3.
  • Deletion variants therefore, include all fragments of the FGFBP3 peptide.
  • the invention also relates to substitution variants of disclosed polypeptides of the invention. Substitution variants include those polypeptides wherein one or more amino acid residues of FGFBP3 are removed and replaced with alternative residues.
  • SEQ ID NOs: 1 or 2 are known to exist and the invention contemplates the use of these known variants in the methods described herein.
  • a variant of FGFBP3 wherein Alanine at position 107 of SEQ ID NO:l is replaced with Threonine (A107T) is included in the methods of the present invention.
  • Another variant of FGFBP3 wherein Glutamate at position 206 of SEQ ID NO:l is replaced with Valine (E206V) is included in the methods of the present invention.
  • positions 107 and 206 of SEQ ID NO:l correspond to positions 81 and 180 of SEQ ID NO:2.
  • the substitutions are conservative in nature; however, the invention embraces substitutions that are also non-conservative.
  • Conservative substitutions for this purpose may be defined as set out in the tables below. Amino acids can be classified according to physical properties and contribution to secondary and tertiary protein structure. A conservative substitution is recognized in the art as a substitution of one amino acid for another amino acid that has similar properties. Exemplary conservative substitutions are set out in below.
  • conservative amino acids can be grouped as described in Lehninger (1975) Biochemistry, Second Edition; Worth Publishers, pp. 71-77, as set forth below.
  • Val (V) lie, Leu, Met, Phe, Ala
  • peptides or polypeptides of the invention is intended to include polypeptides bearing modifications other than insertion, deletion, or substitution of amino acid residues.
  • the modifications may be covalent in nature, and include for example, chemical bonding with polymers, lipids, other organic and inorganic moieties.
  • Such derivatives may be prepared to increase circulating half-life of a polypeptide, or may be designed to improve the targeting capacity of the polypeptide for desired cells, tissues or organs.
  • the invention further embraces FGFBP3 or variants thereof that have been covalently modified to include one or more water- soluble polymer attachments such as polyethylene glycol, polyoxyethylene glycol or polypropylene glycol.
  • compositions in which the FGFBP3 or variants thereof is linked to a polymer are included within the scope of the present invention.
  • the polymer may be water soluble to prevent precipitation of the protein in an aqueous environment, such as a physiological environment.
  • Suitable water-soluble polymers may be selected from the group consisting of, for example, polyethylene glycol (PEG), monomethoxypolyethylene glycol, dextran, cellulose, or other carbohydrate based polymers, poly-(N- vinyl pyrrolidone) polyethylene glycol, polypropylene glycol homopolymers, a polypropylene oxide/ethylene oxide copolymer polyoxyethylated polyols (e.g., glycerol) and polyvinyl alcohol.
  • the selected polymer is usually modified to have a single reactive group, such as an active ester for acylation or an aldehyde for alkylation, so that the degree of polymerization may be controlled.
  • Polymers may be of any molecular weight, and may be branched or unbranched, and mixtures of such polymers may also be used. When the chemically modified NgR polymer is destined for therapeutic use, pharmaceutically acceptable polymers will be selected for use.
  • Pegylation of FGFBP3 or variants thereof may be carried out by any of the pegylation reactions known in the art.
  • the pegylation is carried out via an acylation reaction or an alkylation reaction with a reactive polyethylene glycol molecule (or an analogous reactive water-soluble polymer).
  • a preferred water-soluble polymer for pegylation of polypeptides is polyethylene glycol (PEG), including, but not limited to bi-functional PEGs.
  • PEG polyethylene glycol
  • polyethylene glycol is meant to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (CI-CIO) alkoxy- or aryloxy-polyethylene glycol.
  • FGFBP3 or variants thereof may be performed under any suitable conditions used to react a biologically active substance with an activated polymer molecule.
  • Methods for preparing pegylated FGFBP3 or variants thereof will generally comprise the steps of (a) reacting the polypeptide with polyethylene glycol, such as a reactive ester or aldehyde derivative of PEG, under conditions whereby FGFBP3 or variants thereof becomes attached to one or more PEG groups, and (b) obtaining the reaction products. It will be apparent to one of ordinary skill in the art to select the optimal reaction conditions or the acylation reactions based on known parameters and the desired result.
  • Pegylated and other polymer-modified FGFBP3 or variants thereof may generally be used in the methods of the current invention.
  • the chemically-derivatized polymer-modified FGFBP3 or variants thereof disclosed herein may have additional activities, enhanced or reduced biological activity, or other characteristics, such as increased or decreased half-life, as compared to the nonderivatized molecules.
  • the modified FGFBP3 or variants thereof may be employed alone, together, or in combination with other pharmaceutical compositions.
  • cytokines, growth factors, antibiotics, antiinflammatories and/or chemotherapeutic agents may be co-administered as is appropriate for the indication being treated.
  • compositions comprising purified polypeptides of the invention.
  • Preferred compositions comprise, in addition to the complex of FGF19 and FGFBP3 or variants thereof of the invention, a pharmaceutically acceptable, i.e., sterile and non-toxic, liquid, semisolid, or solid diluent that serves as a pharmaceutical vehicle, excipient or medium. Any diluent known in the art may be used.
  • Exemplary diluents include, but are not limited to, water, saline solutions, polyoxyethylene sorbitan monolaurate, magnesium stearate, methyl- and propylhydroxybenzoate, talc, alginates, starches, lactose, sucrose, dextrose, sorbitol, mannitol, glycerol, calcium phosphate, mineral oil and cocoa butter.
  • the invention provides fusion proteins comprising at least a first and a second fusion peptide.
  • the fusion partners are, generally speaking, covalently bonded to one another via a typical amine bond between the fusion peptides, thus creating one contiguous amino acid chain.
  • Types of fusion proteins provided by the present invention include but are not limited to, fusions with secretion signals and other heterologous functional regions.
  • a region of additional amino acids, particularly charged amino acids may be added to the N-terminus of the FGFBP3 or variant thereof to improve stability and persistence in the host cell, during purification or during subsequent handling and storage.
  • Additional fusion proteins include fusions for enhancing translocation of the protein across cell membranes.
  • Tat is an 86-amino acid protein involved in the replication of human immunodeficiency virus type 1 (HIV-1).
  • HIV-1 Tat transactivation protein is efficiently taken up by cells, and it has been demonstrated that low concentrations (nM) are sufficient to transactivate a reporter gene expressed from the HIV-1 promoter.
  • Exogenous Tat protein is able to translocate through the plasma membrane and reach the nucleus to transactivate the viral genome. Tat peptide-mediated cellular uptake and nuclear translocation have been demonstrated in several systems.
  • Tat-derived peptide (residues 37-72 of Tat) to several proteins results in their internalization in several cell lines or tissues (Fawell (1994) Proc. Natl. Acad. Sci. USA 91, 664-668.
  • Tat-NLS-f-Gal a fusion protein consisting of Tat amino acids 48-59 fused by their amino-terminus to ⁇ -galactosidase amino acids 9- 1023 translocates to the cell nucleus in an ATP-dependent, cytosolic factor-independent manner.
  • the fusion proteins of the present invention may comprise all or a portion of HIV-Tat, such as any sequential residues of the Tat protein basic peptide motif 37-72 (37-
  • the minimum number of amino acid residues can be in the range of from about three to about six.
  • the Tat portion of the fusion protein is from about three to about five contiguous amino acids in length.
  • the Tat portion of the fusion protein is about four amino acids in length, i.e., the minimal requirement for one alpha helical turn.
  • the Tat portion of the fusion protein comprises Tat protein residues 48-57 (GRKKRRQRRR) (SEQ ID NO: 5).
  • a region may be added to facilitate purification.
  • "histidine tags" (“his tags”) or "lysine tags” may be appended to the first fusion peptide.
  • histidine tags include, but are not limited to hexaH, heptaH and hexaHN.
  • lysine tags include, but are not limited to pentaL, heptaL and FLAG.
  • Such regions may be removed prior to final preparation of the FGFBP3 or variant thereof.
  • Other examples of a second fusion peptide include, but are not limited to, glutathione S-transferase (GST) and alkaline phosphatase (AP).
  • peptide moieties to proteins, whether to engender secretion or excretion, to improve stability and to facilitate purification or translocation, among others, is a familiar and routine technique in the art and may include modifying amino acids at the terminus to accommodate the tags.
  • the N-terminus amino acid may be modified to, for example, arginine and/or serine to accommodate a tag.
  • the amino acid residues of the C-terminus may also be modified to accommodate tags.
  • One particularly useful fusion protein comprises a heterologous region from immunoglobulin that can be used solubilize proteins.
  • EP A0464 533 discloses fusion proteins comprising various portions of constant region of immunoglobin molecules together with another human protein or part thereof.
  • the Fc part in a fusion protein is thoroughly advantageous for use in therapy and diagnosis and thereby results, for example, in improved pharmacokinetic properties (EP A0232 262).
  • the fusion proteins of the current invention can be recovered and purified from recombinant cell cultures by well-known methods including, but not limited to, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose
  • HPLC high performance liquid chromatography
  • Well-known techniques for refolding protein may be employed to regenerate active conformation when the fusion protein is denatured during isolation and/or purification.
  • Fusion proteins of the present invention include, but are not limited to, products of chemical synthetic procedures and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect and mammalian cells. Depending upon the host employed in a recombinant production procedure, the fusion proteins of the present invention may be glycosylated or may be non-glycosylated. In addition, fusion proteins of the invention may also include an initial modified methionine residue, in some cases as a result of host- mediated processes.
  • the complex of FGFBP3 or variant thereof and FGF19 can be prepared as a pharmaceutical composition.
  • one or more cofactors may also be added to complex of FGFBP3 or variant thereof and FGF19, to form a composition.
  • Cofactors that may be added include, but are not limited to, heparin, hyaluronic acid, a fibronectin, an elastin, a laminin, albumin, a proteoglycan, collagen, gelatin, a divalent cation, calcium chloride, zinc sulfate, magnesium chloride, sodium bicarbonate, sodium chloride, sodium acetate, or sodium phosphate.
  • a protein or a protein fragment may be added as a cofactor to the complex of FGFBP3 or variant thereof and FGF19.
  • pharmaceutically acceptable carrier or “pharmaceutical carrier” is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration.
  • the nature of the pharmaceutical carrier or other ingredients will depend on the specific route of administration and particular embodiment of the invention to be administered. Examples of techniques and protocols that are useful in this context are, inter alia, found in Remington: The Science and Practice of Pharmacy (2010), Lippincott Williams & Wilkins. Examples of such pharmaceutical carriers or diluents include, but are not limited to, water, saline, Ringer's solution, dextrose solution and 5% human serum albumin.
  • Liposomes and non-aqueous vehicles such as fixed oils may also be used.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.
  • a pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration.
  • routes of administration include oral and parenteral (e.g., intravenous, intradermal, subcutaneous, inhalation, transdermal (topical), transmucosal and rectal administration).
  • Solutions or suspensions used for parenteral, intradermal or subcutaneous application can include, but are not limited to, a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents, antibacterial agents such as benzyl alcohol or methyl parabens, antioxidants such as ascorbic acid or sodium bisulfite, chelating agents such as ethylenediaminetetraacetic acid, buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • the pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide.
  • the parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • suitable pharmaceutical carriers include physiological saline, bacteriostatic water, Cremophor ELTM (BASF) or phosphate buffered saline (PBS).
  • the compositions must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi.
