AU2008201780B2 - Fibroblast growth factor-like polypeptides - Google Patents

Fibroblast growth factor-like polypeptides Download PDF

Info

Publication number
AU2008201780B2
AU2008201780B2 AU2008201780A AU2008201780A AU2008201780B2 AU 2008201780 B2 AU2008201780 B2 AU 2008201780B2 AU 2008201780 A AU2008201780 A AU 2008201780A AU 2008201780 A AU2008201780 A AU 2008201780A AU 2008201780 B2 AU2008201780 B2 AU 2008201780B2
Authority
AU
Australia
Prior art keywords
fgf
polypeptide
seq
cells
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU2008201780A
Other versions
AU2008201780A1 (en
Inventor
Benxian Liu
Arien Read Thomason
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Amgen Inc
Original Assignee
Amgen Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2004242465A external-priority patent/AU2004242465C1/en
Application filed by Amgen Inc filed Critical Amgen Inc
Priority to AU2008201780A priority Critical patent/AU2008201780B2/en
Publication of AU2008201780A1 publication Critical patent/AU2008201780A1/en
Application granted granted Critical
Publication of AU2008201780B2 publication Critical patent/AU2008201780B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Abstract

The present invention relates to a novel Fibroblast Growth Factor-like (FGF-like) polypeptides and nucleic acid molecules encoding the same. The invention also relates to vectors, host cells, antibodies and methods for producing FGF-like polypeptides. Also provided for are methods for the diagnosis and treatment of diseases associated with FGF-like polypeptides.

Description

FIBROBLAST GROWTH FACTOR-LIK POLYPEPTIDES Field of the Invention The present invention relates to novel Fibroblast Growth Factor-like 5 (FGF-like) polypeptides and nucleic acid molecules encoding the same. The invention also relates to vectors, host cells, antibodies and methods for producing FGF-like polypeptides. Also provided for are methods for the diagnosis and treatment of diseases associated with FGF-like polypeptides. 10 Backround of the Invention Technical advances - in the identification, cloning, expression, and manipulation of nucleic acid molecules have greatly accelerated the discovery of novel therapeutics based upon deciphering the human genone. Rapid nucleic acid' sequencing techniques can now generate sequence information at i5 unprecedented rates and, coupled wit computational analyses, allow the assembly of overlapping sequences into entire genomes and the identification of polypeptide-encoding regions. Comparison of a predicted amino acid sequence against a database compilation of known amino acid sequences can allow one to determine the extent of homology to previously identified sequences and/or 20 tuctural landmaarks. Cloning and expression of a polypeptide-encoding region of a nncleic acid molecule provides a polypeptide product for structural and functional analysis. Manipulation of nucleic acid molecule and encoded polypeptides to give variants and derivatives thereof may confer advantageous properties on a product for use as a therapeutic. 25 In spite of the significant technical advances in genome research over the past decade, the potential for development of novel therapeutics based on the human genome is still largely unrealized. Genes encoding potentially beneficial protein therapeutics, or those encoding polypeptides that may act as "targets" for. therapeutic molecules, have still not been identified. In addition, structural and functional analyses of polypeptide products from many human genes have not been undertaken. Accordingly, it is an object of the invention to identify novel polypeptides and nucleic acid molecules encoding the same -which have diagnostic or 5 therapeutic benefit, Summary of the Invention The present invention relates to novel FGF-like -nucleic acid molecules and encoded polypeptides. 10 The invention provides for an isolated nucleic acid molecule comprising a nucleotide sequence selected from the group consisting of: (a) the nucleotide sequence as set forti in SEQ ID NO: I or SEQ ID NO: 3; (b) the nucleotide sequence of the DNA insert in ATCC Deposit No. 15 PTA-626; (c) a nucleotide sequence encoding the polypeptide as set forth in SEQ ID NO: 2 or SBQ ID NO: 4; (d) a nucleotide sequence which hybridizes under moderately or highly stringent conditions to the complement of any of (a)-(c); and 20 (e) a nucleotide sequence complementary to any of(a)-(c). The invention also provides for an isolated nucleic acid molecule comprising a nucleotide sequence selected from the group consisting of; (a) a nucleotide sequence encoding a polypptide that is at least about 25 80 percent identical to the polypeptide as set forth in SEQ ID NO: 2 or SEQ ID NO: 4, wherein the encoded polypeptide activates one or more FGF receptors, regulates the growth and differentiation of cells within the liver or pancreas, regulates other cell types following secretion from the liver or pancreas, plays a role in liver or pancreas chemotaxis, or has an oncogenic activity; -3 (b) a nucleotide sequence encoding an allelic variant or splice variant of the nucleotide sequence as set forth in SEQ ID NO: 1, SEQ ID) NO: 3, or (a); (c) a region of the nucleotide sequence of SEQ ID NO: 1, SEQ ID NO: 3, (a), or (b) encoding a polypeptide fragment of at least about 25 amino acid 5 residues, wherein the encoded polypeptide activates one or more FGF receptors, regulates the growth and differentiation of cells within the liver or pancreas, regulates other cell types following secretion from the liver or pancreas, plays a role in liver or pancreas chemnotaxis, has an oncogenic activity, or serves as an antigen for generating antibodies; 10 (d) a region of the nucleotide sequence of SEQ ID NO: 1, SEQ ID NO; 3, or any of (a) - (c) comprising a fragment of at least about 16 nucleotides; (e) a nucleotide sequence which hybridizes under moderately or highly stringent conditions to the complement of any of (a) - (d); and (1) a nucleotide sequence complementary to any of (a) - (d). 15 The invention further provides for an isolated nucleic acid molecule comprising a nucleotide sequence selected from the group consisting of: (a) a nucleotide sequence encoding a polypeptide as set forth in SEQ ID NO: 2 with at least one conservative amino acid substitution, wherein the 20 encoded polypeptide activates one or more FGF receptors, regulates the growth and differentiation of cells within the liver or pancreas, regulates other cell types following secretion from the liver or pancreas, plays a role in liver or pancreas chemotaxis, or has an oncogenic activity; (b) a nucleotide sequence encoding a polypeptide as set forth in SEQ 25 ID NO: 2 with at least one amino acid insertion, wherein the encoded polypeptide activates one or more FGF receptors, regulates the growth and differentiation of cells within the liver or pancreas, regulates other cell types following secretion from the liver or pancreas, plays role in liver or pancreas chemotaxis, or has an oncogenic activity; -4 (c) a nucleotide sequence encoding a polypeptide as set forth in SEQ ID NO: 2 with at least one amino acid deletion, wherein the encoded polypeptide activates one or more FGF receptors, regulates the growth and differentiation of cells within the liver or pancreas, regulates other cell types following secretion 5 from the liver or pancreas, plays a role in liver or pancreas chemotaxis, or has an oncogenic activity; (d) a nucleotide sequence encoding a polypeptide as set forth in SEQ ID NO: 2 which has a carboxyl- and/or amino-tenninal truncation, wherein the encoded polypeptide activates one or more FGF receptors, regulates the growth 10. and differentiation of cells within the liver or pancreas, regulates other cell types following secretion from the liver or pancreas, plays a role in liver or pancreas chemotaxis. or has an oncogenic activity; (e) a nucleotide sequence encoding a polypeptide as set forth in SEQ ID NO: 2 with at least one modification selected from the group consisting of 15 amino acid substitutions, amino acid insertions, amino acid deletions, carboxyl tenninal truncation, and amino-terminal truncation, wherein the encoded polypeptide activates one or more FGF receptors, regulates the growth and differentiation of cells within the liver or pancreas, regulates other cell types following secretion from the liver or pancreas, plays a role in liver or pandreas 20 chemotaxis, or has an oncogenic activity; (f) a region of the nucleotide sequence of any of (a) - (e) comprising a fragment of at least about 16 nucleotides; (g) a nucleotide sequence which hybridizes under moderately or highly stringent conditions to the complement of any of (a) - (f); and 25 (h) a nucleotide sequence complementary to any of (a) - (e). The present invention provides for an isolated polypeptide comprising the amino acid sequence selected from the group consisting of: (a) the amino acid sequence as set forth in SEQ ID NO: 2 or SEQ ID 3o NO: 4; and -5 (b) the amino acid sequence encoded by the DNA insert of ATCC Deposit No. PTA-626. The invention also provides for an isolated polypeptide comprising the 5 amino acid sequence selected from the group consisting of: (a) the amino acid sequence as set forth in either SEQ ID NO: 5 or SEQ ID NO: 6, optionally further comprising an ainino-terninal methionine; (b) an amino acid sequence for an ortholog of either SEQ ID NO: 2 or SEQ ID NO: 4, wherein the encoded polypeptide activates one or more FGF 10 receptors, regulates the growth and differentiation of cells within the liver or pancreas, regulates other cell types following secretion from the liver or pancreas, plays a role in liver or pancreas chenotaxis, or has an oncogenic activity; (c) an amino acid sequence that is at least about 80 percent identical to the amino acid sequence of either SEQ IDI NO: 2 or SEQ ID NO: 4, wherein the is encoded polypeptide activates one or more FGF receptors, regulates the growth and differentiation of cells within the liver or pancreas, regulates other cell types following secretion from the liver or pancreas, plays a role in liver or pancreas chemotaxis, or has an oncogenic activity; (d) a fragment of the amino acid sequence set forth in either SEQ ID 20 NO: 2 or SEQ ID NO 4 comprising at least about 25 amino acid residues, wherein the encoded polypeptide activates one or more FGF receptors, regulates the growth and differentiation of cells within the liver or pancreas, regulates other cell types following secretion from the liver or pancreas, plays a role in liver or pancreas chemotaxs, has an oncogenic activity, or serves as an antigen for 25 generating antibodies; and (e) an amino acid sequence for an allelic variant or splice variant of either the amino acid sequence as set forth in either SEQ ID NO: 2 or SEQ ID NO: 4; the amino acid sequence encoded by the DNA insert of ATCC Deposit No. PTA-626; (a), (b), or (e). 30 -6 The invention further provides for an isolated polypeptide comprising the amino acid sequence selected from the group consisting of: (a) the amino acid sequence as set forth in either SEQ ID NO: 2 or SEQ ID NO: 4 with at least one conservative amino acid substitution, wherein the 5 encoded polypeptide activates one or more FGF receptors, regulates the growth and differentiation of cells within the liver or pancreas, regulates other cell types following secretion from the liver or pancreas, plays a role in liver or pancreas chemotaxis, or has an oncogenic activity; (b) the amino acid sequence as set forth in either SEQ ID NO: 2 or lo SEQ ID NO: 4 with at least one amino acid insertion, wherein the encoded polypeptide activates one or more FGF receptors, regulates the growth and differentiation of cells within the liver or pancreas, regulates other cell types following secretion from the liver or pancreas, plays a role in liver or pancreas chemotaxis, or has an oncogenic activity; 15 (c) the amino acid sequence as set forth in either SEQ ID NO: 2 or SEQ MD NO: 4 with at least one amino acid deletion, wherein the encoded polypeptide activates one or more FGF receptors, regulates the growth and differentiation of cells within the liver or pancreas, regulates other cell types following secretion from the liver or pancreas, plays a role in liver or pancreas 20 chemotaxis, or has an oncogenic activity (d) the amino acid sequence as set forth in either SEQ ID NO: 2 or SEQ ID NO: 4 which has a C- and/or N- terinal truncation, wherein the encoded polypeptide activates one or more FGF receptors, regulates the growth and differentiation of cells within the liver or pancreas, regulates other cell types 25 following secretion from the liver or pancreas, plays a role in liver or pancreas chemotaxis, or has an oncogenic activity; and (e) the amino acid sequence as set forth in either SEQ ID NO: 2 or SEQ ID NO: 4 with at least one modification selected from the group consisting of amino acid substitutions, amino acid insertions, amino acid deletions, C 30 terminal trucation, and N-terminal truncation, wherein the encoded polypeptide -7/1 activates one or more FGF receptors, regulates the growth and differentiation of cells within the liver or pancreas, regulates other cell types following secretion from the liver or pancreas, plays a role in liver or pancreas chemotaxis, or has an oncogenic activity. The invention also provides for an expression vector comprising the nucleic acid 5 molecules as set forth above, host cells comprising the expression vectors of the invention, and a method of production of an FOF-like polypeptide comprising culturing the host cells and optionally isolating the polypeptide so produced. The invention also provides a recombinant host cell comprising an isolated nucleic acid molecule having a nucleotide sequence that encodes the polypeptide of SEQ ID NO: 10 5. The invention also provides a process of producing a polypeptide encoded by an isolated nucleic acid molecule having a nucleotide sequence that encodes the polypeptide of SEQ ID NO: 5 comprising, culturing a recombinant host cell comprising the nucleic acid molecule having a nucleotide sequence that encodes the polypeptide of SEQ ID NO: 15 5 under suitable conditions to express the polypeptide. The invention also provides a vector comprising an isolated nucleic acid molecule having a nucleotide sequence that encodes the polypeptide of SEQ ID NO: 5. The invention also provides a method of identifying a modulator of FGF-like polypeptide activity comprising: (a) incubating a polypeptide comprising the amino acid 20 sequence of SEQ ID NO: 2, the amino acid sequence of SEQ ID NO: 5, or an amino acid sequence that is at least 95% identical to the amino acid sequence of either SEQ ID NO: 2 or SEQ ID NO: 5, with a test molecule, under conditions that permit interaction of the polypeptide with the test molecule; and (b) measuring FGF-like polypeptide activity to identify a test molecule that modulates FGF-like polypeptide activity. 25 The invention also provides a method of identifying a modulator of FGF-like polypeptide activity comprising: (a) transforming a host cell with a vector comprising the nucleotide sequence of SEQ ID NO: 1, the DNA insert in ATCC Deposit No. PTA-626, or a nucleotide sequence that is 95% identical to the nucleotide sequence of either SEQ ID NO: 1 or the DNA insert in ATCC Deposit No. PTA-626; (b) incubating the host cell with 30 a test molecule, under conditions that permit interaction of the polypeptide encoded by the nucleotide sequence of either SEQ ID NO: 1 or the DNA insert in ATCC Deposit No.