  • the pharmaceutical carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol and liquid polyethylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like.
  • isotonic agents for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions can be prepared by incorporating the active compound/composition in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Oral compositions generally include an inert diluent or an edible pharmaceutical carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches or capsules. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition.
  • the tablets, pills, capsules, troches and the like may contain any of the following ingredients, or compounds of a similar nature, such as but not limited to a binder, such as microcrystalline cellulose, gum tragacanth or gelatin, an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel or corn starch, a lubricant such as magnesium stearate or Sterotes, a glidant such as colloidal silicon dioxide, a sweetening agent such as sucrose or saccharin, or a flavoring agent such as peppermint, methyl salicylate or flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose
  • a disintegrating agent such as alginic acid, Primogel or corn starch
  • a lubricant such as magnesium stearate or Sterotes
  • a glidant such as colloidal silicon dioxide
  • the active complex is prepared with pharmaceutical carriers that will protect the complex against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems.
  • pharmaceutical carriers that will protect the complex against rapid elimination from the body
  • a controlled release formulation including implants and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova
  • Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers.
  • compositions can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811. It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active complex calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved.
  • compositions can be included in a container, pack or dispenser together with instructions for administration.
  • the dosage of the FGFBP3 -FGF19 complex will depend on the disorder or condition to be treated and other clinical factors such as weight and condition of the human or animal and the route of administration of the compound.
  • the complex can be administered at a dose of between about 0.005 mg/kg of body weight to 500 mg/kg of body weight. Therapy is typically administered at lower dosages and is continued until the desired therapeutic outcome is observed.
  • the proper dosage depends on various factors such as the type of disorder being treated, the particular composition being used and the size and physiological condition of the patient.
  • Therapeutically effective doses for the compositions described herein can be estimated initially from cell culture and animal models.
  • a dose can be formulated in animal models to achieve a circulating concentration range that initially takes into account the IC 50 as determined in cell culture assays.
  • the animal model data can be used to more accurately determine useful doses in humans.
  • the invention also relates to methods of altering intracellular signaling of a cell, comprising contacting cells with the complex of FGFBP3 or a variant thereof plus FGF19, wherein the cell possesses a receptor that specifically binds to the complex.
  • the receptor is the fibroblast growth factor receptor 4 (FGF 4).
  • FGF 4 fibroblast growth factor receptor 4
  • the specific binding of the complex to a receptor will, in turn, initiate the intracellular signaling cascade that is normally associated with the complex.
  • Figure 4 demonstrates that administration of the complex of FGFBP3 or a variant thereof and FGF19 results in phosphorylation of Erkl/2.
  • the present invention provides for methods of stimulating phosphorylation of Erkl/2 in a cell comprising contacting the cell(s) with a complex of FGF19 and FGFBP3 or a variant thereof. Additional methods of the present invention comprise assessing the levels of Erkl/2 phosphorylation, both before and after contacting the cell(s) with the complexes of the present invention and determining the increase or decrease of Erkl/2 phosphorylation in response to the complexes of the present invention.
  • one embodiment of the present invention comprises methods of stimulating phosphorylation of p90RSK, rpS6 and/or elF4B in cells. These methods of phosphorylating p90RSK, rpS6 and/or elF4B comprise contacting the cells with a complex of FGF19 and FGFBP3 or a variant thereof in an amount sufficient to stimulate phosphorylation thereof.
  • One target of a phosphorylated p90RSK is glycogen synthase kinase 3a and 3 ⁇ (GSK3 kinases), which, when phosphorylated, are responsible for inhibition of glycogen synthase (GS).
  • the GSK3 kinases are also inhibited or inactivated when they themselves are phosphorylated.
  • phosphorylated p90RSK inhibits or inactivates the GSK3 kinases which block the inhibition of GS. Once the inhibition of GS is removed, GS is activated and, in turn, can trigger production of glycogen.
  • one embodiment of the present invention is directed to methods of increasing glycogen production in a subject in need thereof, with the methods comprising administering a complex of FGF19 and FGFBP3 or a variant thereof in an amount sufficient to stimulate production of glycogen.
  • Phosphorylated p90 SK also stimulates protein synthesis at least in the liver. Accordingly, one embodiment of the present invention is directed towards increasing protein synthesis in a subject in need thereof, with the methods comprising administering a complex of FGF19 and FGFBP3 or a variant thereof to the subject in an amount sufficient to stimulate protein synthesis.
  • a liver- synthesized protein is albumin.
  • one specific embodiment of the present invention is directed towards increasing production of albumin in a subject in need thereof, with the methods comprising administering a complex of FGF19 and FGFBP3 or a variant thereof to the subject in an amount sufficient to stimulate production of albumin.
  • the present invention provides methods of stimulating promoter activity in a cell or population of cells, where the promoter is responsive to activated Erkl/2 or p90 SK with the methods comprising contacting the cell(s) with a complex of FGFBP3 or a variant thereof and FGF19.
  • the promoter is responsive to activated Erkl/2 or p90 SK with the methods comprising contacting the cell(s) with a complex of FGFBP3 or a variant thereof and FGF19.
  • One of skill in the art would be aware of promoters that respond to activated Erkl/2 or p90RSK.
  • the activity of a complex of a variant of FGFBP3 and FGF19 with respect to stimulating Erkl/2-responsive promoters or p90RSK-responsive promoters may or may not be altered relative to a complex with wild-type FGFBP3 and FGF19.
  • Additional methods of the present invention comprise assessing the activity of an Erkl/2-responsive promoter both before and after contacting the cell(s) with the complexes of the present invention and determining the increase or decrease of the promoter in response to the complexes of the present invention.
  • contacting when used in connection with the methods of the present invention means bringing the complexes of the present invention in proximity to the target cells such that a specific binding event or a biological effect is possible.
  • contacting can include adding the complexes in culture medium and applying the culture medium to cells in culture.
  • contacting would also include administration of the complexes, or pharmaceutical compositions thereof, of the present invention to cells in an intact organism.
  • Compositions for administering the complexes of the present invention have been described herein.
  • administering and “administer” are used to mean introducing at least one complex of the present invention into a subject.
  • the complex or composition When administration is for the purpose of treatment, the complex or composition is provided at, or after the onset of, a symptom or condition in need of treatment.
  • the therapeutic administration of this complex or composition serves to attenuate any symptom, or prevent additional symptoms from arising.
  • administration is for the purposes of preventing a condition from arising (“prophylactic administration"), the complex or composition is provided in advance of any visible or detectable symptom.
  • the prophylactic administration of the complex or comporision serves to attenuate subsequently arising symptoms or prevent symptoms from arising altogether.
  • the route of administration of the complex or composition includes, but is not limited to, topical, transdermal, intranasal, vaginal, rectal, oral, subcutaneous intravenous, intraarterial, intramuscular, intraosseous, intraperitoneal, epidural and intrathecal as previously disclosed herein.
  • the methods would also include coadministering one or more substances in addition to the complexes the present invention.
  • coadminister indicates that each of at least two substances, with one of the substances being a complex of FGFBP3 or a variant thereof and FGF19, is administered during a time frame wherein the respective periods of biological activity or effects of each of the substances overlap.
  • the term includes sequential as well as coextensive administration of the complexes of the present invention with another substance.
  • coadministration of more than one substance can be for therapeutic and/or prophylactic purposes. If more than one substance is coadministered, the routes of administration of the two or more substances need not be the same.
  • the invention also relates to methods of lowering blood glucose levels in a subject, the method comprising administering a complex of FGF19 and FGFBP3 to a subject in need of lowering of blood glucose levels.
  • the subject is screened prior to administration of the complex.
  • the invention also relates to methods of lowering a subject's body weight, the method comprising administering a complex of FGF19 and FGFBP3 to a subject that is in need of lowering its body weight.
  • the subject is screened prior to administration of the complex.
  • mice Five week old female ob/ob mice, obtained from Jackson Laboratories, were randomly assigned to treatment groups with FGF19 alone, or FGF19 in combination with BP3. The substances were administered in the morning by single intraperitoneal (i.p.) injection. To assess potential therapeutic effects on the metabolic profile with FGF19 or FGF19+BP3 treatments, glucose tolerance tests were performed and changes in body weights were measured. Two series of experiments were performed to investigate acute and sustained effects with the different treatments.
  • mice were fasted overnight (14 h) before they were subjected to a standard glucose tolerance test (GTT).
  • GTT is used to evaluate the ability of an organism to metabolize exogenous glucose.
  • the GTT is used to uncover patients with latent diabetes or patients at risk for diabetes, e.g. during pregnancy and is performed after a fasting period by oral administration of a glucose containing drink.
  • oral GTT is performed by gavage of a glucose solution, i.e. orally administration. Intraperitoneal injection of a sterile glucose solution was chosen because this allows for a tighter control of dosing of the glucose.
  • the animals were weighed to determine the amount of glucose to inject.
  • the glucose tolerance test was performed in a quiet room and handling was kept down to a minimum to reduce stress during the procedure.
  • a bolus of glucose (1 g/kg) was injected into the intraperitoneal cavity (30 % D-glucose:H 2 0 solution) and blood was sampled from the tail tip at 0, 15, 30, 60, 120 and 180 minutes after glucose injection. Blood glucose levels were determined with a portable glucose meter (Contour ® , Bayer Healthcare).
  • Ob/ob mice exhibit glucose intolerance and are generally used as a leptin-deficient diabetes model and reflect the human disease well.
  • IPGTT intraperitoneal glucose tolerance test
  • the FGF19+BP3 group displayed an improved AUC compared with baseline even 9 days after completed treatment (p ⁇ 0.01), whereas no such effect was observed in mice receiving FGF19 alone ( Figure 1C).
  • BP3 enhances and prolongs the effects of FGF19 on glucose homeostasis, i.e., there is an improvement of the glucose tolerance, from either single or multiple doses.
  • the combination of FGF19+BP3 also reduced the body weight per mouse after 1 dose/day for 5 days, while the animals in FGF19 treatment group gained weight.
  • the combination of BP3 and FGF19 has an unexpectedly better therapeutic effect on obesity than FGF19 alone.
  • FGFBP3 enhanced the effects of FGF19, i.e., a molecule that that will not be retained by the extracellular matrix and that
  • HepG2 cells hepatocellular carcinoma cells
  • FGF19 hepatocellular carcinoma cells
  • Figure 5 shows that the presence of BP3 enhanced the ability of FGF19 to induce phosphorylation of Erkl/2.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The invention relates to methods of treating a metabolic disorder in a subject, the method comprising administering a complex of fibroblast growth factor 19 (FGF19), fibroblast growth factor 21 (FGF21) or fibroblast growth factor 23 (FGF23), plus fibroblast growth factor binding protein 3 (FGFBP3) to a subject in need of treatment of a metabolic disorder.