- 712 PTA-626 with the test molecule; and (c) measuring FGF-like polypeptide activity to identify a test molecule that modulates FGF-like polypeptide activity.A transgenic non human animal comprising a nucleic acid molecule encoding an FGF-like polypeptide is also encompassed by the invention. The FGF-like nucleic acid molecules are introduced 5 into the animal in a manner that allows expression and increased levels of an FGF-like polypeptide, which may include increased circulating levels. Alternatively, the FGF-like nucleic acid molecules are introduced into the animal in a manner that prevents expression of endogenous FGF-like polypeptide (i. e., generates a transgenic animal possessing an FGF-like polypeptide gene knockout). The transgenic non-human animal is preferably a 10 mammal, and more preferably a rodent, such as a rat or a mouse, Preferably the FGF-like transgene is expressed in the liver under the control of the apolioprotein E promoter, or ubiquitously under the control of the beta actin promoter. Also provided are derivatives of the FGF-like polypeptides of the invention, fusion polypeptides comprising the FGF-like polypeptides of the invention, and antibodies 15 specifically binding the FGF-like polypeptides of the invention. Compositions comprising the nucleotides or polypeptides of the invention and a carrier, adjuvant, solubilizer, stabilizer or anti-oxidant, or other pharmaceutically acceptable agent are also encompassed by the invention, The compositions may include pharmaceutical compositions comprising 20 -8 therapeutically effective amounts of the nucleotides or polypeptides of the invention, and methods of using the polypeptides and nucleic acid molecules. Surprisingly, FGF-like polypeptide appeared to be primarily expressed in the liver (Northern analysis) and pancreatic islets (in situ analysis), thereby 5 distinguishing it from all other members of the FGF family. The present polypeptide, and its useful nucleic acid intermediates, may have utility, therefore, in differentiating liver cells or pancreatic islet cells from background. Further, given the localization of FGF-like polypeptide expression, the structural similarity of FGF-like polypeptide to members of the FGF family, and the likelihood that 10 FGF-like polypeptide is secreted into the bloodstream. where it may exert effects on distal sites, the present polypeptides may provide benefits in the stimulation of cells within or near the liver, regulation of intestinal cell activity, stimulation of cells within or near pancreatic islets, regulation of neuronal cells, stimulation or inhibition of angiogenesis, stimulation of epithelium or mesenchymal components 15 of granulation tissue, stimulation of corneal epithelium, lens, or retinal tissue, regeneration of renal -tubules, hematopoietic cell regulation, regulation of hair follicle growth, regulation of pulmonary epithelium, or stimulation of either epithelial, mesenchymal, hematopoictic, or neuronal cells or tissues, particularly as a therapeutic pharmaceutical composition. 20 FGF-like polypeptides may also be useful as growth or fat deposition inhibitors, and therefore may be useful in the treatment of excessive growth (for example, acromegaly), premature maturation, obesity or diabetes. Inhibitors such as antibodies, binding proteins or small molecules - that interfere with the interaction of FGF-like polypeptides and their receptor(s) may be useful in 25 stimulating body growth and maturation. Therefore, such inhibitors may be useful in treating short stature, delayed maturation, or other conditions generally associated with impainnent of signaling of growth hormone or its mediator, insulin-like growth factor. The FGF-like polypeptides and nucleic acid molecules of the invention, or. 30 agonists or antagonists of their biological activity, may be used for therapeutic or -9 diagnostic purposes to treat, prevent and/or detect a medical condition such as cirrhosis or other toxic insult of the liver; inflammatory bowel disease, mucositis, Crohn's disease, or other gastrointestinal abnormality; diabetes; obesity; neurodegenerative diseases; wounds; damage to the corneal epithelium, lens, or 5 retinal tissue; damage to renal tubules as a result of acute tubular necrosis; hematopoietic cell reconstitution following chemotherapy; wasting syndromes (for example, cancer associated cachexia), multiple sclerosis, myopathies; short stature, delayed maturation, excessive growth (for example, acromegaly), premature maturation; alopecia; diseases or abnormalities of androgen target 10 organs; infantile respiratory distress syndrome, bronchopulmonary dysplasia, acute respiratory distress syndrome, or other lung abnormalities; ef tumors of the. eye -or other tissues; atherosclerosis; bypercholesterolemia; diabetes; obesity; stroke; osteoporosis; osteoarthritis; degenerative joint disease; muscle atrophy; sarcopenia; decreased lean body mass; baldness; wrinkles; increased fatigue; 15 decreased stamina; decreased cardiac function; immune system dysfunction; cancer; Parkinson's disease; senile dementia; Alzheimer's disease; and decreased cognitive function. , The invention provides for treating, preventing or ameliorating a disorder comprising administering to an animal an FGF-like polypeptide. The invention also provides for a method of diagnosing such a 20 disorder or a susceptibility to such a disorder in an animal which includes both determining the presence or amount of expression of an FO-like polypeptide and diagnosing such a disorder or a susceptibility to such a disorder based on the presence or amount of expression of an FGF-like polypeptide. The animal is preferably a mammal, and more preferably a human. The present invention also 25 relates to methods for the manufacture of a medicament for the treatment of a disorder such as those mentioned above. The invention aso provides for the use of antibodies or other inhibitors of the binding of FG-like polypeptide to its receptor for the treatment of the same diseases listed above, and for the treatment of tumors.
- 10 The invention also provides for a method of identifying a test molecule which binds to an FGF-like polypeptide wherein the method comprises contacting an FGF-like polypeptide with a test molecule and determining the extent of binding of the test molecule to the polypeptide. The method further comprises 5 determining whether such test molecules are agonists or antagonists of an FGF like polypeptide. The invention also provides for a method of testing the impact of molecules on the expression of FGF-like polypeptide or on the activity of FGF like polypeptide. 10 A method of regulating expression and modulating (Ite., increasing or decreasing) levels of an FGF-like polypeptide are also encompassed by the invention. One method comprises administering to an aimal a nucleic acid molecule encoding an FGF-like polypeptide. In another method, a nucleic acid molecule domprising elements that regulate expression of an FGF-like 15 polypeptide may be administered. Exarples of these methods include gene therapy and anti-sense therapy. Description of the Figures Figure 1 illustrates the nucleotide sequence of the urine FGF-like gene (SEQ ID 20 NO: 3) and the deduced amino acid sequence of murine FGF-like protein (SEQ ID NO: 4); Figures 2A-2B illustrate the nucleotide sequence of the human FOF-like gene (SEQ ID NO: 1) and the deduced amino acid sequence of human FGF-like protein 25 (SEQ ID NO: 2); Figures 3A-3D illustrate the amino acid sequence alignment of human FGF-like protein (hAgp-26257; SEQ ID NO: 2), murine FGF-lile protein (mAgp-26257; SEQ ID NO: 4), human FGF-14 (Hfgfl4; SEQ ID NO: 16), murine FGF-14 30 (Mfgfl4; SEQ ID NO: 26), human FGF-12 (Hfgfl2; SEQ ID NO: 15), murine - 11 FGF-13 (Mfgfl3; SEQ ID NO: 25), human FGF-5 (Hfgf5; SEQ ID NO: 20), murine FGF-5 (Mfgf5; SEQ ID NO: 30), human FGF-6 (Hfgf6; SEQ ID NO: 21), murine FGF-6 (Mfgf6; SEQ ID NO: 31), human FGF-4 (Hfgf4; SEQ ID NO; 19), mutine FGF-4 (Mfgf4; SEQ ID NO: 29), human FGF-3 (Hfgf3; SEQ ID NO: 18), 5 urine FGF-3 (Mfgf3; SEQ ID NO: 28), human FGF-7 (Hfgf7; SEQ ID NO: 22), murine FGF-7 (Mfgf7; SEQ ID NO: 32), human FGF-9 (Hfgf9; SEQ ID NO: 23), murine FGF-9 (Mfgf9; SEQ ID NO: 33), human FGF-I (Hfgfl; SEQ ID NO: 14), murine FGF-1 (Mfgfl; SEQ ID NO: 24), human FGF-2 (Hfgf2; SEQ ID NO: 17), murine FGF-2 (Mfgf2; SEQ ID. NO: 27), and the resulting FGF consensus lo sequence (cons); Figures 4A-4C illustrate the results of (A) a Northern blot analysis of marine FGF-like polypeptide expression, (B) a Northern blot analysis of human FGF-like polypeptide expression, and (C) a dot, blot analysis of human FGF-like 15 polypeptide expression. Detailed Description of the Invention The section headings herein are for organizational purposes only and are not to be construed as limiting the subject matter described therein. All references 20 cited in this application are expressly incorporated by reference hereim Definitions The term "FO-like nucleic acid molecule" refers to a nucleic acid molecule comprising or consisting essentially of a nucleotide sequence as set forth 25 in SEQ ID NO: 1 or SEQ ID NO: 3, comprising or consisting essentially of a nucleotide sequence encoding the polypeptide as set forth in SEQ ID NO: 2 or SEQ ID NO: 4, comprising or consisting essentially of a nucleotide sequence of the DNA insert in ATCC Deposit No. PTA-626, or nucleic acid molecules related thereto.
-12 Related nucleic acid molecules comprise or consist essentially of a nucleotide sequence that is about 80 percent identical to the nucleotide sequence as shown in SEQ ID NO: 1 or SEQ ID NO: 3, or comprise or consist essentially of a nucleotide sequence encoding a polypeptide that is about 80 percent identical to 5 ihe polypeptide as set forth in SEQ ID NO: 2 or SEQ I) NO: 4. In preferred embodiments, the nucleotide sequences are about 85 percent, or about 90 percent; or about 95 percent, or about 96, 97, 98, or 99 percent identical to the nucleotide sequence as shown in SEQ ID NO: 1 or SEQ ID NO: 3, or the nucleotide sequences encode a polypeptide that is about 85 percent, or about 90 percent, or 10 about 95 percent, or about 96, 97, 98, or 99 percent identical to the polypeptide sequence as set forth in SEQ ID NO: 2 or SEQ ID NO: 4. Related nucleic acid molecules also- include fragments of the above FGF-like nucleic acid molecules which are at least about 16 contiguous nucleotides, or -about 18, or about 20, or about 25, or about 50, or about 75, or about 100, or greater than about 100 1- contiguous nucleotides. Related nucleic acid molecules also include fragments of the above FGF-like nucleic acid molecules which encode a polypeptide of at least about 25 amino acid residues, or about 50, or about 75, or about 100, or greater than about 100 amino acid residues. Related nucleic acid molecules also include a nucleotide sequence 20, encoding a polypeptide as set forth in either SEQ ID NO: 2 or SEQ ID NO: 4 with at least one conservative amino acid substitution and wherein the polypeptide retains at least one FGF-like polypeptide activity, or a nucleotide sequence encoding a polypeptide as set forth in either SEQ ID NO: 2 or SEQ ID NO: 4 with at least one amino acid insertion and wherein the polypeptide retains at least one 25 FGF-like polypeptide activity, or a nucleotide sequence encoding a polypeptide as set-forth in either SEQ ID NO: 2 or SEQ ID NO: 4 with at least one amino acid deletion and wherein the polypeptide retains at least one FGF-like polypeptide activity. Related nucleic acid molecules further include a nucleotide sequence encoding a polypeptide as set forth in either SEQ ID NO: 2 or SEQ ID NO: 4 30 which has a C- and/or N- terminal truncation and wherein the polypeptide retains -13 at least one FGF-like polypeptide activity. Related nucleic acid molecules also include a nucleotide sequence encoding a polypeptide as set forth in either SEQ ID NO: 2 or SEQ ID NO: 4 with combinations of modifications selected from the group consisting of amino acid substitutions, amino acid insertions, amino acid s deletions, C-terminal truncations, and N-teninal truncations and wherein the polypeptide retains at least one FGF-like polypeptide activity. Related FGF-like nucleic acid molecules include,-those molecules that comprise nucleotide sequences which hybridize under moderately or highly stringent conditions as defined herein with the complements of any of the above 10 nucleic acid molecules. In preferred embodiments, the related nucleic acid molecules comprise sequences which hybridize under moderately or highly stringent conditions with the sequence as shown in SEQ ID NO: 1 or SEQ ID NO: 3, or with a molecule encoding a polypeptide, which polypeptide comprises the sequence as shown in SEQ ID NO: 2 or SEQ ID NO: 4, or with a nucleic acid 15 fragment as defined above, or with a nucleic acid fragment encoding a polypeptide as defined above. It is also understood that related nucleic acid molecules include allelic or splice variants of any of the above nucleic acids, and include sequences wbich are complementary to any of the above nucleotide sequences. The term "isolated nucleic acid molecule" refers to a nucleic acid molecule 20 of the invention that is free from at least one contaminting nucleic acid molecule with which it is naturally associated, and preferably substantially free from any Pther contaminating mammalian nucleic acid molecules which would interfere with its use in protein production or its therapeutic or diagnostic use. The term "allelic variant"- refers to one of several possible naturally 25 occurring alternate forms of a gene occupying a given locus on a chromosome of an organism or a population of organisms. The term "splice variant" refers to a nucleic acid molecule, usually RNA, which is generated by alternative. processing of intron sequences in an RNA transcript.
-14 The term "expression vector" refers to a vector that is suitable for propagation in a host cell and contains nucleic acid sequences that direct and/or control the expression of inserted heterologous nucleic acid sequences. Expression includes, but is not limited to, processes such as transcription, 5 translation, and RNA splicing, if introns are present. The term "highly stringency conditions" refers to those conditions that (1) employ low ionic strength reagents and high temperature for washing, for example, 0.015 M NaCI/0.0015 M sodium citrate/0.1% NaDodSO4 (SDS) at 50*C, or (2) employ during hybridization a denaturing agent such as formamide, 10 for example, 50%. (vol/vol) formamide with 0.1% bovine serumn albumin, 0.2% Ficoll, 0.1% polyvinylpyrrolidone, 50 m.M sodium phosphate buffer (pH 6.5), 750 mM NaCl, and 75 mM sodium citrate at 42*C. Another example is the use of 50% formamide, 5X SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyropbosphate, 5X Denhardt's solution, 15 sonicated salmon sperm DNA (50 pg/ml), 0.1% SDS, and 10% dextran sulfate at 42 0 C, with washes at 42*C in O.2X SSC and 0.1% SDS. The term "moderately stringency conditions" refers to conditions which generally include the use of a washing solution and hybridization conditions (e.g., temperature, ionic strength, and percentage of SDS) less stringent than described 20 above. An example of moderately stringent conditions are conditions such as overnight incubation at 37"C in a solution comprising 20% formanide, 5X SSC (150 mM NaC, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5X Deuhardt's solution, 10% dextran sulfate, and 20 d/nl denatured sheared salmon sperm DNA, followed by washing in IX SSC at about 37-50*C. The skilled 25 artisan will recognize how to adjust the temperature, ionic strength, etc. as necessary to accommodate factors such as probe length and the like. In certain prefened embodiments, where oligonucleotide probes are used to screen cDNA or genomic libraries, high stringency conditions are used which depend upon the melting temperature (Tm) of oligonucleotide probes to target -15 sequences. The Tm may be estimated using the following fonnula (Bolton et al., Proc. Na. Acad. Sci. US.A. 48:1390 (1962)): Tm= 81.5 - 16.6 (logfNa+]) + 0.41(%G+C) - (600/N) 5 wherein [Na+] is the sodium ion concentration in the hybridization (or washing) solution; %G+C is gusuine and cytosine content in the oligonucleotide probe; and N is the probe length in nucleo tides. 10 Au example of a high stringency solution is 6X SSC and 0.05% sodium pyrophosphate at a temperature of 35-63"C, depending on the length of the oligonucleotide probe. For example, according to certain embodiments, 14 base pair probes are washed at 35-40*C, 17 base pair probes are washed at 45-50*C, 20 15 base pair probes are washed at 52-57*C, and 23 base pairprobes are washed at 57 63*C. The temperature can be increased 2-3 0 C where the background non specific binding appears high. A second high stringency solution utilizes tetramethylammoninm chloride (TMAC) for washing oligonucleotide probes. One stringent washing solution is 3 M TMAC, 50 mM Tris-HCI, pH 8.0, and 20 0.2% SDS. The washing temperature using this solution is a function of the length of the probe. For example, 14 base pair probes are washed at 35-40"C, 17 - base pair probes are washed at about 45-50"C, 20 base pair probes are washed at 52-57"C, and 23 base pair probes are washed at 57-63*C. The term "FGF-like polypeptides" refers to a polypeptide comprising the 2S amino acid sequence of SEQ ID NO: 2 or SEQ M NO: 4, and related polypeptides described herein. Related polypeptides include: allelic variants; splice variants; fragments; derivatives; substitution, deletion, and insertion variants; fusion polypeptides; and orthologs. FGF-like polypeptides may be mature polypeptides, as defined herein, and may or 'may not have an amino terminal methionine 30 residue, depending on the method by which they are prepared.
-16 The term "FGF-like polypeptide fragment" refers to a peptide or polypeptide that comprises less than the full length amino acid sequence of an FGF-like polypeptide as set forth in SEQ ID NO: 2 or SEQ ID NO: 4. Such a fragment may arise, for example, from a truncation at the amino tenninus, a 5 trMncation at the carboxyl temiinus, and/or an intemal deletion of a residue(s) from the amino acid sequence. FGF-like fragments may result from alternative' RNA splicing or from in vivo protease activity. The term "FGF-like polypeptide variants" refers to FGF-like polypeptides comprising amino acid sequences which contain one or more amino acid sequence 1o substitutions, deletions, and/or additions as compared to the FGF-like polypeptide amino acid sequence set forth in SEQ ID NO: 2 or SEQ ID NO: 4. Variants may be naturally occurring or artificially constructed. Such FGF-like polypeptide variants may be prepared from the corresponding nucleic acid molecules encoding said variants, which have a DNA sequence that varies accordingly from the DNA 15 sequences for wild type FGF-like polypeptides as set forth in SEQ ID NO: 1 or SEQ ID NO: 3. The term 'TGF-like fusion polypeptide" refers to a fusion of an FGF-like polypeptide, fragment, variant, or derivative thereof, with a heterologous'peptide or polypeptide. 20 The term "FGF-like polypeptide derivatives" refers to FGF-like polypeptides, variants, or fragments thereof, that have been chemically modified, as for example, by covalent attachment of one or more polymers, including, but nt limited to, water soluble polymers, N-linked or O-linked. carbohydrates, sugars, phosphates, and/or other such molecules. The derivatives are modified in 25 a manner that is different from naturally occurring FGF-like polypeptide, either in the type or location of the molecules attached to the polypeptide. Derivatives further include the deletion of one or more chemical groups naturally attached to the FOF-like polypeptide. The terms "biologically active FGF-like polypeptides," "biologically 30 active FGF-like polypeptide fragments," "biologically active FGF-like -17 polypeptide variants," and "biologically active FGF-like polypeptide derivatives" refer to FGF-like polypeptides having at least one activity characteristic of an FGF-like polypeptide, such as stimulation of cells within or near the liver, regulation of intestinal cell activity, stimulation of cells within or near pancreatic 5 islets, regulation of neuronal cells, stiulation or inhibition of angiogenesis, stimulation of epithelium or mesenchymal components of grannlation tissue, stimulation of corneal epithelium, lens, or retinal tissue, regeneration of renal tubules hematopoietic cell regulation, regulation of hair follicle growth, regulation of pulmonary pithelium, or stimulation of either epithelial, 10 mesenchymal, hematopoietic, or neuronal cells or tissues. In general, FGF-like polypeptides, and.variants, fragments and derivatives thereof, will have, at least one activity characteristic of an FGF-like polypeptide such as those activities listed above. In addition, an FOP-like polypeptide .may be active as an immunogen (i.e., the polypeptide contains at least one epitope towhicb antibodies iS may be raised). "Naturally occurring" when used in connection with biological materials such as nucleic acid molecules, polypeptides, host cells, and the like, refers to that which are found in nature and not manipulated by a human being. The term "isolated polypeptide" refers to a polypeptide of the invention 20 that is free from at least one contaminating polypeptide that is found in its natural environment and preferably substantially free from any other contaminating mammalian polypeptides which would interfere with its therapeutic or diagnostic use. The term "ortholog" refers. to a polypeptide that corresponds to a 25 polypeptide identified from a different species. For example, marine and human FGF-like polypeptides are considered orthologs of one another. The term "mature FGF-like polypeptide" refers to a polypeptide lacking a leader sequence and mayalso include other modifications of a polypeptide such as proteolytic processing of the amino terminus (with or without a leader sequence) 30 and/or the carboxyl terminus, cleavage of a smaller polypeptide from a larger - 18 precursor, N-linked and/or O-linked glycosylation, and other post-translational modifications understood by those with skill in the art. The terms "effective amount" and "therapentically effective amount" refer to the amount of a FGF-like polypeptide that is useful or necessary to support an 5 observable level of one or more biological activities of the FGF-like polypeptides as set forth above. Relatedness ofNucleic Acid Molecules and/or Polypeptides The term "identity," as known in the art, refers to a relationship between 10 the sequences of two or more polypeptide molecules or two or more nucleic acid molecules, as determined by comparing the sequences. In the art, "identity".also means the degree of sequence relatedness between polypeptide .or nucleic acid molecule sequences, as the case may be, as determined by the match between strings of nucleotide or aino acid sequences. "Identity" measures the percent of 15 identical matches between two or more sequences with gap alignments addressed by a particular mathematical model of computer programs (i e., "algorithms"). The term "similarity" is a related concept, but in contrast to 'identity," refers to a measure of similarity which includes both identical matches and conservative substitution matches. Since conservative substitutions apply to 20 polypeptides and not nucleic acid molecules, similarity only deals with polypeptidb sequence comparisons. If two polypeptide sequences have, for example, 10 out of 20 identical amino acids, and the remainder are all non conservative substitutions, then the percent identity and similarity would both be 50%. If in the same example, there are 5 more positions where there are 25 conservative substitutions, then the percent identity remains 50%, but the percent similarity would be 75% (15 out of 20). Therefore, in cases where there are conservative substitutions, the degree of similarity between two polypeptide sequences will be higher than the percent identity between those two sequences. The term "conservative amino acid substitution" refers to a substitution of 30 a native amino acid residue with a nonnative residue such that there is little or no - 19 effect on the polarity or charge of the amino acid residue at that position. For example, a conservative substitution results from the replacement of a non-polar residue in a polypeptide with any other non-polar residue. Furthermore, any native residue in the polypeptide may also be substituted with alanine, as has been 5 previously described for "alanine scanning mutagenesis" (Cunnigham et al., Science 244:1081-85 (1989)). General rules for conservative amino acid substitutions are set forth in Table . Table.I 10 Conservative Amino Acid Substitutions Original Residues Exemplary Substituti6ns Preferred Substitutions Ala Val, Leu, le Val Arg Lys, Gn. Asn Lys Asn Gin, His, Lys. Arg Gln Asp Glu Gin Cys Ser Ser Gln Asn Asn Gin Asp Asp Gly Pro, Ala Ala His Asn, Gln, Lys, Arg Arg Ile Leu, Val, Met, Ala, Leu Phe, Norleucine Leu Norleucine, le,. Ile Val, Met, Ala, Phe Lys Ax Gln, Asn Arg Met Leu, Phe, Ile Leu Phe Leu, Val, lie, Ala, Leu Tyr Pro Ala Ala - 20 Ser Thr Thr Tir Ser Ser Trp Tyr, Phe Tyr Tyr Trp, Phe, Thr, Ser Phe Val Ile, Met, Leu, Phe, Leu Ala,.Norleucine Conservative amino acid substitutions also encompass non-naturally occurring amino acid residues that are typically incorporated by chemical peptide synthesis rather than by synthesis in biological systems. These include peptidomimetics, 5 and other reversed or inverted forms of amino acid moieties. Conservative , modifications to the amino acid sequence (and the corresponding modifications to the encoding nucleotides) are expected to produce FGF-like polypeptide having functional and chemical characteristics similar to those of naturally occurring FGF-like polypeptide. In contrast, substantial 10 modifications in the functional and/or chemical characteristics of FGF-like polypeptide may be' accomplished by selecting substitutions that differ significantly in their effect on maintaining (a) the structure of the molecular backbone in the area of the substitution, for example, as a sheet or helical conformation, (b) the charge or hydrophobicity of the molecule at the target site, 15 or (c) the bulk of the side chain. Naturally occurring residues may be divided into groups based on common side chain properties: 1), hydrophobic: norleucine, Met, Ala, Val, Len, lle; 2) neutral hydrophilic: Cys, Ser, Thr; 3) acidic: Asp, Glu; 20 4) basic: Asn, Gin, His Lys, Arg; 5) residues that influence chain orientation: Gly, Pro; and 6) aromatic: Trp, Tyr, Phe. Non-conservative substitutions may involve the exchange of a member of one of these classes for a member from another class. Such substituted residues -21 may be introduced into regions of the human FGF-like molecule that are homologous with non-human FGF-like polypeptide, or into the non-homologous regions of the molecule. Identity and similarity of related nucleic acid molecules and polypeptides 5 can be readily calculated by known methods, including but not limited to those described in Computational Molecular Biology (A.M. Lesk, ed., Oxford University Press 1988); Biocomputing: informatics and Genome Projects (D.W. Smith, ed., Academic Press 1993); Computer Analysis of Sequence Data (Part 1, A.M. Griffin and H.G. Griffin, eds., Humana Press 1994); G. von Heinle, 10 Sequence Analysis in Molecular Biology (Academic Press 1987); Sequence Analysis Primer (M. Gribskov and 1. Devereux, eds., M. Stockton Press 1991); and Carillo et al., SIAMJ. Applied Math. 48:1073 (1988). Preferred methods to determine identity and/or similarity are designed to give the largest match between the sequences tested. Methods to determine 15' identity and similarity are codified in publicly available computer programs. Preferred computer program methods to determine identity and similarity between two sequences include, but are not limited to, the GCG program package, including GAP (Devereux et al., Nuc. Acids Res. 12:387 (1984); Genetics Computer Group, University of Wisconsin, Madison, W1), BLASTP, BLASTN, 20 and FASTA (Atschnl et al., J. Mol. Biol. 215:403-10 (1990)). The BLAST X program is publicly. available from the National Center for Biotechnology Information (NCB) and other sources (Altschul et al., BLAST Manual (NCB NLM NIH, Bethesda, MD); Altschul et al., 1990; supra). The well-known Smith Waterman algorithm may also be used to determine identity. 