Description

COMPOSITIONS AND TREATMENTS OF METABOLIC DISORDERS USING
FGF BINDING PROTEIN 3 AND FGF19
Cross-Reference to Related Applications
[0001] This application claims priority to United States Provisional Application No. 61/782,347, filed 14 March 2013, which is incorporated by reference.
Statement Regarding Federally Sponsored Research or Development
[0002] Part of the work performed during development of this invention utilized U.S. Government funds through National Institutes of Health Grant Nos. Ol CA71508 and POl HL068686. The U.S. Government has certain rights in this invention.
Sequence Listing Information
[0003] A computer readable text file, entitled "036681-5014-WO-SequenceListing.txt," created on or about 13 March 2014 with a file size of about 6 kb contains the sequence listing for this application and is hereby incorporated by reference in its entirety.
Background of the Invention
Field of the Invention
[0004] The invention relates to methods of treating a metabolic disorder in a subject, the method comprising administering a complex of fibroblast growth factor 19 (FGF19), fibroblast growth factor 21 (FGF21) or fibroblast growth factor 23 (FGF23), plus fibroblast growth factor binding protein 3 (FGFBP3) to a subject in need of treatment of a metabolic disorder.
Background of the Invention
[0005] Fibroblast growth factor 19 (FGF19) and other members of the FGF19 family are involved in the regulation of metabolism. FGF19 has also been recently described as a sensitizer to insulin. In addition, there some members of the FGF19 family interact with the co-receptor klotho to affect metabolism and in response to bile acid production.
[0006] Previous studies indicate that FGF-binding proteins (FGFBP) can enhance the effects of FGF by mobilizing FGFs from their storage depots found in extracellular glycosaminoglycans or heparansulfate proteoglycans. In contrast to most other FGFs, members of the FGF19 family of proteins, i.e., FGF19, FGF21 and FGF23, show only very little binding to glycosaminoglycans or heparansulfates in the extracellular matrix. The most recently discovered member of the FGFBP3 family also appears to mobilize FGFs from such storage depots and bind to FGFs including FGF19 family members.
Summary of the Invention
[0007] The invention relates to methods of treating a metabolic disorder in a subject, the method comprising administering a complex of fibroblast growth factor 19 (FGF19), fibroblast growth factor 21 (FGF21) or fibroblast growth factor 23 (FGF23), plus fibroblast growth factor binding protein 3 (FGFBP3) to a subject in need of treatment of a metabolic disorder.
[0008] The invention also relates to methods of lowering blood glucose levels in a subject, the method comprising administering a complex of FGF19, FGF21 or FGF23 and FGFBP3 to a subject in need of lowering of blood glucose levels.
[0009] The invention also relates to methods of lowering a subject's body weight, the method comprising administering a complex of FGF19, FGF21 or FGF23 and FGFBP3 to a subject that is in need of lowering its body weight.
Brief Description of the Drawings
[0010] FIGURE 1 depicts the effects of multiple FGF19 or FGF19+BP3 treatments on glucose blood levels in ob/ob mice after a bolus injection of glucose (=glucose tolerance test). A, Five treatments with FGF19 reduced blood glucose levels at 180 minutes after the beginning of the glucose tolerance test, and the curve returned to the baseline 9 days after receiving no treatment. Filled Squares: Baseline; Open Squares: FGF19 (one dose per day for 5 days); Open Circles: FGF19 (one dose per day for 5 days + 9 days of no treatment). B, Five treatments with FGF19+BP3 improved the glucose tolerance at 60, 120, and 180 minutes with a sustained effect for 9 days. Filled Squares: Baseline; Open Squares: FGF19+BP3 (one dose per day for 5 days); Open Circles: FGF 19+BP3 (one dose per day for 5 days + 9 days of no treatment). C, The area under the curve of the glucose tolerance test (AUC) was improved after 5 treatments in both groups. After another 9 days without any treatment, only the FGF19+BP3 group exhibited an improved glucose tolerance test. Solid Bars: Baseline, Open Bars: FGF19 or FGF19+BP3 (one dose per day for 5 days); Hatched Bars, FGF19 or FGF19+BP3 (one dose per day for 5 days + 9 days of no treatment). * denotes P<0.01. [0011] FIGURE 2 depicts the effects of a single dose of FGF19 or FGF19+BP3 treatment on glucose blood levels in ob/ob mice after a bolus injection of glucose (=glucose tolerance test). A, The glucose levels were not changed 2 days after a single dose of FGF19 alone. Filled Squares: Baseline, Open Squares: single dose treatment. B, In the FGF19+BP3 group, the glucose levels were significantly reduced at 15, 30 and 60 minutes post-injection of glucose. C, The area under the curve of the glucose tolerance test (AUC) was reduced 2 days after a single treatment in FGF19+BP3 group. Filled Squares: Baseline, Open Squares: Single treatment. * denotes P<0.05, ** P<0.01.
[0012] FIGURE 3 depicts the effects of acute single doses of BP3, FGF19, and the complex of BP3+FGF19 in ob/ob mice in response to a glucose tolerance test. Figures 3A, 3B show the effect when the test was conducted two hours after treatment. Figure 3C shows the effect when the test was performed 24 hours after treatment. Even 7 days of dosing of BP3 alone with a single daily dose of BP3 lacked an effect on the baseline blood glucose or on the IPGTT test (Figure 3D).
[0013] FIGURE 4 depicts the body weight changes in ob/ob mice following single or multiple treatments with FGF19 or FGF19+BP3. A, The changes in body weight were significantly different between the FGF19 group and the combination group immediately after 5 treatments (2.2 ± 0.8g vs. -1.0 ± 0.7g), and a clear trend was also observed 2 days after a single treatment (p=0.055). Filled Squares: single treatment, Open Squares: 5 treatments at one dose/day. B, The percentage changes in body weight is also different between the two groups immediately after 5 daily treatments (6.04 ± 2.33% vs. -3.40 ± 2.50%). Filled Squares: single treatment; Open Squares: 5 treatments (one dose/day). * denotes P<0.05.
[0014] FIGURE 5 depicts the presence of BP3 enhancing the ability of FGF19 to induce phosphorylation of Erkl/2 in HepG2 cells. HepG2 cells were treated with FGF19 ± BP3 or the negative control protein MBP. pERKl/2 was measured in cell lysates at different times after treatment.
Detailed Description of the Invention
[0015] The invention relates to methods of treating a metabolic disorder in a subject, the methods comprising administering a complex of fibroblast growth factor 19 (FGF19), fibroblast growth factor 21 (FGF21) or fibroblast growth factor 23 (FGF23), plus fibroblast growth factor binding protein 3 (FGFBP3) or variant thereof to a subject in need of treatment of a metabolic disorder. As used herein, the term "subject" is used interchangeably with the term "human" or "patient," and is also used to mean an animal, in particular a mammal, and even more particularly a non-human or human primate.
[0016] Fibroblast growth factor 19 (FGF19) is a well-known member of the FGF family of proteins that is involved in nutrient metabolism. FGF19 is a protein of 216 amino acids, with the signal peptide being amino acids 1-24. As used herein, "fibroblast growth factor 19" or "FGF19" can mean the full length FGF19 with or without the N-terminus signal sequence. The mouse ortholog to human FGF19 is known as FGF15 and is 218 amino acids in length, including the 25-amino acid signal sequence at the N- terminus. As used herein, "fibroblast growth factor 15" or "FGF15" is used to indicate any ortholog to FGF19 and can include the full length amino acid sequence, with or without the N-terminus signal sequence. It is understood that a reference to "FGF19" herein will also include a reference to its art- accepted orthologs, such as mouse FGF15. In general the concentration of FGF19 (or mFGF15) is upregulated after feeding and binds to FGF Receptor 4 (FGFR4). Specifically, FGF19 is synthesized in the distal small intestine in response to uptake of bile acids via the nuclear bile receptor and controlled by food intake. FGF21 expression in the liver and fat tissues is also regulated by the feeding or starving status and function in a temporal cascade with insulin, glucagon and other hormones to regulate responses to nutrition (Potthoff, et al., Genes and Development 2012).
[0017] The fibroblast growth factor binding protein 3 (FGFBP3) is a secreted protein that binds to FGF19 and is believed to act as a co-receptor with FGFR4. FGFBP3 is not known to bind to other FGF receptors to any significant extent. The full length amino acid sequence of human FGFBP3 is shown below as SEQ ID NO:l. The full length amino acid sequence of human FGFBP3, without the 26 amino- acid signal sequence, is shown below as SEQ ID NO:2. The C-terminus of FGFBP3 is shown below as SEQ ID NO:3.
[0018] As used herein, "FGFBP3" means a peptide that comprises the amino acid sequence of SEQ ID NO:3 or a variant thereof that still retains activity similar to the wild-type FGFBP3. Thus, the amino acid sequence of SEQ ID NOs:l and 2 are just two embodiments of the term FGFBP3 as it is used herein. "Variants" of FGFBP3 are discussed below.
MTPPKLRASL SPSLLLLLSG CLLAAARREK GAASNVAEPV PGPTGGSSGR FLSPEQHACS 60 WQLLLPAPEA AAGSELALRC QSPDGARHQC AYRGHPERCA AYAARRAHFW KQVLGGLRKK 120 RRPCHDPAPL QARLCAGKKG HGAELRLVPR ASPPARPTVA GFAGESKPRA RNRGRTRERA 180 SGPAAGTPPP QSAPPKENPS ERKTNEGKRK AALVPNEERP MGTGPDPDGL DGNAELTETY 240 CAEKWHSLCN FFVNFWNG (SEQ ID NO : l ) 258
RREK GAASNVAEPV PGPTGGSSGR FLSPEQHACS WQLLLPAPEA AAGSELALRC QSPDGARHQC 64
AYRGHPERCA AYAARRAHFW KQVLGGLRKK RRPCHDPAPL QARLCAGKKG HGAELRLVPR 124
ASPPARPTVA GFAGESKPRA RNRGRTRERA SGPAAGTPPP QSAPPKENPS ERKTNEGKRK 184
AALVPNEERP MGTGPDPDGL DGNAELTETY CAEKWHSLCN FFVNFWNG (SEQ ID NO: 2 ) 232
LDGNAELTET YCAEKWHSLC NFFVNFWNG (SEQ ID NO: 3 ) 29
[0019] The present invention is directed to methods that include administration of a complex of FGF19 and FGFBP3. As used herein, the term "complex" as it relates to FGF19 and FGFBP3 means the presence of both FGFBP3 and FGF19. The FGF19 and FGFBP3 can, but need not, be specifically interacting, i.e., specifically binding to one another. In one embodiment, the FGF19 and FGFBP3 within the complex are specifically binding to one another. In another embodiment, the FGF19 and FGFBP3 within the complex are not necessarily specifically binding to one another.
[0020] Accordingly, in some embodiment of the methods of the present invention, full length FGFBP3 (a peptide amino acid sequence of SEQ ID NO:l) is complexed with FGF19. In select of these embodiments, the FGFBP3 in the complex comprises a peptide with an amino acid sequence that is 100% identical to the amino acid sequence of SEQ ID NO:l. In additional embodiments, the FGFBP3 in the complex consists of a peptide with an amino acid sequence that is 100% identical to the amino acid sequence of SEQ ID NO:l.
[0021] In additional embodiments of the methods of the present invention, full length FGFBP3 without the signal sequence (a peptide amino acid sequence of SEQ ID NO:2) is complexed with FGF19. In select of these embodiments, the FGFBP3 in the complex comprises a peptide with an amino acid sequence that is 100% identical to the amino acid sequence of SEQ ID NO:2. In additional embodiments, the FGFBP3 in the complex consists of a peptide with an amino acid sequence that is 100% identical to the amino acid sequence of SEQ ID NO:2.
[0022] In still additional embodiment of the methods of the present invention, the C-terminal FGFBP3 (a peptide amino acid sequence of SEQ ID NO:3) is complexed with FGF19. In select of these embodiments, the FGFBP3 in the complex comprises a peptide with an amino acid sequence that is 100% identical to the amino acid sequence of SEQ ID NO:3. In additional embodiments, the FGFBP3 in the complex consists of a peptide with an amino acid sequence that is 100% identical to the amino acid sequence of SEQ ID NO:3.
[0023] The terms "peptide," "polypeptide" and "protein" are used interchangeably herein. As used herein, an "isolated polypeptide" is intended to mean a polypeptide that has been completely or partially removed from its native environment. For example, polypeptides that have been removed or purified from cells are considered isolated. In addition, recombinantly produced polypeptides molecules contained in host cells are considered isolated for the purposes of the present invention. Moreover, a peptide that is found in a cell, tissue or matrix in which it is not normally expressed or found is also considered as "isolated" for the purposes of the present invention. Similarly, polypeptides that have been synthesized are considered to be isolated polypeptides. "Purified," on the other hand is well understood in the art and generally means that the peptides are substantially free of cellular material, cellular components, chemical precursors or other chemicals beyond, perhaps, buffer or solvent.
"Substantially free" is not intended to mean that other components beyond the novel peptides are undetectable.
[0024] The invention also relates to the use of variants of FGFBP3 that still retain their ability to specifically interact, at least partially, with FGF19. In one embodiment, FGFBP3 variants comprise an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequences of SEQ ID NO: 3. In another embodiment, the FGFBP3 variant consists of a peptide with an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequences of SEQ ID NO: 3.
[0025] In one embodiment, FGFBP3 variants comprise an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequences of SEQ ID NO: 1. In another embodiment, the FGFBP3 variant consists of a peptide with an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequences of SEQ ID NO: 1.
[0026] In one embodiment, FGFBP3 variants comprise an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequences of SEQ ID NO: 2. In another embodiment, the FGFBP3 variant consists of a peptide with an amino acid sequence that is at least 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98% or 99% identical to the amino acid sequences of SEQ ID NO: 2.
[0027] As used herein, a metabolic disorder can be any disorder associated with metabolism, and examples include but are not limited to, obesity, central obesity, insulin resistance, glucose intolerance, abnormal glycogen metabolism, type 2 diabetes, hyperlipidemia, hypoalbuminemia,
hypertriglyceridemia, metabolic syndrome, syndrome X, a fatty liver, fatty liver disease, polycystic ovarian syndrome, and acanthosis nigricans. In one embodiment, the methods are directed towards treating at least one component of postprandial metabolism, such as, but not limited to hepatic glycogen synthesis, protein synthesis and clearance of plasma glucose.