25 By way of example, using the computer algorithm GAP (Genetics Computer Group), two polypeptides for which the percent sequence identity is to be determined are aligned for optimal matching of their respective amino acids (the "matched span," as determined by the algorithm). A gap opening penalty (which is calculated as 3X the average diagonal; the "average diagonal" is the 30 average of the diagonal of the comparison matrix being used; the "diagonal" is the -22 score or number assigned to each perfect amino acid match by the particular comparison matrix) and a gap extension penalty (which is usually O.IX the gap opening penalty), as well as a comparison matrix such as PAM 250 or BLOSUM 62 are used in conjunction with the algorithm. A standard comparison matrix (see 5 Dayhoff et al,, 5 Atlas of Protein Sequence and Structure (Supp. 3 1978) for the PAM250 comparison matrix; see Henikoff et al, Proc. Natl. Acad, Sd USA 89:10915-19 (1992) for the BLOSUIM 62 comparison matrix) is also used by the , algorithm. Preferred parameters -for polypeptide sequence comparison include the 10 following: Algorithm: Needleman and Wunsch, J Mol. Biol. 48:443-53 (1970) Comparison matrix: BLOSUM 62 from Henikoff et al., Proc. NatL. Acad. Sc. U.S.A. 89:10915-19 (1992) 15 Gap Penalty: 12 Gap Length Penalty: 4 Threshold of Similarity: 0 The GAP program is useful with the above parameter. The aforementioned 20 parameters are the default parameters for polypeptide comparisons (along with no penalty for end gaps) using the GAP algorithm. Preferred parameters for nucleic acid molecule sequence comparison include the following: 25 Algorithm: Needleman et al., . Mol BioL 48:443-53 (1970) Comparison matrix: matches = +10, mismatch = 0 Gap Penalty: 50 Gap Length Penalty. 3 -23 The GAP program is also useful with the above parameters. The aforementioned parameters are the default parameters for nucleic acid molecule comparisons. Other exemplary algorithms, gap opening penalties, gap extension penalties, comparison matrices, thresholds of similarity, etc. inay'be used by those 5 of skill in the art, including those set forth in the Program Manual, Wisconsin Package, Version 9, September, 1997. The particular choices to be made will depend on the specific comparison to be made, such as DNA to DNA, protein to protein, protein to DNA; and additionally, whether the comparison is between given pairs of sequences Cm which case GAP or BestFit are generally preferred) or 10 between one sequence and a large database of sequences (in which case FASTA or BLASTA are preferred). Sequence analysis of an isolated mouse cDNA (murine FGF-like protein; SEQ ID NO: 3) indicated that it encoded a novel member of the FGF family of proteins. The muTine FGF-like gene comprises a 630 bp open reading frame is encoding a protein of 210 amino acids (Figure 1). The murine'sequence was used to identify the human'i FGF-like ortholog. Sequence analysis of four human FGF like polypeptide cDNA clones indicated that the human FGF-like gene comprises a 627 bp open reading frame encoding a protein of 209 amino acids (Figures 2A 2B). 20 Figures 3A-3D illustrate the aniino acid sequence alignment of human FGF-like protein, murine FGF-like protein, and other members of the FGF family. Computer analysis of the predicted inurine FGF-like polypeptide, using the FASTA program of the Swissprot database, indicated that the protein was most closely related to murine FGF-6, FGF-15, and FGF-4. Using the GAP program, 25 murine FGF-like polypeptide was fund to be 32% identical to urine FGF-6 and 28% identical to marine FGF-4. Computer analysis also indicated that the urine FGF-like polypeptide, similar to FGF-6, FGF-4, and FGF-15 but in contrast to FGF-i and FGF-2j possessed a potential signal peptide at its amino terminus. The ihurine FGF-like polypeptide is 79% identical to the human FGF-like protein. 30 -24 Nicleic Acid Molecules Recombinant DNA methods used herein are generally those set forth in Sambrook et al., Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Laboratory Press, 1989) and/or Current Protocols in Molecular Biology (Ausubel 5 et al., eds., Green Publishers Inc. and Wiley and Sons 1994). The invention provides for nucleic acid molecules as described herein and methods for obtaining -the molecules. A gene or cDNA encoding an FGF-like polypeptide or fragment thereof may be obtained by hybridization screening of a. genomic or cDNA library, or by PCR amplification. Probes or primers useful for 10 screening a library by hybridization can be generated based on sequence information for other known genes or gene fragments from the same or a related family.of genes, such as, for example, conserved motifs. In addition, where a gene encoding FGF-like polypeptide has been identified from one species, all or a portion of that gene may be used as a probe to identify corresponding genes from 15 other species (orthologs) or related genes from the same species (homologs). The probes or primers may be used to screen cDNA libraries from various tissue sources believed to express the FGF-like gene. In addition, part or all of a nucleic acid molecule having the sequence as set forth in SEQ ID NO: 1 or SEQ ID NO: 3 may be used to screen a genomic library to identify and isolate a gene encoding 20 FGF-like polypeptide. Typically, conditions of moderate or high stringency will be employed for screening to minimize the number of false positives obtained from the screen. Nucleic acid molecules encoding FGF-like polypeptides may also be identified by expression cloning which employs detection of positive clones based 25 upon a property of the expressed protein. Typically, nucleic acid libraries are screened by binding an antibody or other binding partner (e.g., receptor or ligand) to cloned proteins which are expressed and displayed on the host cell surface. The antibody or binding partner is modified with a detectable label to identify those cells expressing the desired clone.
-25 Another means of preparing a nucleic acid molecule encoding an FGF-like polypeptide ot fragment thereof is chemical synthesis using methods well known to the skilled artisan such as those described by Engels et al., Angew. Chem. Intl, Ed. 28:716-34 (1989). These methods include, inter alia, the phosphotriester, 5 phosphoramidite, and H-phosphonate methods for nucleic acid synthesis. A preferred method for such chemical synthesis is polymer-supported synthesis using standard phosphoramidite chemistry. Typically, the DNA encoding the FGF-like polypeptide will be several hundred nucleotides in length. Nucleic acids larger than about 100 nucleotides can be synthesized as several fragments using 10 these methods. The fragments can then be ligated together to form the full-length FGF-like polypeptide. Usually, the DNA fragment encoding the amino terminus of the polypeptide will have an ATG, which encodes a methionine residue. This methionine may or may not be present on the mature form of the FGF-like polypeptide, depending on whether the polypeptide produced in the host cell is 15 designed to be secreted from that cell. In some cases, it may be desirable to prepare nucleic acid molecules encoding FGF-like polypeptide variants. Nucleic acid molecules encoding variants may be produced using site directed mutagenesis, PCR amplification, or other appropriate methods, where the primer(s) have the desired point mutations 20 (see Sambrook et al., supra, and Ausubel et al., supra, for descriptions of mutagenesis techniques). Chemical synthesis using methods described by Engels et at, supra, may also be used to prepare such variants. Other methods known to the skilled artisan may be used as well. In certain embodiments, nucleic acid variants contain codons which have 25 been altered for optimal expression of an FGF-like polypeptide in a given host cell. Particular codon alterations will depend upon the FOE-like polypeptide and host cell selected for expression. Such "codon optimization" can be carried out by a variety of methods, for example, by selecting codons which are preferred for use m highly expressed genes in a given host cell. Computer algorithms which 30 incorporate codon frequency tables such as "Ecohigh.Cod" for codon preference -26 of highly expressed bacterial genes may be used and are provided by the University of Wisconsin Package Version 9,0, Genetics Computer Group, Madison, WL Other useful codon frequency tables include "Celeganshigh.cod;" "Celegans-low.cod," "Drosophila high.cod,' "Humanhigh.cod," 5 "Maize high,cod," and "Yeasthigh.cod." In other embodiments, nucleic acid molecules encode FGF-like variants with conservative amino acid substitutions as defined above, FGF-like variants comprising an addition and/or a deletion of one or more N-linked or 0-linked glycosylation sites, FGF-like variants having deletions and/or substitutions of one 10 or more cysteine residues, or FGF-like polypeptide fragments as described above, .n addition, nucleic acid molecules may encode any combination of FGF-like variants, fragments, and fusion polypeptides described herein. Vectois and Host Cells J.5 A nucleic acid molecule encoding am FGF-like polypeptide is inserted into an appropriate expression vector using standard ligation techniques. The vector is typically selected to be functional in the particular host cell employed (i.e., the vector is compatible with the host cell machinery such that amplification of the gene and/or expression of the gene can occur). A nucleic acid molecule encoding 20 an FGF-like polypeptide may be amplified/expressed in prokaryotic, yeast, insect (baculovirms systems) and/or eukaryotic host cells. Selection of the host cell will depend in part on whether an FGF-like polypeptide is to be post-translationally modified (eg., glycosylated and/or phosphorylated). If so, yeast, insect, or mammalian host cells are preferable. For a review of expression vectors, see 185 25 Meth. Enz. (D.V. Goeddel, ed., Academic Press 1990). Typically, expression vectors used in any of the host cells will contain sequences for plasmid maintenance and for cloning and expression of exogenous nucleotide sequences. Such sequences, collectively referred to as "flanking sequences" in certain embodiments will typically include one or more of the 30 following nucleotides: a promoter, one or niore enhancer sequences, an origin of -27 replication, a transcriptional termination sequence, a complete itron sequence containing a donor and acceptor splice site, a leader sequence for secretion, a ribosome binding site, a polyadenylation sequence, a polylinker region for inserting the nucleic acid encoding the polypeptide to be expressed, and a 5 selectable marker element. Each of these sequences is discussed below. Optionally, the vector may contain a "tag" sequence, L., an oligonucleotide molecule located at the S' or 3' end of the FGF-like polypeptide coding sequence; the oligonucleotide molecule encodes polyHis (such as hexaHis), or other "tag" such as FLAG, HA (hemaglutinin Infuenza virus) or myc 10 for which commercially available antibodies exist, This tag is typically fused to the polypeptide upon expression of the polypeptide, and can serve as a means for affinity purification of the FGF-like polypeptide from the host cell. Affinity purification can be accomplished, for example, by colunm chromatography using antibodies against the tag as an affinity matrix. Optionally, the tag can 1s subsequently be removed from the purified FGF-like polypeptide by various means such as using certain peptidases for cleavage. Flanking sequences may be homologous (i.e., from the same species and/or strain as the host cell), heterologous (i.e., from a species other than the host cell species or strain), hybrid (L.e., a combination of flanking sequences from more 20 than one source), or synthetic, or native, sequences which normally function to regulate PGF-like expression. As such, the source of flanking sequences may be any prokaryotic or eukaryotic organism, any vertebrate or invertebrate organism, or any plant provided that the flanking sequences is functional in, and can be activated by, the host cell machinery. 