[0028] The terms "trait" and "phenotype" are used interchangeably herein and refer to any visible, detectable or otherwise measurable property of an organism such as symptoms of or susceptibility to a disorder. Typically the terms "trait" or "phenotype" are used herein to refer to symptoms of a metabolic disorder, or a susceptibility to an metabolic disorder. Examples of traits of metabolic disorders include but are not limited to high total cholesterol, low high-density lipoprotein (HDL) cholesterol, impaired fasting glucose levels, insulin resistance, hyperproinsulinemia, central obesity, elevated triglyceride levels, postprandial glucose levels, elevated uric acid levels, thyroid dysfunction, increased body-mass index (BMI), hypertension, impaired glucose tolerance, alterations in hormone and peptide levels (e.g., leptin, ghrelin, obstatin, adiponectin, perilipin, omentin), interactions with substances involved in insulin signaling, lipid, amino acid and glucose metabolism, life expectancy, increased systemic inflammatory state (e.g., as reflected in levels of C-reactive protein, interleukin-6, and TNF-alpha), depression, and sleep disordered breathing.
[0029] In additional embodiments, the peptide variants described herein are functional and capable of altering a subject's response in a glucose tolerance test when complexed with FGF19. In some embodiments, the FGFBP3 variants of the present invention, when complexed with FGF19, have enhanced ability to alter a subject's response in a glucose tolerance test compared to FGF19 complexed with the wild-type FGFBP3. In some embodiments, the FGFBP3 variants of the present invention also have enhanced stability compared to the wild-type FGFBP3 regardless of their association with FGF19.
[0030] A polypeptide having an amino acid sequence at least, for example, about 95% "identical" to a reference an amino acid sequence, e.g., SEQ ID NO: 1, is understood to mean that the amino acid sequence of the polypeptide is identical to the reference sequence except that the amino acid sequence may include up to about five modifications per each 100 amino acids of the reference amino acid sequence. In other words, to obtain a peptide having an amino acid sequence at least about 95% identical to a reference amino acid sequence, up to about 5% of the amino acid residues of the reference sequence may be deleted or substituted with another amino acid or a number of amino acids up to about 5% of the total amino acids in the reference sequence may be inserted into the reference sequence. These modifications of the reference sequence may occur at the N- terminus or C-terminus positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among amino acids in the reference sequence or in one or more contiguous groups within the reference sequence.
[0031] As used herein, "identity" is a measure of the identity of nucleotide sequences or amino acid sequences compared to a reference nucleotide or amino acid sequence. In general, the sequences are aligned so that the highest order match is obtained. "Identity" per se has an art-recognized meaning and can be calculated using well known techniques. While there are several methods to measure identity between two polynucleotide or polypeptide sequences, the term "identity" is well known to skilled artisans (Carillo (1988) J. Applied Math. 48, 1073). Examples of computer program methods to determine identity and similarity between two sequences include, but are not limited to, GCG program package (Devereux (1984) Nucleic Acids Research 12, 387), BLASTP, ExPASy, BLASTN, FASTA (Atschul (1990) J. Mol. Biol. 215, 403) and FASTDB. Examples of methods to determine identity and similarity are discussed in Michaels (2011) Current Protocols in Protein Science, Vol. 1, John Wiley & Sons.
[0032] In one embodiment of the present invention, the algorithm used to determine identity between two or more polypeptides is BLASTP. In another embodiment of the present invention, the algorithm used to determine identity between two or more polypeptides is FASTDB, which is based upon the algorithm of Brutlag (1990) Comp. App. Biosci. 6, 237-245). In a FASTDB sequence alignment, the query and reference sequences are amino sequences. The result of sequence alignment is in percent identity. In one embodiment, parameters that may be used in a FASTDB alignment of amino acid sequences to calculate percent identity include, but are not limited to: Matrix=PAM, k-tuple=2, Mismatch Penalty=l, Joining Penalty=20, Randomization Group Length=0, Cutoff Score=l, Gap
Penalty=5, Gap Size Penalty 0.05, Window Size=500 or the length of the subject amino sequence, whichever is shorter. [0033] If the reference sequence is shorter or longer than the query sequence because of N-terminus or C-terminus additions or deletions, but not because of internal additions or deletions, a manual correction can be made, because the FASTDB program does not account for N-terminus and C-terminus truncations or additions of the reference sequence when calculating percent identity. For query sequences truncated at the N- or C- termini, relative to the reference sequence, the percent identity is corrected by calculating the number of residues of the query sequence that are N-and C- terminus to the reference sequence that are not matched/aligned, as a percent of the total bases of the query sequence. The results of the FASTDB sequence alignment determine matching/alignment. The alignment percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score. This corrected score can be used for the purposes of determining how alignments "correspond" to each other, as well as percentage identity. Residues of the reference sequence that extend past the N- or C-termini of the query sequence may be considered for the purposes of manually adjusting the percent identity score. That is, residues that are not matched/aligned with the N- or C-termini of the comparison sequence may be counted when manually adjusting the percent identity score or alignment numbering.
[0034] For example, a 90 amino acid residue query sequence is aligned with a 100 residue reference sequence to determine percent identity. The deletion occurs at the N-terminus of the query sequence and therefore, the FASTDB alignment does not show a match/alignment of the first 10 residues at the N- terminus. The 10 unpaired residues represent 10% of the reference sequence (number of residues at the N- and C-termini not matched/total number of residues in the reference sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 residues were perfectly matched (100% alignment) the final percent identity would be 90% (100% alignment - 10% unmatched overhang). In another example, a 90 residue query sequence is compared with a 100 reference sequence, except that the deletions are internal deletions. In this case the percent identity calculated by FASTDB is not manually corrected, since there are no residues at the N- or C- termini of the subject sequence that are not matched/aligned with the query. In still another example, a 110 amino acid query sequence is aligned with a 100 residue reference sequence to determine percent identity. The addition in the query occurs at the N-terminus of the query sequence and therefore, the FASTDB alignment may not show a match/alignment of the first 10 residues at the N-terminus. If the remaining 100 amino acid residues of the query sequence have 95% identity to the entire length of the reference sequence, the N-terminal addition of the query would be ignored and the percent identity of the query to the reference sequence would be 95%.
[0035] As used herein, the terms "correspond(s) to" and "corresponding to," as they relate to sequence alignment, are intended to mean enumerated positions within the reference protein, e.g., wild-type FGFBP3, and those positions in the variant or ortholog FGFBP3 that align with the positions with the reference protein. Thus, when the amino acid sequence of a subject FGFBP3 is aligned with the amino acid sequence of a reference FGFBP3, e.g., SEQ ID NO: 2, the amino acids in the subject sequence that "correspond to" certain enumerated positions of the reference sequence are those that align with these positions of the reference sequence, e.g., SEQ ID NO: 2, but are not necessarily in these exact numerical positions of the reference sequence. Methods for aligning sequences for determining corresponding amino acids between sequences are described herein.
[0036] The invention further embraces other species, preferably mammalian, homologs with amino acid sequences that correspond to FGFBP3. Species homologs, sometimes referred to as "orthologs," in general, share at least 35%, 40%, 45%, 50%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% sequence identity with the human version of the full length binding proteins or the full length binding proteins without the signal sequence. Such corresponding sequences account for FGFBP3 from across a variety of species, such as canine, feline, mouse, rat, rabbit, monkey, etc.
[0037] FGFBP3 with an additional methionine residue at position -1 (Met_1-peptide) are contemplated, as are variants with additional methionine and lysine residues at positions -2 and -1 (Met^-Lys 1- peptide). Variants of FGFBP3 with additional Met, Met-Lys, or Lys residues (or one or more basic residues in general) are particularly useful for enhanced recombinant protein production in bacterial host cells.
[0038] Variants resulting from insertion of the polynucleotide encoding FGFBP3 into an expression vector system are also contemplated. For example, variants (usually insertions) may arise from when the amino terminus and/or the carboxy terminus of FGFBP3 is/are fused to another polypeptide.
[0039] In another aspect, the invention provides deletion variants wherein one or more amino acid residues in FGFBP3 are removed. Deletions can be effected at one or both termini of the FGFBP3 peptide, or with removal of one or more non-terminal amino acid residues of the FGFBP3. Deletion variants, therefore, include all fragments of the FGFBP3 peptide. [0040] Within the confines of the disclosed percent identity, the invention also relates to substitution variants of disclosed polypeptides of the invention. Substitution variants include those polypeptides wherein one or more amino acid residues of FGFBP3 are removed and replaced with alternative residues. For example two variants of SEQ ID NOs: 1 or 2 are known to exist and the invention contemplates the use of these known variants in the methods described herein. Specifically, a variant of FGFBP3 wherein Alanine at position 107 of SEQ ID NO:l is replaced with Threonine (A107T) is included in the methods of the present invention. Another variant of FGFBP3 wherein Glutamate at position 206 of SEQ ID NO:l is replaced with Valine (E206V) is included in the methods of the present invention. Of course, positions 107 and 206 of SEQ ID NO:l correspond to positions 81 and 180 of SEQ ID NO:2. In one aspect, the substitutions are conservative in nature; however, the invention embraces substitutions that are also non-conservative. Conservative substitutions for this purpose may be defined as set out in the tables below. Amino acids can be classified according to physical properties and contribution to secondary and tertiary protein structure. A conservative substitution is recognized in the art as a substitution of one amino acid for another amino acid that has similar properties. Exemplary conservative substitutions are set out in below.
Table I: Conservative Substitutions
Side Chain Characteristic Amino Acid
Aliphatic
Non-polar Gly, Ala, Pro, Iso, Leu, Val
Polar-uncharged Cys, Ser, Thr, Met, Asn, Gin
Polar-charged Asp, Glu, Lys, Arg
Aromatic His, Phe, Trp, Tyr
Other Asn, Gin, Asp, Glu
[0041] Alternatively, conservative amino acids can be grouped as described in Lehninger (1975) Biochemistry, Second Edition; Worth Publishers, pp. 71-77, as set forth below.
Table II: Conservative Substitutions
Side Chain Characteristic Amino Acid
Non-polar (hydrophobic)
Aliphatic: Ala, Leu, Iso, Val, Pro
Aromatic: Phe, Trp
Sulfur-containing: Met
Borderline: Gly
Uncharged-polar
Hydroxyl: Ser, Thr, Tyr
Amides: Asn, Gin
Sulfhydryl: Cys
Borderline: Gly
Positively Charged (Basic): Lys, Arg, His
Negatively Charged (Acidic) Asp, Glu
[0042] And still other alternative, exemplary conservative substitutions are set out below. Table III: Conservative Substitutions
Original Residue Exemplary Substitution
Ala (A) Val, Leu, lie
Arg (R) Lys, Gin, Asn
Asn (N) Gin, His, Lys, Arg
Asp (D) Glu
Cys (C) Ser
Gin (Q) Asn
Glu (E) Asp
His (H) Asn, Gin, Lys, Arg
He CO Leu, Val, Met, Ala, Phe
Leu (L) lie, Val, Met, Ala, Phe
Lys (K) Arg, Gin, Asn
Met (M) Leu, Phe, lie
Phe (F) Leu, Val, lie, Ala
Pro (P) Gly
Ser (S) Thr
Thr (T) Ser
Trp (W) Tyr
Tyr (Y) Trp, Phe, Thr, Ser
Val (V) lie, Leu, Met, Phe, Ala
[0043] It should be understood that the definition of peptides or polypeptides of the invention is intended to include polypeptides bearing modifications other than insertion, deletion, or substitution of amino acid residues. By way of example, the modifications may be covalent in nature, and include for example, chemical bonding with polymers, lipids, other organic and inorganic moieties. Such derivatives may be prepared to increase circulating half-life of a polypeptide, or may be designed to improve the targeting capacity of the polypeptide for desired cells, tissues or organs. Similarly, the invention further embraces FGFBP3 or variants thereof that have been covalently modified to include one or more water- soluble polymer attachments such as polyethylene glycol, polyoxyethylene glycol or polypropylene glycol.
[0044] Compositions in which the FGFBP3 or variants thereof is linked to a polymer are included within the scope of the present invention. The polymer may be water soluble to prevent precipitation of the protein in an aqueous environment, such as a physiological environment. Suitable water-soluble polymers may be selected from the group consisting of, for example, polyethylene glycol (PEG), monomethoxypolyethylene glycol, dextran, cellulose, or other carbohydrate based polymers, poly-(N- vinyl pyrrolidone) polyethylene glycol, polypropylene glycol homopolymers, a polypropylene oxide/ethylene oxide copolymer polyoxyethylated polyols (e.g., glycerol) and polyvinyl alcohol. The selected polymer is usually modified to have a single reactive group, such as an active ester for acylation or an aldehyde for alkylation, so that the degree of polymerization may be controlled. Polymers may be of any molecular weight, and may be branched or unbranched, and mixtures of such polymers may also be used. When the chemically modified NgR polymer is destined for therapeutic use, pharmaceutically acceptable polymers will be selected for use.
[0045] Pegylation of FGFBP3 or variants thereof may be carried out by any of the pegylation reactions known in the art. In one method, the pegylation is carried out via an acylation reaction or an alkylation reaction with a reactive polyethylene glycol molecule (or an analogous reactive water-soluble polymer). A preferred water-soluble polymer for pegylation of polypeptides is polyethylene glycol (PEG), including, but not limited to bi-functional PEGs. As used herein, "polyethylene glycol" is meant to encompass any of the forms of PEG that have been used to derivatize other proteins, such as mono (CI-CIO) alkoxy- or aryloxy-polyethylene glycol.
[0046] Chemical derivatization of FGFBP3 or variants thereof may be performed under any suitable conditions used to react a biologically active substance with an activated polymer molecule. Methods for preparing pegylated FGFBP3 or variants thereof will generally comprise the steps of (a) reacting the polypeptide with polyethylene glycol, such as a reactive ester or aldehyde derivative of PEG, under conditions whereby FGFBP3 or variants thereof becomes attached to one or more PEG groups, and (b) obtaining the reaction products. It will be apparent to one of ordinary skill in the art to select the optimal reaction conditions or the acylation reactions based on known parameters and the desired result.
[0047] Pegylated and other polymer-modified FGFBP3 or variants thereof may generally be used in the methods of the current invention. The chemically-derivatized polymer-modified FGFBP3 or variants thereof disclosed herein may have additional activities, enhanced or reduced biological activity, or other characteristics, such as increased or decreased half-life, as compared to the nonderivatized molecules. The modified FGFBP3 or variants thereof may be employed alone, together, or in combination with other pharmaceutical compositions. For example, cytokines, growth factors, antibiotics, antiinflammatories and/or chemotherapeutic agents may be co-administered as is appropriate for the indication being treated.
[0048] The present invention provides compositions comprising purified polypeptides of the invention. Preferred compositions comprise, in addition to the complex of FGF19 and FGFBP3 or variants thereof of the invention, a pharmaceutically acceptable, i.e., sterile and non-toxic, liquid, semisolid, or solid diluent that serves as a pharmaceutical vehicle, excipient or medium. Any diluent known in the art may be used. Exemplary diluents include, but are not limited to, water, saline solutions, polyoxyethylene sorbitan monolaurate, magnesium stearate, methyl- and propylhydroxybenzoate, talc, alginates, starches, lactose, sucrose, dextrose, sorbitol, mannitol, glycerol, calcium phosphate, mineral oil and cocoa butter.
[0049] In one embodiment, the invention provides fusion proteins comprising at least a first and a second fusion peptide. The fusion partners are, generally speaking, covalently bonded to one another via a typical amine bond between the fusion peptides, thus creating one contiguous amino acid chain. Types of fusion proteins provided by the present invention include but are not limited to, fusions with secretion signals and other heterologous functional regions. Thus, for instance, a region of additional amino acids, particularly charged amino acids, may be added to the N-terminus of the FGFBP3 or variant thereof to improve stability and persistence in the host cell, during purification or during subsequent handling and storage.
[0050] Additional fusion proteins include fusions for enhancing translocation of the protein across cell membranes. For example, Tat is an 86-amino acid protein involved in the replication of human immunodeficiency virus type 1 (HIV-1). The HIV-1 Tat transactivation protein is efficiently taken up by cells, and it has been demonstrated that low concentrations (nM) are sufficient to transactivate a reporter gene expressed from the HIV-1 promoter. Exogenous Tat protein is able to translocate through the plasma membrane and reach the nucleus to transactivate the viral genome. Tat peptide-mediated cellular uptake and nuclear translocation have been demonstrated in several systems. Chemically coupling a Tat-derived peptide (residues 37-72 of Tat) to several proteins results in their internalization in several cell lines or tissues (Fawell (1994) Proc. Natl. Acad. Sci. USA 91, 664-668.
[0051] It is well-known that a region of the Tat protein centered on a cluster of basic amino acids is responsible for this translocation activity. A synthetic peptide consisting of the Tat basic amino acids 48- 60 with a cysteine residue at the C-terminus coupled to fluorescein maleimide translocates to the cell nucleus as determined by fluorescence microscopy. In addition, a fusion protein (Tat-NLS-f-Gal) consisting of Tat amino acids 48-59 fused by their amino-terminus to β-galactosidase amino acids 9- 1023 translocates to the cell nucleus in an ATP-dependent, cytosolic factor-independent manner.
Accordingly, the fusion proteins of the present invention may comprise all or a portion of HIV-Tat, such as any sequential residues of the Tat protein basic peptide motif 37-72 (37-
CFITKALGISYG KK Q PPQGSQTHQVSLSKQ-72 (SEQ ID NO: 4). The minimum number of amino acid residues can be in the range of from about three to about six. In one embodiment, the Tat portion of the fusion protein is from about three to about five contiguous amino acids in length. In another embodiment, the Tat portion of the fusion protein is about four amino acids in length, i.e., the minimal requirement for one alpha helical turn. In another embodiment, the Tat portion of the fusion protein comprises Tat protein residues 48-57 (GRKKRRQRRR) (SEQ ID NO: 5).
[0052] In additional embodiments of fusion proteins, a region may be added to facilitate purification. For example, "histidine tags" ("his tags") or "lysine tags" may be appended to the first fusion peptide. Examples of histidine tags include, but are not limited to hexaH, heptaH and hexaHN. Examples of lysine tags include, but are not limited to pentaL, heptaL and FLAG. Such regions may be removed prior to final preparation of the FGFBP3 or variant thereof. Other examples of a second fusion peptide include, but are not limited to, glutathione S-transferase (GST) and alkaline phosphatase (AP).
[0053] The addition of peptide moieties to proteins, whether to engender secretion or excretion, to improve stability and to facilitate purification or translocation, among others, is a familiar and routine technique in the art and may include modifying amino acids at the terminus to accommodate the tags. For example in SEQ ID NOs: 1, 2 or 3, the N-terminus amino acid may be modified to, for example, arginine and/or serine to accommodate a tag. Of course, the amino acid residues of the C-terminus may also be modified to accommodate tags. One particularly useful fusion protein comprises a heterologous region from immunoglobulin that can be used solubilize proteins. For example, EP A0464 533 discloses fusion proteins comprising various portions of constant region of immunoglobin molecules together with another human protein or part thereof. In many cases, the Fc part in a fusion protein is thoroughly advantageous for use in therapy and diagnosis and thereby results, for example, in improved pharmacokinetic properties (EP A0232 262). On the other hand, for some uses, it would be desirable to be able to delete the Fc part after the fusion protein has been expressed, detected and purified in the advantageous manner described.
[0054] The fusion proteins of the current invention can be recovered and purified from recombinant cell cultures by well-known methods including, but not limited to, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose
chromatography, hydrophobic interaction chromatography, affinity chromatography, e.g., immobilized metal affinity chromatography (IMAC), hydroxylapatite chromatography and lectin chromatography. High performance liquid chromatography ("HPLC") may also be employed for purification. Well-known techniques for refolding protein may be employed to regenerate active conformation when the fusion protein is denatured during isolation and/or purification.
[0055] Fusion proteins of the present invention include, but are not limited to, products of chemical synthetic procedures and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect and mammalian cells. Depending upon the host employed in a recombinant production procedure, the fusion proteins of the present invention may be glycosylated or may be non-glycosylated. In addition, fusion proteins of the invention may also include an initial modified methionine residue, in some cases as a result of host- mediated processes.
[0056] The complex of FGFBP3 or variant thereof and FGF19, can be prepared as a pharmaceutical composition. For example, one or more cofactors may also be added to complex of FGFBP3 or variant thereof and FGF19, to form a composition. Cofactors that may be added include, but are not limited to, heparin, hyaluronic acid, a fibronectin, an elastin, a laminin, albumin, a proteoglycan, collagen, gelatin, a divalent cation, calcium chloride, zinc sulfate, magnesium chloride, sodium bicarbonate, sodium chloride, sodium acetate, or sodium phosphate. In some embodiments, a protein or a protein fragment may be added as a cofactor to the complex of FGFBP3 or variant thereof and FGF19.
[0057] As used herein, "pharmaceutically acceptable carrier" or "pharmaceutical carrier" is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The nature of the pharmaceutical carrier or other ingredients will depend on the specific route of administration and particular embodiment of the invention to be administered. Examples of techniques and protocols that are useful in this context are, inter alia, found in Remington: The Science and Practice of Pharmacy (2010), Lippincott Williams & Wilkins. Examples of such pharmaceutical carriers or diluents include, but are not limited to, water, saline, Ringer's solution, dextrose solution and 5% human serum albumin. Liposomes and non-aqueous vehicles such as fixed oils may also be used. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.
[0058] A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include oral and parenteral (e.g., intravenous, intradermal, subcutaneous, inhalation, transdermal (topical), transmucosal and rectal administration). Solutions or suspensions used for parenteral, intradermal or subcutaneous application can include, but are not limited to, a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents, antibacterial agents such as benzyl alcohol or methyl parabens, antioxidants such as ascorbic acid or sodium bisulfite, chelating agents such as ethylenediaminetetraacetic acid, buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
[0059] Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. For intravenous administration, suitable pharmaceutical carriers include physiological saline, bacteriostatic water, Cremophor EL™ (BASF) or phosphate buffered saline (PBS). In all cases, the compositions must be sterile and should be fluid to the extent that easy syringeability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The pharmaceutical carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol and liquid polyethylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like. In many cases, it may be desirable to include isotonic agents, for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
[0060] Sterile injectable solutions can be prepared by incorporating the active compound/composition in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, methods of preparation are vacuum drying and freeze-drying that yields a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
[0061] Oral compositions generally include an inert diluent or an edible pharmaceutical carrier. They can be enclosed in gelatin capsules or compressed into tablets. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, troches or capsules. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like may contain any of the following ingredients, or compounds of a similar nature, such as but not limited to a binder, such as microcrystalline cellulose, gum tragacanth or gelatin, an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel or corn starch, a lubricant such as magnesium stearate or Sterotes, a glidant such as colloidal silicon dioxide, a sweetening agent such as sucrose or saccharin, or a flavoring agent such as peppermint, methyl salicylate or flavoring. [0062] In one embodiment, the active complex is prepared with pharmaceutical carriers that will protect the complex against rapid elimination from the body, such as a controlled release formulation, including implants and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for preparation of such formulations will be apparent to those skilled in the art. The materials can also be obtained commercially from Alza Corporation and Nova
Pharmaceuticals, Inc. Liposomal suspensions (including liposomes targeted to infected cells with monoclonal antibodies to viral antigens) can also be used as pharmaceutically acceptable carriers.
These compositions can be prepared according to methods known to those skilled in the art, for example, as described in U.S. Patent No. 4,522,811. It is especially advantageous to formulate oral or parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the subject to be treated; each unit containing a predetermined quantity of active complex calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the dosage unit forms of the invention are dictated by and directly dependent on the unique characteristics of the active compound and the particular therapeutic effect to be achieved.
[0063] The pharmaceutical compositions can be included in a container, pack or dispenser together with instructions for administration.
[0064] The dosage of the FGFBP3 -FGF19 complex will depend on the disorder or condition to be treated and other clinical factors such as weight and condition of the human or animal and the route of administration of the compound. For treating human or animals, the complex can be administered at a dose of between about 0.005 mg/kg of body weight to 500 mg/kg of body weight. Therapy is typically administered at lower dosages and is continued until the desired therapeutic outcome is observed.
[0065] Methods of determining the dosages of composition to be administered to a patient and modes of administering compositions to an organism are disclosed in, for example, WO 96/22976. Those skilled in the art will appreciate that such descriptions are applicable to the present invention and can be easily adapted to it.
[0066] The proper dosage depends on various factors such as the type of disorder being treated, the particular composition being used and the size and physiological condition of the patient.