25 The fiauking sequences useful in the vectors of this invention may be obtained by any of several methods well known. in the art. Typically, flanking sequences useful herein other than the FGF-like gene flanking sequences will have been previously identified by mapping and/or by restriction endonuclease digestion and can thus be isolated from the- proper tissue source using the 30 appropriate restriction endonucleases. In some cases, the full nucleotide sequence .- 28 of one or more flanking sequence may be known. Here, the flanking sequence may be synthesized using the methods described above for nucleic acid synthesis or cloning. Where. all or only a portion of the flanking sequence is known, it may be 5 obtained -using PCR and/or by screening a genomic library with suitable oligonucleotide and/or flanking sequence fragments from the same or another species. Where the flanking sequence is not known, a fragment of DNA containing a flanking sequence may be isolated from a larger piece of DNA that may contain, 10 for example, a coding sequence or even another gene 0'r genes. Isolation may be accomplished by.restriction endonuclease digestion to produce the proper DNA fragment followed by isolation using agarose gel purification, Qiagen (Valencia, CA) column chromatography, or other methods known to the skilled artisan. Selection of suitable enzymes to accomplish this purpose will be readily apparent 15 to one of ordinary skill in the art. An origin- of replication is typically a part of prokaryotic expression vectors purchased commercially, and aids in the amplification of the vector in a host cell. Amplification of the vector to a certain copy number can, in some cases, be important for optimal expression of the FGF-like polypeptide. It the 20 vector of choice does not contain an origin of replication site, one may be chemically synthesized based on a known sequence, and ligated into the vector. The origin of replication from the plasmid pBR322 (Product No. 303-3s, New England Biolabs, Beverly, MA) is suitable for most Gram-negative bacteria and various origins (e.g., SV40, polyoma, adenovirs, vesicular stomatitus virus 25 (VSV) or papillornaviruses such as HPV or BPV) are useful for cloning vectors in mammalian cells. Generally, the origin of replication component is not needed for mammalian expression vectors (for example, the SV40 origin is often used only because it contains the early promoter). A transcription termination sequence is typically located 3' of the end of a 30 polypeptide coding regions and serves to tenninate transcription. Usually, a - 29 transcription termination sequence in prokaryotic cells is a G-C rich fragment followed by a poly-T sequexice. While the sequence is easily cloned from a library or even purchased commercially as part of a vector, it can also be readily synthesized using methods for nucleic acid synthesis such as those described 5 above. A selectable marker gene element encodes a protein necessary for the survival and growth of a host cell grown in a selective culture medium. Typical selection marker genes encode proteins that (a) confer resistance to antibiotics or other toxins, for example, ampicillin, tetracycline, or kanamycinfor prokaryotic io host cells, (b) complement auxotrophic deficiencies of the cell; or (c) supply critical nutrients not available from.complex media. Preferred selectable markers are the kanamycin resistance gene, the ampicillin resistance gene, and the tetracycline resistance gene. A neomycin resistance gene may also be used for selection in prokaryotic and eukaryotic host cells. is Other selection genes may be used .to amplify the gene that will be expressed. Amplification is the process wherein genes that are in greater demand for the production of a protein critical for growth are reiterated in tandem within the chromosomes of successive generations of recombinant cells. Examples of suitable selectable markers for mammalian cells include dihydrofolate reductase 20 (DHFR) and thynildine kinase. The.mammalian cell transfornants are placed under selection pressure that only the transformants are uniquely adapted to survive by virtue of the marker present in the vector. Selection pressure is imposed by culturing the transformed cells under conditions in which the concentration of selection agent in the medium is successively changed, thereby 25 leading to amplification of both the selection gene and the DNA that encodes FGF-like polypeptide. As a result, increased quantities of FGF-like polypeptide are synthesized from the amplified DNA. A ribosome binding site is usually necessary for translation initiation of mRNA and is characterized by a Shine-Dalgarno sequence (prokaryotes) or a 30 Kozak sequence (eukaryotes). The element is typically located 3' to the promoter - 30 and 5' to the coding sequence of the FGF-like polypeptide to be expressed. The Shine-Dalgarno sequence is varied but is typically a polypurine (i.e., having a high A-G content). Many Shine-Dalgarno sequences have been identified, each of which can be readily synthesized using methods set forth above and used in a 5 prokaryotic vector. A leader, or signal, sequence. may be used to direct an FGF-like polypeptide out of the host cell. Typically, the signal sequence is positioned in the coding region of the FGF-like nucleic acid molecule, or directly at the 5' end of the FGF-like polypeptide coding region. Many signal sequences have been 10 identified, and any of them that are functional in the selected host cell may be used in conjunction with the FGF-ike gene or eDNA. Therefore, a signal sequence may be homologous (naturally occurring) or heterologous to the FGF like gene or cDNA, Additionally, a signal sequence may be chemically synthesized using methods set forth above. In most cases, secretion of an FGF 15 like polypeptide from the host cell via the presence of a signal peptide will result in the removal of the signal peptide from the FGF-like polypeptide. The signal sequence may be a component of the vector, or it may be a part of FGF-like DNA that is inserted into the vector. Included within the scope of this invention is the native FGF-like signal 20 sequence joined to an FGF-like coding region and a heterologous signal sequence joined to an FGF-like coding region. The heterologous signal sequence selected should be one that is recognized and processed, i.e., cleaved by a signal peptidase, by the host cell. For prokaryotic host cells that do not recognize and process the native FGF-like signal sequence, the signal sequence is substituted by a 25 prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, or heat-stable enterotoxin II leaders. For yeast secretion, the native FGF-like signal sequence may be substituted by the yeast invertase, alpha factor, or acid phosphatase leaders. In mammalian cell expression the native signal sequence is satisfactory, although other mammalian signal 30 sequences may be suitable.
-31 In some cases, such as where glycosylation is desired in a eukaryotic host cell expression system, one may manipulate the various presequences to improve glycosylation'or yield. For example, one may alter the peptidase cleavage site of a particular signal peptide, or add prosequences, which also may affect 5 glycosylation. The final protein product may have, in the -1 position (relative to the first amino acid of the mature protein) one or more additional amino acids incident to expression, which may not have been totally removed. For example, the final protein product may have one or two amino acid found in the peptidase cleavage site, attached to the N-terminus. Alternatively, use of some enzyme 10 cleavage sites may result in a slightly truncated form of the desired FOE-like polypeptide, if the enzyme cuts at such area within the mature polypeptide. In many cases, transcription of a nucleic acid molecule is increased by the presence of one or more introns in the vector; this is particularly true where a polypeptide is produced in eukaryotic host cells, especially mammalian host cells. 15 The introns used may be naturally occurring within the FGF-like gene especially where the gene used is a full-length genomic sequence or a fragment thereof. Where the intron is not naturally occurring within the gene (as for most cDNAs), the intron may be obtained from another source. The position of the intron with respect to flanking sequences and the FGF-like gene is generally important, as the 20 intron must be transcribed to be effective. Thus, when an FGF-like cDNA molecule is being expressed, the preferred position for the intron is 3' to the transcription start site and 5' to the poly-A transcription termination sequence. Preferably, the intron or introns will be located on one side or the other (i.e., 5' or 3') of the cDNA such that it does not interrupt the coding sequence. Any intron 25 from any source, including any viral, prokaryotic and eukaryotic (plant or animal) organisms, may be used to practice this invention, provided that it is compatible with the host cell into which it is inserted. Also included herein are synthetic introns. Optionally, more than one intron may be used in the vector. The expression and cloning vectors of the present invention will typically 30 contain a promoter that is recognized by the host organism and operably linked to -32 the molecule encoding the FGF-like protein. Promoters are untranslated sequences located upstream (ia, 5') to the start codon of a structural gene (generally within about 100 to 1000 bp) that control the transcription and translation of the structural gene. Promoters are conventionally grouped into one 5 of two classes: inducible promoters and constitutive jiromoters. Inducible promoters initiate increased levels of transcription from DNA under their control in response to some change in culture conditions such as the presence or absence of a nutrient or a change in temperature. A large number of promoters, recognized by a variety of potential host cells, are well known. These promoters are operably 1.0 linked to the DNA encoding FGF-like polypeptide by removing the promoter from the source DNA by restriction enzyme digestion and inserting the desired promoter sequence into the vector. The native FGF-like promoter sequence may be used to direct amplification and/or expression of FGF-like DNA. A heterologous promoter is preferred, however, if it permits greater transcription and is higher yields of the expressed protein as compared to the native promoter, and if it is compatible with the host cell system that has been selected for use. Promoters suitable for use with prokaryotic hosts include the beta lactamase and lactose promoter systems; alkaline phosphatase, a tryptophan (trp) promoter system; and hybrid promoters such as the tac promoter. Other known 20 bacterial promoters are also suitable. Their sequences have been published, thereby enabling one skilled in the art to ligate them to the desired DNA sequence, using linkers or adapters as needed to supply any required restriction sites. Suitable promoters for use with yeast hosts are also well known in the art. Yeast enhancers are advantageously used with yeast promoters. Suitable 25 promoters for use with mammalian host cells are well known and include those obtained from the genomes of viruses such as polyoma virus, fowlpox virms, adenovirus (such as Adenovirus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and most preferably Simian Vims 40 (SV40). Other suitable mammalian promoters include heterologous 3 o mammalian promoters, for example, heat-shock promoters and the actin promoter.