Therapeutically effective doses for the compositions described herein can be estimated initially from cell culture and animal models. For example, a dose can be formulated in animal models to achieve a circulating concentration range that initially takes into account the IC50 as determined in cell culture assays. The animal model data can be used to more accurately determine useful doses in humans.
[0067] The invention also relates to methods of altering intracellular signaling of a cell, comprising contacting cells with the complex of FGFBP3 or a variant thereof plus FGF19, wherein the cell possesses a receptor that specifically binds to the complex. In one embodiment, the receptor is the fibroblast growth factor receptor 4 (FGF 4). The specific binding of the complex to a receptor will, in turn, initiate the intracellular signaling cascade that is normally associated with the complex. For example, Figure 4 demonstrates that administration of the complex of FGFBP3 or a variant thereof and FGF19 results in phosphorylation of Erkl/2. Accordingly, the present invention provides for methods of stimulating phosphorylation of Erkl/2 in a cell comprising contacting the cell(s) with a complex of FGF19 and FGFBP3 or a variant thereof. Additional methods of the present invention comprise assessing the levels of Erkl/2 phosphorylation, both before and after contacting the cell(s) with the complexes of the present invention and determining the increase or decrease of Erkl/2 phosphorylation in response to the complexes of the present invention.
[0068] Recently, it was shown that FGF19 alone was able to induce phosphorylation of the p90 ribosomal S6 kinase (p90RSK), which is a downstream target of phosphorylated ERK1/2. In turn, phosphorylated p90RSK is known to phosphorylate both ribosomal protein S6 (rpS6) and the eukaryotic translation initiation factor 4B (elF4B). It was also recently shown that FGF19 alone was able to induce phosphorylation of both rpS6 and elF4B. Thus one embodiment of the present invention comprises methods of stimulating phosphorylation of p90RSK, rpS6 and/or elF4B in cells. These methods of phosphorylating p90RSK, rpS6 and/or elF4B comprise contacting the cells with a complex of FGF19 and FGFBP3 or a variant thereof in an amount sufficient to stimulate phosphorylation thereof.
[0069] One target of a phosphorylated p90RSK is glycogen synthase kinase 3a and 3β (GSK3 kinases), which, when phosphorylated, are responsible for inhibition of glycogen synthase (GS). The GSK3 kinases are also inhibited or inactivated when they themselves are phosphorylated. Specifically, phosphorylated p90RSK inhibits or inactivates the GSK3 kinases which block the inhibition of GS. Once the inhibition of GS is removed, GS is activated and, in turn, can trigger production of glycogen. Thus one embodiment of the present invention is directed to methods of increasing glycogen production in a subject in need thereof, with the methods comprising administering a complex of FGF19 and FGFBP3 or a variant thereof in an amount sufficient to stimulate production of glycogen.
[0070] Phosphorylated p90 SK also stimulates protein synthesis at least in the liver. Accordingly, one embodiment of the present invention is directed towards increasing protein synthesis in a subject in need thereof, with the methods comprising administering a complex of FGF19 and FGFBP3 or a variant thereof to the subject in an amount sufficient to stimulate protein synthesis. One example of a liver- synthesized protein is albumin. Accordingly, one specific embodiment of the present invention is directed towards increasing production of albumin in a subject in need thereof, with the methods comprising administering a complex of FGF19 and FGFBP3 or a variant thereof to the subject in an amount sufficient to stimulate production of albumin.
[0071] Likewise, the present invention provides methods of stimulating promoter activity in a cell or population of cells, where the promoter is responsive to activated Erkl/2 or p90 SK with the methods comprising contacting the cell(s) with a complex of FGFBP3 or a variant thereof and FGF19. One of skill in the art would be aware of promoters that respond to activated Erkl/2 or p90RSK. The activity of a complex of a variant of FGFBP3 and FGF19 with respect to stimulating Erkl/2-responsive promoters or p90RSK-responsive promoters may or may not be altered relative to a complex with wild-type FGFBP3 and FGF19. One of skill in the art can readily determine if a promoter is more or less activated over control groups using well known techniques such as transcription of reporter genes, ELISA assays, etc. Additional methods of the present invention comprise assessing the activity of an Erkl/2-responsive promoter both before and after contacting the cell(s) with the complexes of the present invention and determining the increase or decrease of the promoter in response to the complexes of the present invention.
[0072] As used herein, "contacting," when used in connection with the methods of the present invention means bringing the complexes of the present invention in proximity to the target cells such that a specific binding event or a biological effect is possible. Thus, contacting can include adding the complexes in culture medium and applying the culture medium to cells in culture. Of course, contacting would also include administration of the complexes, or pharmaceutical compositions thereof, of the present invention to cells in an intact organism. Compositions for administering the complexes of the present invention have been described herein. [0073] As used herein, "administering," and "administer" are used to mean introducing at least one complex of the present invention into a subject. When administration is for the purpose of treatment, the complex or composition is provided at, or after the onset of, a symptom or condition in need of treatment. The therapeutic administration of this complex or composition serves to attenuate any symptom, or prevent additional symptoms from arising. When administration is for the purposes of preventing a condition from arising ("prophylactic administration"), the complex or composition is provided in advance of any visible or detectable symptom. The prophylactic administration of the complex or comporision serves to attenuate subsequently arising symptoms or prevent symptoms from arising altogether. The route of administration of the complex or composition includes, but is not limited to, topical, transdermal, intranasal, vaginal, rectal, oral, subcutaneous intravenous, intraarterial, intramuscular, intraosseous, intraperitoneal, epidural and intrathecal as previously disclosed herein.
[0074] Furthermore, the methods would also include coadministering one or more substances in addition to the complexes the present invention. The term "coadminister" indicates that each of at least two substances, with one of the substances being a complex of FGFBP3 or a variant thereof and FGF19, is administered during a time frame wherein the respective periods of biological activity or effects of each of the substances overlap. Thus the term includes sequential as well as coextensive administration of the complexes of the present invention with another substance. And similar to administering the compositions of the present invention, coadministration of more than one substance can be for therapeutic and/or prophylactic purposes. If more than one substance is coadministered, the routes of administration of the two or more substances need not be the same.
[0075] The invention also relates to methods of lowering blood glucose levels in a subject, the method comprising administering a complex of FGF19 and FGFBP3 to a subject in need of lowering of blood glucose levels. In one embodiment, the subject is screened prior to administration of the complex.
[0076] The invention also relates to methods of lowering a subject's body weight, the method comprising administering a complex of FGF19 and FGFBP3 to a subject that is in need of lowering its body weight. In one embodiment, the subject is screened prior to administration of the complex.
[0077] The following examples are illustrative and are not intended to limit the scope of the invention described herein. Examples
[0078] Five week old female ob/ob mice, obtained from Jackson Laboratories, were randomly assigned to treatment groups with FGF19 alone, or FGF19 in combination with BP3. The substances were administered in the morning by single intraperitoneal (i.p.) injection. To assess potential therapeutic effects on the metabolic profile with FGF19 or FGF19+BP3 treatments, glucose tolerance tests were performed and changes in body weights were measured. Two series of experiments were performed to investigate acute and sustained effects with the different treatments.
Figure imgf000025_0001
[0079] For all experiments reported here mice were fasted overnight (14 h) before they were subjected to a standard glucose tolerance test (GTT). The GTT is used to evaluate the ability of an organism to metabolize exogenous glucose. In clinical practice the GTT is used to uncover patients with latent diabetes or patients at risk for diabetes, e.g. during pregnancy and is performed after a fasting period by oral administration of a glucose containing drink. In the animal model oral GTT is performed by gavage of a glucose solution, i.e. orally administration. Intraperitoneal injection of a sterile glucose solution was chosen because this allows for a tighter control of dosing of the glucose. Before starting the experiments, the animals were weighed to determine the amount of glucose to inject. The glucose tolerance test was performed in a quiet room and handling was kept down to a minimum to reduce stress during the procedure. A bolus of glucose (1 g/kg) was injected into the intraperitoneal cavity (30 % D-glucose:H20 solution) and blood was sampled from the tail tip at 0, 15, 30, 60, 120 and 180 minutes after glucose injection. Blood glucose levels were determined with a portable glucose meter (Contour®, Bayer Healthcare).
[0080] Ob/ob mice exhibit glucose intolerance and are generally used as a leptin-deficient diabetes model and reflect the human disease well. To examine the metabolic capacity of the animals for glucose the intraperitoneal glucose tolerance test (IPGTT) was performed in ob/ob mice and blood glucose levels were read at 0, 15, 30, 60, 120 and 180 minutes post-injection of glucose. The baseline blood glucose levels were much higher than the normal range (70-120 mg/dl) (Figure 1A, IB, Baseline). After 5 days of treatment (one dose/day) with FGF19, ob/ob mice (n=6) showed an improved glucose tolerance as evidenced by reduced blood glucose levels at 180 minutes post-injection of glucose (p<0.05). The glucose tolerance test (IPGTT) was repeated 9 days after treatment and glucose levels returned to the levels seen before the treatment (Figure 1A).
[0081] For the combination treatment group (FGF19+BP3, n=5), the glucose tolerance test (IPGTT) was seen improved at 60, 120 and 180 minutes post-injection of glucose (p<0.05). The respective blood glucose levels were significantly lower than those of the control group. This effect was sustained even 9 days after the last of 5 doses of FGF19+BP3 (Figure IB). The results from the glucose tolerance test, shown as the area under the curve of the blood glucose levels after glucose injection (AUC), was reduced by 26% compared with baseline in the FGF19 group and by 33% in the FGF19+BP3 group. Most strikingly, the FGF19+BP3 group displayed an improved AUC compared with baseline even 9 days after completed treatment (p<0.01), whereas no such effect was observed in mice receiving FGF19 alone (Figure 1C). In addition, the combination treatment (n=5) improved the AUC even 2 days after a single dose of FGF19+BP3, while FGF19 alone (n=3) did not (p<0.01, Figure 2). These data suggest that BP3 dramatically enhances the FGF19 effect in a standardized IPGTT glucose tolerance test in diabetic mice.
[0082] The effects of acute single doses of BP3 or FGF19 alone, and the combination of BP3+FGF19 were tested in ob/ob mice by IPGTT described above (Figure 3A, B). Mice were starved overnight (14 hours) and then treated for two hours by i.p. injection of vehicle (filled squares, Fig. 3A,B) or FGF19 alone or BP3+FGF19 (open squares Fig. 3A,B). The IPGTT test was initiated two hours after treatments. The combination of BP3+FGF19 induced a striking effect and normalized the baseline blood glucose and the IPGTT blood glucose curve (Fig. 3A). This effect was still present 24 hours after the single dose of BP3+FGF19 (Figure 3C). FGF19 alone showed no significant effect in the test (Fig. 3B). Also, treatment of animals with BP3 alone (without FGF19) showed no significant effect, and even 7 days of dosing of BP3 with a single daily dose of BP3 lacked an effect on the baseline blood glucose or on the IPGTT test (Fig. 3D).
[0083] Two days after a single dose treatment with FGF19 (n=3) or FGF19+BP3 (n=5), ob/ob mice were weighed and the changes in body weight were compared between the two groups. No significant changes in body weight were found (Figure 4A, 4B, 1 treatment) between the groups. However, 5 days of treatment (one dose/day) with FGF19+BP3 lowered the body weight by 1.0±0.7 g/animal, whereas 5 days of treatment (one dose/day) with FGF19 alone was associated with an increase in body weight (2.2±0.8g/animal) which was similar to vehicle treated animals. For the 5 days of treatment, the changes in body weight were significantly different, in both grams and in percent body weight (p<0.05, Figure 4A, 4B, 5 daily treatments).
[0084] BP3 enhances and prolongs the effects of FGF19 on glucose homeostasis, i.e., there is an improvement of the glucose tolerance, from either single or multiple doses. The combination of FGF19+BP3 also reduced the body weight per mouse after 1 dose/day for 5 days, while the animals in FGF19 treatment group gained weight. The combination of BP3 and FGF19 has an unexpectedly better therapeutic effect on obesity than FGF19 alone.
[0085] Given the proposed mechanism of action, it was surprising that a FGFBP3 enhanced the effects of FGF19, i.e., a molecule that that will not be retained by the extracellular matrix and that
administration of a complex of FGF19 and FGFBP3 would affect metabolism more quickly, (i.e. after a single dose), for a longer period (i.e for up to two days after a single dose) and more profoundly (i.e. better efficacy in normalizing glucose tolerance) than FGF19 alone. These data support the concept that BP3+FGF19 treatment improves glucose metabolism in subjects with diabetes and that a single daily dose may be sufficient to last beyond 24 hours.
[0086] To further explore a possible mechanism of action, HepG2 cells (hepatocellular carcinoma cells) were treated with FGF19 with or without BP3. Figure 5 shows that the presence of BP3 enhanced the ability of FGF19 to induce phosphorylation of Erkl/2.

Claims

What is Claimed is:
1. A method of treating a metabolic disorder in a subject, the method comprising administering a complex of fibroblast growth factor 19 (FGF19) and fibroblast growth factor binding protein 3 (FGFBP3) to a subject in need of treatment of a metabolic disorder.
2. The method of claim 1, wherein the metabolic disorder is diabetes.
3. The method of claim 1, wherein the metabolic disorder is obesity.
4. The method of claim 1, wherein the metabolic disorder is hyperglycemia.
5. The method of claim 1, wherein the metabolic disorder is glycogen metabolism.
6. A method of lowering blood glucose levels in a subject, the method comprising administering a complex of fibroblast growth factor 19 (FGF19) and fibroblast growth factor binding protein 3 (FGFBP3) to a subject in need of lowering of blood glucose levels.
7. A method of lowering a subject's body weight, the method comprising administering a complex of fibroblast growth factor 19 (FGF19) and fibroblast growth factor binding protein 3 (FGFBP3) to a subject in need of lowering its body weight.
8. A method of regulating at least one component of postprandial metabolism in a subject, the method comprising administering a complex of fibroblast growth factor 19 (FGF19) and fibroblast growth factor binding protein 3 (FGFBP3) to a subject in need of regulation of postprandial metabolism.
9. The method of claim 8, wherein the at least one component of postprandial metabolism is hepatic glycogen synthesis.
10. The method of claim 8, wherein the at least one component of postprandial metabolism is protein synthesis.
11. The method of claim 8, wherein the at least one component of postprandial metabolism is clearance of plasma glucose.
12. The method any of claims 8-11, wherein hepatic triglyceride levels are not affected.
13. A method of increasing protein synthesis is a cell, the method comprising contacting the cell with a complex of FGF19 and FGFBP3 in an amount sufficient to stimulate protein production in the cell.
14. The method of claim 13, wherein the cell is in cell culture.
15. The method of claim 13, wherein the cell is part of an organism.
PCT/US2014/026938 2013-03-14 2014-03-14 Compositions and treatments of metabolic disorders using fgf binding protein 3 and fgf 19 WO2014152090A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US14/853,482 US9789160B2 (en) 2013-03-14 2015-09-14 Treatments for lowering glucose levels using FGF binding protein 3
US15/784,730 US20180214514A1 (en) 2013-03-14 2017-10-16 Compositions and Treatments of Metabolic Disorders Using FGF Binding Protein 3
US16/840,046 US20200246425A1 (en) 2013-03-14 2020-04-03 Treatments of Metabolic Disorders Using FGF Binding Protein 3

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361782347P 2013-03-14 2013-03-14
US61/782,347 2013-03-14

Related Child Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2014/026937 Continuation-In-Part WO2014152089A1 (en) 2013-03-14 2014-03-14 Compositions and treatments of metabolic disorders using fgf binding protein 3
US14/853,482 Continuation-In-Part US9789160B2 (en) 2013-03-14 2015-09-14 Treatments for lowering glucose levels using FGF binding protein 3

Publications (1)

Publication Number Publication Date
WO2014152090A1 true WO2014152090A1 (en) 2014-09-25

Family

ID=51581110

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/026938 WO2014152090A1 (en) 2013-03-14 2014-03-14 Compositions and treatments of metabolic disorders using fgf binding protein 3 and fgf 19

Country Status (1)

Country Link
WO (1) WO2014152090A1 (en)