-33 Additional promoters which may be of interest in controlling FGF-like gene expression include, but are not limited to: the SV40 early promoter region (Bemoist and Chambon, Nature 290:304-10 (1981)); the CMV promoter, the promoter contained in the 3' long terminal repeat of Rous sarcoma virus 5 (Yamamoto, et aL, Cell 22:787-97,(1980)); the herpes thymidine kinase promoter (Wagner et al., Proc NatL. Acad. Sc US.A. 78:1444-45- (1981)); the regulatory' sequences of the metallothionine gene (Brinster et al., Nature 296:39-42 (1982)); prokaryotic expression vectors such as the beta-lactamase promoter (Villa Kamaroff et al., Proc. Nat. Acad. Sci. U.SA, 75:3727-31 (1978)); or the tac 10 promoter (DeBoer et al., Proc NatL Acad Sci, U.SA, 80:21-25 (1983)). Also of interest are the following animal transcriptional control regions, which exhibit tissue specificity and have been utilized. in transgenic animals: the elastase I gene control region which is active in pancreatic acinar cells (Swift et al., Cell 38:639 46 (1984); Omitz et alt, Cold Spring Harbor Symp. Quant. Biol 50:399-409 15 (1986); MacDonald,- Repatology 7:425-515 (1987)); the insulin gene control region which is active in pancreatic beta cells (Hanahan, Nature 315:115-22 (1985)); the immunoglobulin gene control region which is active in lymphoid cells (Grosschedl et al., Cell 38:647-58 (1984); Adames et al., Nature 318:533-38 (1985); Alexander et al., MoL Cell. BiA, 7:1436-44 (1987)); the mouse mammary 20 tumor virus control region which is active in testicular, breast, lymphoid and mast cells (Leder et al., Cell 45:485-95 (1986)); the albumin gene control region which is Active in liver (Pinkert et al., Genes and Devel 1:268-76 (1987)); the alpha-feto protein gene control region which is active in liver (Krumlanf et al., Ma. Cell. Biol., 5:1639-48 (1985); Hammer et al., Science 235:53-58 (1987)); the alpha 1 25 antitrypsin gene control region which is active in the liver (Kelsey et al., Genes and Devel 1;161-71, 1987)); the beta-globin gene control region which is active in mycloid cells (Mogram et al., Nature 315:338-40 (1985); Kollias et al, Cell 46:89-94 (1986)); the myelin basic protein gene control region which is active in oligodendrocyte cells in the brain (Readhead et A, Cell 48:703-12 (1987)); the 30 myosin light chain-2 gene control region which is active in skeletal muscle (Sani, -34 Nature 314:283-86 (1985)); and the gonadotropic releasing hormone gene control region which is active in, the hypothalamus (Mason et al., Science 234:1372-78 (1986)). An enhancer sequence may be inserted into the vector to itLcrease the s transcription of a DNA encoding an FGF-like protein of the present invention by higher eukaryotes. Enhancers are cis-acting elements of DNA, usually about 10 300 bp in length, that act on the promoter to increase its transcription. Enhancers are relatively orientation and position independent. They have been found 5' and 3' to the transcription unit. Several enhancer sequences available from 10 mammalian genes are known (e.g., globin, elastase, albumin, alpha-feto-protein and insulin). Typically, however, an enhancer from a virus will be used. The SV40 enhancer, the cytomegalovirus early promoter enhancer, the polyoma enhancer, and adenovirus enhancers are exemplay enhancing elements for the activation of eukaryotic promoters. While an enhancer may be spliced into the 15 vector at a position 5t or 3' to FGF-like DNA, it is typically located at a site 5' from the promoter. Expression vectors of the invention may be constructed from starting vectors such as a commercially available vector. Such vectors may or may not contain all of the desired flanking sequences. Where one or more of the flanking 20 sequences set forth above are not already present in the vector to be used, they may be individually obtained and ligated into the vector. Methods used for obtaining each of the flanking sequences are well known to one skilled in the art. Preferred vectors for practicing this invention are those which are compatible with bacterial, insect, and mammalian host cells. Such vectors 25 include, inter alia, pCRI pCR3, and pcDNA3.1 (fnvitrogen, San Diego, CA), pBSII (Stratagene, La Jolla, CA), pET15 (Novagen, Madison, WI), pGEX (Phannacia Biotech, Piscataway, NJ), pEGFP-N2 (Clontech, Palo Alto, CA), pETL (BlueBacI, Invitrogen), pDSR-alpha (PCT Pub. No. WO 90/14363) and pFastBacDual (Gibco-BRL, Grand Island, NY). 30 Additional possible vectors include, but are not limited to, cosmids, - 35 plasmids, or modified viruses, but the vector system must be compatible with the selected host cell. Such vectors include, but are not limited to plasmids such as Bluescripte plasmid derivatives (a high copy number CoIEl-based phagemid, Stratagene Cloning Systems, La Jolla CA), PCR cloning plasmids designed for 5 cloning Taq-amplified PCR products (e.g., TOPOTm TA Cloninge Kit, PCR2.1 plasmid derivatives, Invitrogen, Carlsbad, CA), and mammalian, yeast or virus vectors such as a baculovirus expression system (pBacPAK plasmid derivatives, Clontech, Palo Alto, CA).. The recombinant molecules can be introduced into host cells via transformation, transfection, infection, electroporation, or other known .10 techniques. After the vector has been constructed and a nucleic acid molecule encoding an FGF-like polypeptide has been inserted into the proper site of the vector, the completed. vector may be inserted into a suitable host cell for amplification and/or polypeptide expression. -5 Host cells may be prokaryotic host cells (such as E. coli) or eukaryotic host cells (such as a yeast cell, an insect cell, or a vertebrate cell). The host cell, when cultured under appropriate conditions, synthesizes an FGF-like polypeptide which can subsequently be collected from the culture medium (if the host cell secretes it into the medium) or directly from the host cell producing it (if it is not . 20 secreted). Selection of an appropriate host cell will depend upon various factors, such as desired expression levels, polypeptide modifications that are desirable or - necessary for activity, such as glycosylation or phosphorylation, and ease of folding into a biologically active molecule. A number of suitable host cells are known in the art and many are 25 available from the American Type Culture Collection (ATCC), Manassas,'VA. Examples include mammalian cells, such as Chinese hamster ovary cells (CHO), CHO DHFR- cells (Urlaub et al., Proc. NatiL. Acad. Sci US.A, 97:4216-20 (1980)), human embryonic kidney (HEK) 293 or 293T cells, or 3T3 cells. The selection of suitable' mammalian host cells and methods for transformation, 3o culture, amplification, screening, product production and purification are known - 36 in the art. Other suitable manunalian cell lines, are the monkey COS-1 and COS 7 cell lines, and the CV-.1 cell line. Further exemplary mammalian host cells include primate cell lines and rodent cell lines, including transformed cell lines. Normal diploid cells, cell strains derived from in vitro culture of primary tissue, as s .well as primary explants, are also suitable. Candidate cells may be genotypically deficient in the selection gene, or may contain a dominantly acting selection gene. Other suitable mammalian dell lines include but are not limited to, mouse neuroblastoma N2A cells, HeLa, mouse L-929 cells, 3T3 lines derived from Swiss, Balb-c or NIH mice, BHK or HaK hamster cell lines. Each of these cell 10 lines is known by alid available to those skilled in the at of protein expression Similarly useful as. host cells suitable for the present invention are' bacterial cells. For example, the various strains of E. coli (e.g., HB101, DR5, DH10, and MC1061) are well known as host cells in the field of biotechnology. Various strains of B. subtilis, Pseudomonas spp., other Bacillus spp., 15 Streptomyces spp., and the like may also be employed in this method. Many strains of yeast cells known to those skilled in the art are also available as host cells for expression of the polypeptides of the present invention. Preferred yeast cells include, for example, Saccharonyces cerivisae. Additionally, where desired, insect cell systems may be utilized in the 20 methods of the present invention.~ Such systems are described, for example, in Kitts et al., Biotechniques, 14:810-17 (1993); Lucklow, Czar. Opin. Biotechnol. 4:564-72 (1993); and Lucklow et al, J. Virol., 67:4566-79 (1993). Preferred insect cells are Sf-9 and HiS (Invitrogen). Transformation or transfection of an expression vector for an FGF-like 25 polypeptide into a selected host cell may be accomplished by well known methods including methods such as calcium chloride, electroporation, microinjection, lipofection or the DEAE-dextran method. The method selected will in part be a function of the type of host cell to be used. . These methods and other suitable methods are well known to the skilled artisan, and are set forth, for example, in 30 Sambrook et al., supra.
- 37 One may also use transgenic animals to express glycosylated FGF-like polypeptides. For example, one may use a transgenic milk-producing animal (a cow or goat, for example) and obtain the present glycosylated polypeptide in the animal milk. One may also use plants to produce FGF-like polypeptides, 5 however, in general, the glycosylation occurring in plants is different from that produced in mammalian cells, and may result in a glycosylated product which is not suitable for human therapeutic use, Polypeptide Production 10 Host cells comprising an FGF-like polypeptide expression vector (i.e., transformed or transfected) may be cultured using standard media well known to the skilled artisan The media will usually contain all nutrients necessary for the growth and survival of the cells. Suitable media for culturing E. coli cells are for example, Luria Broth (1B) and/or Terrifie Broth (TB)-. Suitable media for 15 culturing eukaryotic cells are RPM[ 1640, MEM, DMEM, all of which may be supplemented with serum and/or growth factors as required by the particular cell line being cultured. A suitable medium for insect cultures is Grace's medium supplemented with yeastolate, lactalbumin hydrolysate, and/or fetal calf serum as necessary. 20 Typically, an antibiotic or other compound useful for selective growth of transfected or transformed cells is added as a supplement to the media. The compound to be used will be dictated by the selectable marker element present on the plasmid with which the host cell was transformed. For example, where the selectable marker element is kanamycin resistance, the compound added to the 25 culture medium will be kanamycin. Other compounds for selective. growth include ampicillin, tetracycline, and neomycin. The amount of an FGF-like polypeptide produced by a host cell can be evaluated using standard methods known in the art. Such methods include, without limitation, Westem blot analysis, SDS-polyacrylamide gel -38 electrophoresis, non-denaturing gel electrophoresis, HPLC separation, immunoprecipitation, and/or activity assays such as DNA binding gel shift assays. If an FGF-like polypeptide has been designed to be secreted flom the host cells, the majority of polypeptide may be found in the cell culture medium. If 5 however, the FGF-like polypeptide is not secreted from the host cells, it will be present in the cytoplasm and/or the nucleus (for eukaryotic host cells) or in the cytosol (for gram-negative bacteria host cells). For an FGF-like polypeptide situated in the host cell cytoplasm and/or nucleus, the host cells are typically first disrupted mechanically or with detergent 10 to release the intra-cellular contents into a buffered solution. FGF-like polypeptide can then be isolated from this solution. Purification of an FGF-like polypeptide from solution can be accomplished using a variety of techniques. If the polypeptide has been synthesized such that it contains a tag such as Hexahistidine (FGF-like is polypeptide/hexaHis) or other small peptide such as FLAG (Eastman Kodak Co., New Haven, CT) or myc (Invitrogen) at either its carboxyl or amino teninus, it may essentially be purified in a one-step process by passing the solution through an affinity column where the column matrix has a high affinity for the tag or for the polypeptide directly (Le., a monoclonal antibody specifically recognizing 20 FOF-like polypeptide). For example, polyhistidine binds with great affinity and specificity to nickel and thus an affinity column of nickel (such as the Qiagen nickel columns) can be used for purification of FGF-like polypeptide/polyHis. See, e.g., Current Protocols in Molecdar Biology § 10.11.8 (Ausubel et al., eds., John Wiley & Sons 1993). 25 Where an FGF-like polypeptide is prep ared without a tag attached, and no antibodies are available, other well-known procedures for purification can be used. Such procedures include, without limitation, ion exchange chromatography, molecular sieve chromatography, HPLC, native gel electrophoresis in combination with gel elution, and preparative isoelectric focusing ("Isoprime" -39 machine/technique, Hoefer Scientific). In some cases, two or more of these techniques maybe combined to achieve increased purity. If an FGF-like polypeptide is produced intracellularly, the intracellular material (including inclusion bodies for gram-negative bacteria) can be extracted 5 from the host cell using any standard technique known to the skilled artisan, For example, the host cells can be lysed to release the contents of the periplasm/cytoplasm by French press, homogenization, and/or sonicatipn followed by centrifugation. If an FGF-like polypeptide has formed inclusion bodies in the cytosol, the 3.0 inclusion bodies can often bind to the inner and/or outer cellular membranes and thus will b'e found primarily in the pellet material after centrifugation. The pellet material can then be treated at-pH extremes or with chaotropic agent such as a detergent, guanidine, guanidine derivatives, urea, or ured derivatives in the presence of a reducing -agent such as dithiotbreitol at alkaline pH or tris is carboxyethyl phosphine at acid pH to release, break apart, and solubilize the inclusion bodies. The FGF-like polypeptide in its now soluble form can then be analyzed using gel electrophoresis, immunoprecipitation, or the like. If it is desired to isolate the FGF-like polypeptide, isolation may be accomplished using standard methods such as those set forth below and in Marston et al., Meth. Enz., 20 182:264-75 (1990). In some cases, an FGF-like .polypeptide may not be biologically active upon isolation. Various methods for refoldingg" or converting the polypeptide to its tertiary structure and generating disulfide linkages can be used to restore biological activity. Such methods include exposing the solubilized polypeptide to 25 a pH usually above 7 and in the presence of a particular concentration of a chaotrope. The selection of chaotrope is very similar to the choices used for inclusion body solubilization, but usually the chaotrope is used at a lower concentration and is not necessarily the same as chaotropes used for the solubilization. In most cases the refolding/oxidation solution will also contain a 30 reducing agent or the reducing agent plus its oxidized form in a specific ratio to -40 generate a particular redx potential allowing for disulfide shuffling to occur in the formation of the protein's cysteine bridges. Some of the *commonly used redox couples include cysteine/cystamine, glutathione (GSH)/dithiobis GSH, cupric chloride, dithiothreitol(DTT)/dithiane. DTT, and 2 5 mercaptoethanol(bME)/dithio-b(M). In many instances, a cosolvent may be used or may be needed to increase the efficiency of the refolding and the more common reagents used for this purpose include glycerol, polyethylene glycol of various molecular weights, arginine and the like. If inclusion bodies are not formed to a significant degree upon expression 10 of an FGF-like polypeptide, the polypeptide will be found primarily in the supernatant after centrifugation. of -the cell homogenate and may be further isolated from the supernatant using methods such as those set forth below. In situations