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9089525B1 (en) 2011-07-01 2015-07-28 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants and fusions of FGF19 polypeptides for reducing glucose levels in a subject
US9273107B2 (en) 2012-12-27 2016-03-01 Ngm Biopharmaceuticals, Inc. Uses and methods for modulating bile acid homeostasis and treatment of bile acid disorders and diseases
US9290557B2 (en) 2012-11-28 2016-03-22 Ngm Biopharmaceuticals, Inc. Compositions comprising variants and fusions of FGF19 polypeptides
WO2016090546A1 (en) 2014-12-09 2016-06-16 Wentao Zhang Nbp158 and uses thereof
US9925242B2 (en) 2012-12-27 2018-03-27 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants and fusions of FGF19 polypeptides for treatment of nonalcoholic steatohepatitis
US9963494B2 (en) 2012-11-28 2018-05-08 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants and fusions of FGF19 polypeptides for reducing glucose levels in a subject
US10093735B2 (en) 2014-01-24 2018-10-09 Ngm Biopharmaceuticals, Inc. Beta-klotho binding proteins
US10369199B2 (en) 2013-10-28 2019-08-06 Ngm Biopharmaceuticals, Inc. Methods of using variants of FGF19 polypeptides for the treatment of cancer
US10398758B2 (en) 2014-05-28 2019-09-03 Ngm Biopharmaceuticals, Inc. Compositions comprising variants of FGF19 polypeptides and uses thereof for the treatment of hyperglycemic conditions
US10434144B2 (en) 2014-11-07 2019-10-08 Ngm Biopharmaceuticals, Inc. Methods for treatment of bile acid-related disorders and prediction of clinical sensitivity to treatment of bile acid-related disorders
US10456449B2 (en) 2014-06-16 2019-10-29 Ngm Biopharmaceuticals, Inc. Methods and uses for modulating bile acid homeostasis and treatment of bile acid disorders and diseases
US10517929B2 (en) 2014-10-23 2019-12-31 Ngm Biopharmaceuticals, Inc. Pharmaceutical compositions comprising FGF19 variants
US10744185B2 (en) 2015-11-09 2020-08-18 Ngm Biopharmaceuticals, Inc. Methods of using variants of FGF19 polypeptides for the treatment of pruritus
US10800843B2 (en) 2015-07-29 2020-10-13 Ngm Biopharmaceuticals, Inc. Beta klotho-binding proteins
US11370841B2 (en) 2016-08-26 2022-06-28 Ngm Biopharmaceuticals, Inc. Methods of treating fibroblast growth factor 19-mediated cancers and tumors

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020155543A1 (en) * 1997-11-25 2002-10-24 Genentech, Inc. Fibroblast growth factor-19 (FGF-19) nucleic acids and polypeptides and methods of use for the treatment of obesity and related disorders

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020155543A1 (en) * 1997-11-25 2002-10-24 Genentech, Inc. Fibroblast growth factor-19 (FGF-19) nucleic acids and polypeptides and methods of use for the treatment of obesity and related disorders

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BEENKEN ET AL.: "The FGF family: biology, pathophysiology and therapy.", NAT REV DRUG DISCOV, vol. 8, no. 3, March 2009 (2009-03-01), pages 235 - 253 *
KIR ET AL.: "FGF19 as a postprandial, insulin-independent activator of hepatic protein and glycogen synthesis.", SCIENCE, vol. 331, no. 6024, 25 March 2011 (2011-03-25), pages 1621 - 1624 *
ZHANG ET AL.: "Effect of FGF-binding protein 3 on vascular permeability.", J BIOL. CHEM, vol. 283, no. 42, 17 October 2008 (2008-10-17), pages 28329 - 28337 *

Cited By (38)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9089525B1 (en) 2011-07-01 2015-07-28 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants and fusions of FGF19 polypeptides for reducing glucose levels in a subject
US11065302B2 (en) 2011-07-01 2021-07-20 Ngm Biopharmaceuticals, Inc. Compositions comprising fusion variants of FGF19 polypeptides
US10413590B2 (en) 2011-07-01 2019-09-17 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants of FGF19 polypeptides for reducing body mass in a subject
US9751924B2 (en) 2011-07-01 2017-09-05 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising fusion variants of FGF19 polypeptides for reducing glucose levels in a subject
US9670260B2 (en) 2011-07-01 2017-06-06 Ngm Biopharmaceuticals, Inc. Compositions comprising fusion variants of FGF19 polypeptides
US9580483B2 (en) 2011-07-01 2017-02-28 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants and fusions of FGF19 polypeptides for treatment of diabetes
US10758590B2 (en) 2012-11-28 2020-09-01 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants and fusions of FGF 19 polypeptides for treating diabetes
US11066454B2 (en) 2012-11-28 2021-07-20 Ngm Biopharmaceuticals, Inc. Compositions comprising variants and fusions of FGF19 polypeptides
US9290557B2 (en) 2012-11-28 2016-03-22 Ngm Biopharmaceuticals, Inc. Compositions comprising variants and fusions of FGF19 polypeptides
US9963494B2 (en) 2012-11-28 2018-05-08 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants and fusions of FGF19 polypeptides for reducing glucose levels in a subject
US9889178B2 (en) 2012-12-27 2018-02-13 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants and fusions of FGF19 polypeptides for modulating bile acid homeostasis in a subject having nonalcoholic steatohepatitis
US11103554B2 (en) 2012-12-27 2021-08-31 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants of FGF19 polypeptides for reducing bile acid synthesis in a subject having cirrhosis
US9878009B2 (en) 2012-12-27 2018-01-30 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants and fusions of FGF19 polypeptides for modulating bile acid homeostasis in a subject having error of bile acid synthesis
US9895416B2 (en) 2012-12-27 2018-02-20 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants and fusions of FGF19 polypeptides for modulating bile acid homeostasis in a subject having cholestasis
US11564972B2 (en) 2012-12-27 2023-01-31 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants of FGF19 polypeptides for treating primary biliary cirrhosis in a subject
US9925242B2 (en) 2012-12-27 2018-03-27 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants and fusions of FGF19 polypeptides for treatment of nonalcoholic steatohepatitis
US9974833B2 (en) 2012-12-27 2018-05-22 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants and fusions of FGF19 polypeptides for modulating bile acid homeostasis in a subject having pregnancy intrahepatic cholestasis
US9889177B2 (en) 2012-12-27 2018-02-13 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants and fusions of FGF19 polypeptides for modulating bile acid homeostasis in a subject having primary sclerosing cholangitis
US9273107B2 (en) 2012-12-27 2016-03-01 Ngm Biopharmaceuticals, Inc. Uses and methods for modulating bile acid homeostasis and treatment of bile acid disorders and diseases
US9878008B2 (en) 2012-12-27 2018-01-30 Ngm Biopharmaceuticals, Inc. Methods of using compositions comprising variants and fusions of FGF19 polypeptides for modulating bile acid homeostasis in a subject having bile acid diarrhea or bile acid malabsorption
US10369199B2 (en) 2013-10-28 2019-08-06 Ngm Biopharmaceuticals, Inc. Methods of using variants of FGF19 polypeptides for the treatment of cancer
US10744191B2 (en) 2014-01-24 2020-08-18 Ngm Biopharmaceuticals, Inc. Beta klotho-binding proteins and methods of use thereof
US10093735B2 (en) 2014-01-24 2018-10-09 Ngm Biopharmaceuticals, Inc. Beta-klotho binding proteins
US11596676B2 (en) 2014-01-24 2023-03-07 Ngm Biopharmaceuticals, Inc. Methods of treating nonalcoholic steatohepatitis comprising administering an anti-human beta klotho antibody or binding fragment thereof
US10398758B2 (en) 2014-05-28 2019-09-03 Ngm Biopharmaceuticals, Inc. Compositions comprising variants of FGF19 polypeptides and uses thereof for the treatment of hyperglycemic conditions
US11241481B2 (en) 2014-06-16 2022-02-08 Ngm Biopharmaceuticals, Inc. Methods and uses for modulating bile acid homeostasis and treatment of bile acid disorders and diseases
US10456449B2 (en) 2014-06-16 2019-10-29 Ngm Biopharmaceuticals, Inc. Methods and uses for modulating bile acid homeostasis and treatment of bile acid disorders and diseases
US10517929B2 (en) 2014-10-23 2019-12-31 Ngm Biopharmaceuticals, Inc. Pharmaceutical compositions comprising FGF19 variants
US10434144B2 (en) 2014-11-07 2019-10-08 Ngm Biopharmaceuticals, Inc. Methods for treatment of bile acid-related disorders and prediction of clinical sensitivity to treatment of bile acid-related disorders
US11141460B2 (en) 2014-11-07 2021-10-12 Ngm Biopharmaceuticals, Inc. Methods for treatment of bile acid-related disorders and prediction of clinical sensitivity to treatment of bile acid-related disorders
CN106413739B (en) * 2014-12-09 2019-09-17 张文涛 NBP158 and application thereof
US10954282B2 (en) 2014-12-09 2021-03-23 Wentao Zhang NBP158 and uses thereof
CN106413739A (en) * 2014-12-09 2017-02-15 张文涛 NBP158 and uses thereof
WO2016090546A1 (en) 2014-12-09 2016-06-16 Wentao Zhang Nbp158 and uses thereof
US10800843B2 (en) 2015-07-29 2020-10-13 Ngm Biopharmaceuticals, Inc. Beta klotho-binding proteins
US11667708B2 (en) 2015-07-29 2023-06-06 Ngm Biopharmaceuticals, Inc. Anti-human beta klotho antibody or binding fragment thereof and methods of their use
US10744185B2 (en) 2015-11-09 2020-08-18 Ngm Biopharmaceuticals, Inc. Methods of using variants of FGF19 polypeptides for the treatment of pruritus
US11370841B2 (en) 2016-08-26 2022-06-28 Ngm Biopharmaceuticals, Inc. Methods of treating fibroblast growth factor 19-mediated cancers and tumors

Similar Documents

Publication Publication Date Title
WO2014152090A1 (en) Compositions and treatments of metabolic disorders using fgf binding protein 3 and fgf 19
US20200246425A1 (en) Treatments of Metabolic Disorders Using FGF Binding Protein 3
JP2023071932A (en) Fgf21 compound/glp-1r agonist combinations with optimized activity ratio
JP7028775B2 (en) FGF21 mutant
US20190085043A1 (en) Fusion proteins for treating a metabolic syndrome
WO2014152089A1 (en) Compositions and treatments of metabolic disorders using fgf binding protein 3
KR20180120739A (en) Glucagon and GLP-1 co-agonists for the treatment of obesity
JP2016505627A (en) Therapeutic agents, compositions, and methods for glycemic control
US20190151416A1 (en) Fibroblast growth factor (fgf) 1 proteins with glucose lowering ability and reduced mitogenicity
CA2920679A1 (en) Methods of using interleukin-10 for treating diseases and disorders
WO2019243557A1 (en) Fgf21 compound / glp-1r agonist combinations with optimized activity ratio
US20230265152A1 (en) Glp-1r agonist / fgf21 fusion proteins
US20230097335A1 (en) Fibroblast growth factor 1 (fgf1) mutant proteins that selectively activate fgfr1b to reduce blood glucose
US20220118052A1 (en) Materials and methods for modulating glucose uptake
KR20230034356A (en) GLP-1R agonist peptides with reduced activity
US20190276510A1 (en) Use of fibroblast growth factor 1 (fgf1)-vagus nerve targeting chimeric proteins to treat hyperglycemia
KR102646717B1 (en) Fusion protein comprising cell penetrating peptide and fkbp12 and use thereof
WO2004015113A2 (en) Albumin-fused ciliary neurotrophic factor

Legal Events

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

Ref document number: 14768570

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14768570

Country of ref document: EP

Kind code of ref document: A1