where it is preferable to partially or completely purify an FGF-like polypeptide such that it is partially dr substantially free of contaminants, is standard methods known to the one killed in the art may be used. Such methods include, without limitation, separation by electrophoresis followed by electroelution, various types of chromatography (affinity, immunoafinmity, molecular sieve, and/or ion exchange), and/or high pressure .liquid chromatography. In some cases, it may be preferable to use more than one of 20 these methods for complete purification. FGF-like polypeptides, fragments, and/or derivatives thereof may also be prepared by chemical synthesis methods (such as solid phase peptide synthesis) using techniques known in the art such as those set forth by Merrifield et al., J, Am. Chem. Soc. 85:2149 (1963); Honghten et al., Proc Nat! Acad, ScL USA 25 82:5132 (1985); and Stewart and Young, Solid Phase Peptide Synthesis (Pierce Chemical Co. 1984). Such polypeptides may be synthesized with or without a methionine on the amino tenninus. . Chemically synthesized FOF-like polypeptides or fragments may be oxidized using methods set forth in these references to form disulfide bridges. FGF-like polypeptides, fragments or 30 derivatives are . expected to have comparable biological activity to the -41 corresponding FGF-like polypeptides, fragments or derivatives produced recombinantly or purified from natural sources, and thus may be used interchangeably with recombinant or natural FGF-like polypeptide. Another means of obtaining FGF-like polypeptide is via purification from 5 biological samples such as source tissues and/or fluids in which the FGF-like polypeptide is naturally found. Such purification can be conducted using methods for protein purification as described above. The presence of the FGF-like polypeptide during purification may be monitored using, for example, an antibody prepared against recombiantly produced FGF-like polypeptide or peptde 10 fragments thereof. Polyeotides Polypeptides of the invention include isolated FGF-like polypeptides and polypeptides related thereto including fragments, variants, fusion polypeptides, 15 and derivatives as defined hereinabove. FGF-like polypeptide fragments of the invention may result from truncations at the amino terminus (with or without a leader sequence). truncations at the carboxyl terminus, and/or deletions internal to the polypeptide. In preferred embodiments, trnmcations and/or deletions comprise about 10 amino acids, or 20 about 20 amino acid, or about 50 amino acids, or about 75 amino acids, or about 100 amino acids, or more than about 100 amino acids. The polypeptide fragments so produced will comprise about 25 contiguous amino acids, or about 50 amino acids, or about 75 amino acids, or about 100 amino acids, or about 150 amino acids, or about 200 amino acids. Such FGF-like polypeptide fragments may 25 optionally comprise an amino terminal methionine residue. FGF-like polypeptide variants of the invention -include one or more amino acid substitutions, additions and/or deletions as compared to SEQ ID NO: 2 or' SEQ ID NO: 4. In preferred embodiments, the variants have from 1 to 3, or from 1 to 5, or from I to 10, or from I to 15, or from 1 to 20, or from 1 to 25, or from 1 30 to 50, or from 1 to 75, or from I to 100, or more than 100 amino acid -42 substitutions, insertions, additions and/or deletions, wherein the substitutions may be conservative, as defined above, non-conservative, or any combination thereof, and wherein the FGF-like polypeptide variant retains an FGF-like activity. The variants may have additions of amino acid residues either at the carboxyl terminus 5 or at the amino terminus (with or without a leader sequence). Preferred FGF-like polypeptide variants include glycosylation variants wherein the number and/or type of glycosylation sites has been altered compared to native FGF-like polypeptide. In one embodiment, FGF-like variants comprise a greater or a lesser number of N-linked glycosylation sites. An N-linked glycosylation site 1.0 is characterized by the sequence: Asn-X-Ser or Thr, where the amino acid residue designated as "X" may be any type of amino acid except proline.. Substitution(s) of amino acid residues to create this sequence provides a potential new site for addition of an N-linked carbohydrate chain. Alternatively, substitutions to eliminate this sequence will remove an existing N-linked carbohydrate chain. i5 Also provided is a rearrangement of N-linked carbohydrate chains wherein one or more N-linked glycosylation sites (typically those that are naturally occurring) are eliminated and one or more new N-linked sites are created. Additional preferred FGF-like variants include cysteine variants, wherein one or more cysteine residues are deleted or substituted with another amino acid (eg., serine), Cysteine variants 20 are useful when FGF-like polypeptide must be refolded into a biologically active conformation such as after isolation of insoluble inclusive bodies. Cysteine variants generally have fewer cysteine residues than the native protein, and typically have an even number to minimize interactions resulting from unpaired cysteines. 25 One skilled hi the art will be able to determine suitable variants of the native FGF-like polypeptide using well-known techniques. For example; one may be able to predict suitable areas of the molecule that may be changed without destroying biological activity. Also, one skilled in the art will realize that even areas that may be important for biological activity or for structure may be subject -43 to conservative amino acid substitutions without destroying the biological activity or without adversely affecting the polypeptide structure. For predicting suitable areas of the molecule that may be changed without destroying activity, one skilled in the art may target areas not believed to be 5 important for activity. For example, when similar polypeptides with similar activities from the same species of from other species are known, one skilled in the art may compare the amino acid sequence of FGF-like polypeptide to such similar polypeptides. After making such a comparison, one skilled in the art would be able to determine residues and portions of the molecules that are o conserved among similar polypeptides. One skilled in the art would know that changes in areas of the FGF-like molecule that are not conserved would be less likely to adversely affect biological activity and/or structure. One skilled in the art would also know that, even in relatively conserved regions, one could have likely substituted chemically similar amino acids for the naturally occurring residues 15 while retaining activity (conservative amino acid residue substitutions). Also, one skilled in the art may -review -stucture-function studies identifying residues in similar polypeptides that are important for activity or structure. In view of such a comparison, one skilled in the art can predict the importance of amino acid residues in FGF-like polypeptide that correspond to 20 amino acid residues that are important for activity or structure in similar polypeptides. One skilled in the art may opt for chemically similar amino acid substitutions for such predicted important amino acid residues of FGF-like polypeptide. If available, one skilled in the art can also analyze the tbree-dimensional 25 structure and amino acid sequence in relation to that structure iii similar polypeptides. In view of that information, one skilled in the art may be able to predict the alignment of amino acid residues of FGF-like polypeptide with respect to its three dimensional structure. One skilled in the art may choose not to make radical changes to amino acid residues predicted to be on the surface of the -44 protein, since such residues may be involved in important interactions with other molecules. Moreover, one skilled in the art could generate test variants containing a single amino acid substitution at each amino acid residue, The variants could be S screened using activity assays disclosed in this application. Such variants could be used to gather information about suitable variants. For example, if one discovered that a change to a particular amino acid residue resulted in destroyed activity, variants with such a change would be avoided. In other words, based on information gathered from such experiments, when attempting to find additional 10 acceptable variants, one skilled in the art would have know the amino acids where farther substitutions should be avoided either alone or in combination with other mutations. FGF-like fusion polypeptides of the invention comprise FGF-like polypeptides, fragments, variants, or derivatives fused to a heterologous peptide or 15 protein. Heterologous peptides and proteins inclu4, but are not limited to: an epitope to allow for detection and/or isolation of an FGF-like fusion polypeptide; a transmembranc receptor protein or a portion thereof, such as an extracellular domain, or a transmembrane and intracellular domain; a ligand or a portion thereof which binds to a transmembrane receptor protein; an enzyme or portion 20 thereof which is catalytically active; a protein or peptide which promotes oligomerization, such as leucine zipper domain; and a protein or peptide which increases stability, such as an imminunoglobulin constant region. An FGF-like polypeptide may be fused to itself or to a fragment, variant, or derivative thereof. Fusions may be made either at the amino terminus or at the carboxyl terminus of 25 an FGF-like polypeptide, and may be direct with no liner or adapter molecule or may be through a tinker or adapter molecule, such as one or more amino acid residues up to about 20 amino acids residues, or up to about 50 amino acid residues. A tinker or adapter molecule may also be designed with a cleavage site for a DNA restriction endonuclease or for a protease to allow for separation of the 3o fused moieties, -45 In a further embodiment of the invention, an FGF-like polypeptide, fragment, variant and/or derivative is fused to an Fc region of human IgG. In one example, a human IgG hinge, CH2 and CH3 region may be fused at either the N terminus or C-terminus of the FGF-like polypeptides using methods known to the 5 skilled artisan. In another example, a portion of a hinge regions and CH2 and CH3 regions may be fused. The FGF-like Fc-fusion polypeptide so produced may be purified by use of a Protein A affinity column. In addition, peptides and proteins fused to an Fc region have been found to exhibit a substantially greater half-life in vivo than the unfused counterpart. Also, a fusion to an Fc region 10 allows for dimerization/multimerization of the fusion polypeptide. The Fe region may be a naturally occrring Fe region, or may be altered to improve certain qualities, such as therapeutic qualities, circulation time, reduced aggregation, etc. FGF-like polypeptide derivatives are included in the scope of the present invention. Such derivatives are chemically modified FGF-like polypeptide 15 compositions in which FGF-like polypeptide is linked to a polymer. The polymer selected is typically water-soluble so that the protein to which it is attached does not precipitate in an aqueous environment, such as a physiological environment. The polymer may be of any molecular weight, and may be branched or branched. Included within the scope of FGF-like polypeptide polymers is a 20 mixture of polymers. Preferably, for therapeutic use of the end-product preparation, the polymer will be pharmaceutically acceptable. The water soluble polymer or mixture thereof may be selected from the group consisting of; for example, polyethylene glycol (PEG), monomethoxy polyethylene glycol, dextran (such as low molecular weight dextran, of, for 25 example about 6 kD), cellulose, or other carbohydrate based polymers, poly-(N vinyl pyrrolidone) polyethylene glycol, propylene glycol homopolymers, a polypropylene oxide/ethylene oxide co-polymer, polyoxyethylated polyols (e.g., glycerol) and polyvinyl alcohoL Also encompassed by the invention are bifunctional PEG cross-linking molecules that may be used to prepare covalently 30 attached FGF-like polypeptide multimers -46 For the acylation reactions, the polymer(s) selected should have a single reactive ester group. For reductive alkylation, the polymer(s) selected should have a single reactive aldehyde group. A reactive aldehyde is, for example, polyethylene glycol propionaldehyde, which is water stable, or mono C1-C1O 5 alkoxy or aryloxy derivatives thereof (see U.S. Patent No. 5,252,714). Pegylation of FGF-like polypeptides may be carried out.by any of the pegylation reactions known in the art, as described for example in the following references: Francis et al., Focus on Growth Factors 3, 4-10 (1992); BP 0 154 316; EP 0 401 384 and U.S. Patent No. 4,179,337. Pegylation may be carried out via 10 an acylation reaction or an alkylation reaction with a reactive polyethylene glycol molecule (or an analogous reactive water-soluble polymer) as described below. One water-soluble polymer for use herein is polyethylene glycol,, abbreviated PEG. 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 15 mono-(C1-CIO) alkoxy- or eryloxy-polyethylene glycol. In general, chemical derivatization may be perfouned under any suitable conditions used to react a biologically active substance with an activated polymer molecule. Methods for preparing pegylated FGF-like polypeptides will generally comprise the steps of (a) reacting the polypeptide with polyethylene *glycol (such 2C. as a reactive ester or aldehyde derivative of PEG) under conditions whereby FGF like polypeptide becomes attached to one or more PEG groups, and (b) obtaining the reaction product(s). In general, the optimal reaction conditions for the acylation reactions will be determined based on known parameters and the desired result. For example, the larger the ratio of PEG: protein, the greater the 25 percentage of poly-pegylated product. In a preferred embodiment, the FGF-lke polypeptide derivative will have a single PEG moiety at the amino terminus. See U.S. Patent No. 5,234,784, herein incorporated by reference. Generally, conditions that may be alleviated or modulated by 30 administration of the present FGF-like polypeptide derivative include those -47 described herein for FGF-like polypeptides. However, the FGF-like polypeptido derivative 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 non-derivatized molecules. 5 - Antibodies FGF-like polypeptides; fragments, variants, and derivatives may be used to prepare antibodies using methods known in the art. Thus, antibodies and antibody fragments that bind FGF-like polypeptides are within the scope of the present 10 invention. Antibodies may be polyclonal, monospecific polyclonal, monoclonal, recombinant, chimeric, humanized, fully human, single chain and/or bispecific Polyclonal antibodies directed toward an FGF-like polypeptide generally are raised in animals (e.g., rabbits or mice) by multiple subcutaneous or intraperitoneal injections of FGF-like polypeptide and an adjuvant. It may be 15 useful t6 conjugate an FGF-like polypeptide, or a variant, fragment or derivative thereof to a carrier protein that is immunogenic in the species to be immunized, such as keyhole- limpet hemocyanin, serum, albumin, bovine thyroglobulin, or soybean trypsin inhibitor. Also, aggregating agents such as alum are used to enhance the immune response. After immunization, the animals are bled and the 20 serum is assayed for anti-FGF-like antibody titer. Monoclonal antibodies directed toward FGF-like polypeptide are produced using -any method that provides for the production of antibody molecules by continuous cell lines in culture. Examples of suitable methods for preparing monoclonal antibodies. include hybridoma methods of Kohler, et al., Nature 25 256:495-97 (1975), and the human B-cell hybridoma method, Kozbor, J Immunol. 133:3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications 51-63 (Marcel Dekker 1987). Also provided by the invention are hybridoma cell lines that produce monoclonal antibodies reactive with FGF-like polypeptides.
-48 Monoclonal antibodies of the invention may be modified for use as therapeutics. One embodiment is a "chimeric" antibody in which a portion of the heavy and/or light chain is identical with or homologous to corresponding sequence in antibodies derived from a particular species or belonging to a 5 particular antibody class or subclass, while the remainder of the chain(s) is identical with or homologous to corresponding sequence in antibodies derived from another species or belonging to another.antibody class or subclass, as well as fragments of such antibodies, so long as they exhibit the desired biological activity (see U.S. Patent No 4,816,567; Morrison, et aL, Proc. Natl. Acad. Sci. 10 U.S.A. 81: 6851-55 (1985), In another embodiment, a monoclonal antibody of the invention is a "humanized" antibody. Methods for humanizing non-human antibodies are well known in the art. Generally, a humanized antibody has one or more amino acid residues introduced into it from a source that is non-human. Humanization can be 15 perfonied following methods known in the art (Jones, et al., Nature 321: 522-25 (1986); Riechmann, et al., Nature 332:323-27 (1988); Verhoeyen et al., Science 239:1534-36 (1988)), by substituting-rodent complementarily-deteraining regions (CDRs) for the corresponding regions of a human antibody. Also encompassed by the invention are fully human antibodies that bind 20 FGF-like polypeptides, fragments, variants, and/or derivatives. Such antibodies are produced by immunization with an FGF-like antigen (optionally conjugated to a carrier) of transgenic animals (e.g., mice) that are capable of producing a repertoire of human antibodies in the absence of endogenous immunoglobulin, production. See, e.g., Jakobovits, et al., Proc. Naitl Acad. Sci. U.S.A. 90: 255.1-55 25 (1993); Jakobovits, et al., Nature 362:255-58 (1993); Bruggermann et al., Year in Immune. 7:33 (1993). Human antibodies can also be produced in phage-display libraries (Hoogenboom et al., J. MaL BioL 227:381 (1991); Marks, et aL, J. Mol Biol. 222:581 (1991)). Chimeric, CDR grafted and humanized antibodies are typically produced 30 by recombinant methods. Nucleic acids encoding the antibodies are introduced -49 into host cells and expressed using materials and procedures described herein above. In a preferred embodiment, the antibodies are produced iri maimmalian host cells, such as CHO cells. Fully human antibodies may be produced by expression of recombinant DNA transfected into host cells or by expression in S hybridoma cells as described above. For diagnostic applications, in certain embodiments, anti-FGF-like antibodies typically will be labeled with a detectable moiety. The detectable moiety can be any one that is capable of producing, either directly or indirectly, a detectable signal. For example, the detectable moiety may be a radioisotope, such iao as 3 H, "C 4
_
2 p, IIS, or 125, a fluorescent or chemiluminescent compound, such as fluorescein isothiocyanate, rhodamine, or luciferin; or an enzyme' such as alkaline phosphatase, P-galactosidase or horseradish peroxidase. Bayer, et al., Meth. Enz. 184: 138-63 (1990). The anti-FGF-like antibodies of the invention may be eriployed in any 15 known assay method, such as competitive binding assays, direct and indirect sandwich assays, and dinmunoprecipitation assays (Sola, Monoclonal Antibodies: A Manual of Techniques 147-58 (CRC Press 1987)) for detection and quantitation of FGF-like polypeptides. The antibodies will bind FGF-like polypeptides with an affinity that is appropriate for the assay method being employed. 20 Competitive binding assays rely on the ability of a labeled standard (eg., an FGF-like polypeptide, or an immunologically reactive portion thereof) to compete with the test sample analyte (an FGF-like polypeptide) for binding with a limited amount of anti FGF-like antibody. The amount of an FGF-like polypeptide in the test sample is inversely proportional to the amount of standard 25 that becomes bound to the antibodies. To facilitate determining the amount of standard that becomes bonnd, the antibodies typically are insolubilized before or after the competition, so that the standard and analyte that are bound to the antibodies may conveniently be separated from the standard and analyte which remain unbound.
- 50 Sandwich assays involve the use of .two antibodies, each capable of binding to a different immunogenic portion, or epitppe, of the protein to be detected and/or quantitated. In 4 sandwich assay, the test sample analyte is typically bound by a first antibody which is immobilized on a solid support, and 5 thereafter a second antibody binds to the analyte, thus forming an insoluble three part complex. See, e.g., U.S. Patent No. 4,376,110. The second antibody may itself be labeled with a detectable moiety (direct sandwich assays) or may be measured using an anti-immunoglobulin antibody that is labeled with a detectable moiety (indirect sandwich assays). For example, one type of sandwich assay is an 10 ELISA assay, in which case the detectable moiety is an enzyme. The anti-FGF.-like antibodies of the invention also are useful for in vivo imaging, wherein an antibody labeled with a detectable moiety is administered to an animal, preferably into the bloodstream, and the presence and location of the labeled antibody in the host is assayed. The antibody may be labeled with any 15 moiety that is detectable iu an animal, whether by nuclear magnetic resonance, radiology, or other detection means known in the art. The invention also relates to a kit comprising anti-FOF-like antibodies and other reagents useful for detecting FGF-like polypeptide levels in biological samples. Such reagents may include a secondary activity, a detectable label, 20 blocking serum, positive and negative control samples and detection reagents. Antibodies of the invention may be used as therapeutics. Therapeutic antibodies are generally agonists or antagonists, in that they either enhance or reduce, respectively, at least one of the biological activities of an FGF-like polypeptide. In one embodiment, antagonist antibodies of the invention are 25 antibodies or binding fragments thereof which are capable of specifically binding to an FGF-like polypeptide, fragment, variant, and/or derivative, and which are capable of inhibiting or eliminating the finctional activity' of an FGF-like polypeptide in vivo or in vitro. In preferred embodiments, an antagonist antibody will inhibit the functiouial activity of an FOF-like polypeptide at least about 50%, 30 and preferably at least about 80%. In another embodiment antagonist antibodies -51 are capable of interacting with an FGF-like binding partner thereby inhibiting or eliminating FGF-like activity in vitro or in vivo. Agonist and antagonist anti FGF-like antibodies are identified by screening assays described below. 5 Genetically Engineered Non-Human Animals Additionally included within the scope of the present invention are non human animals such as mice, rats, or other rodents, rabbits, goats, sheep, or other farm animals, in which the genes encoding native FGF-like polypeptide have been disrupted (Le., "knocked out") such that the level of expression of FGF-like 10 polypeptide is significantly decreased or completely abolished. Such animals may be prepared using techniques and methods such as those described in U.S. Patent No. 5,557,032. The present invention further includes non-human animals such as mice, rats, or other rodents, rabbits, goats, sheep, or other farm animals, in which a gene 15 encoding a native form of FGF-like polypeptide for that animal or a heterologous FOE-like polypeptide gene is overexpressed by the animal, thereby creating a "transgenic" animal. Such transgenic animals may be prepared using well known methods such as those described in U.S. Patent No 5,489,743 and PCT Pub. No. WO 94/28122. 20 The present invention further includes non-human animals in which the promoter for one ori more of the FGF-like polypeptides of the present invention is either activated or inactivated (e.g., by using homologous recombination methods as described below) to alter the level of expression of one or more of the native FGF-like polypeptides. 25 Such non-human animals may be used for drug candidate screening- The impact of a drug candidate on the animal may be measured- For example, drug candidates may decrease or increase expression of the FGF-like polypeptide gene. In certain embodiments, the amount of FGF-like polypeptide or an FGF-like polypeptide fragment that is produced may be measured after exposure of the 30 animal to the drug candidate. In certain embodiments, one may detect the actual - 52 impact of the drug candidate on the animal. For example, overexpression of a particular gene may result in, or be associated with, a disease or pathological condition. In such cases, one may test a drug candidate's ability to decrease expression of the gene or its ability to prevent or inhibit a pathological condi6on. 5 In other examples, production of a particular metabolic product such as a fragment of a polypeptide, may result in, or be associated with, a disease or pathological condition. In such cases, one may test a drug candidate's ability to decrease production. of such a metabolic product or its ability to prevent or inhibit a pathological condition. 10 Modulators ofFGF-like polypeptide activity In some situations, it may be desirable to identify molecules tiat are modulators, i.e., agonists or antagonists, of the activity of FGF-like polypeptide. Natural or synthetic molecules that modulate FGF-like polypeptide can be 15 identified using one or more screening assays, such as those described below. Such molecules may be administered either in an ex vivo mainer or in an in viva manner by local or intravenous injection or by oral delivery, implantation device, or the like. The following definition is used herein for describing the assays: 20 "Test molecule" refers to a molecule that is under evaluation for the ability to modulate (i.e., increase or decrease) the activity of an FGF-like polypeptide. Most commonly, a test molecule will interact directly with ah FGF-like polyeptide. However, it is also contemplated that a test molec.nle may also modulate FGF-like polypeptide activity indirectly, such as by affecting FGF-like 25 gene expression, or by binding to an FGF-like binding partner (e.g., receptor or ligand). In one embodiment, a test molecule will bind to an FGF-like polypeptide with an affinity constant of at least about 10e M, preferably about 10~ M, more preferably about 10- 9 M, and even more preferably about If' 0 M. Methods for identifying compounds that interact with FGF-like 30 polypeptides are encompassed by the invention. In certain embodiments, an FGF- -53 like polypeptide is incubated with a test molecule under conditions that permit interaction of the test molecule with an FGF-like polypeptide, and the extent of the interaction can be measured. The test molecule may be screened in a substantially purified fonn or in a crude mixture. Test molecules may be nucleic . acid molecules, proteins, peptides, carbohydrates, lipids, or small molecular weight organic or inorganic compounds. Once a set of test molecules has been identified as interacting with an FGF-like polypeptide, the molecules may be further evaluated for their ability to increase or decrease FGF-like polypeptide activity, 10 Measurement of the. interaction of test molecules with FGF-like polypeptides may be carried out in several formats, including cell-based binding assays, membrane binding assays, solution-phase assays and immunoassays. In general, test molecules are incubated with an FGF-like polypeptide for a specifed period of- time and FGF-like polypeptide activity is determined by one or more 15 assays described herein for measuring biological activity. Interaction of test molecules with FGF-like polypeptides may also be assayed directly using polyclonal or monoclonal antibodies in an innnunoassay. Alternatively, modified forms of FGF-like polypeptides containing epitope tags as described above may be used in solution and immunoassays. 20 In certain embodiments, an FGF-like polypeptide agonist 'or antagonist may be a protein, peptide, carbohydrate, lipid, or small molecular weight molecule that interacts with FGF-like polypeptide to regulate its activity. Potential protein antagonists of FGF-like polypeptide include antibodies that interest with active regions of the polypeptide and inhibit or eliminate at least one activity of FGF-like 25 polypeptide. Molecules which regulate FGF-like polypeptide expression may include nucleic acids which are complementary to nucleic acids encoding an FGF like polypeptide, or are complementary to nucleic acids sequences which direct or control expression of FGF-like polypeptide, and which act as anti-sense regulators of expression.
54 In the event that FGF-like polypeptides display biological activity through interaction with a binding partner (e.g., a receptor or a ligand), a variety of in vitro assays may be used to measure binding of an FGF-like polypeptide to a corresponding binding partner. These assays may be used to screen test molecules 5 for their ability to increase or decrease the. rate and/or the extent of binding of an FGF-like polypeptide to its binding partner. In one assay, an FGF-like polypeptide is immobilized by attachment to the bottom of the wells of a microtiter plate. Radiolabeled FGF-like binding partner (for example, iodinated FOP-like binding partner) and the test molecules can then be added either one at a 10 time (in either order) or simultaneously to the wells. After incubation, the wells can be washed and counted using a scintillation counter for radioactivity' to determine the extent of binding to FGF-like protein by its binding partner. Typically, the molecules willbe rested over a range of concentrations and a series of control wells lacking one or more elements of the test assays can be used for 15 accuracy in evaluation of the results. An alternative to this method involves reversing the "positions" of the proteins, 4e., immobilizing FGF-like binding partner to the microtiter plate wells, incubating with the test molecule and radiolabeled FGF-like polypeptide, and determining the extent of FGF-like binding (see, e.g., Current Protocdls in Molecular Biology, chap. 18 (Ausubel et 20 al, eds., John Wiley & Sons 1995)). As an alternative to radiolabeling, an FGF-like polypeptide or its binding partner may be conjugated to biotin and the presence of biotinylated protein can then be detected using streptavidin linked to an enzyme, such as horse radish peroxidase (HRP) or alkaline phosphatase (AP), that can be detected 25 colorometrically, or by fluorescent tagging of streptavidin. An antibody directed to an FGF-like polypeptide or to an FGF-like binding partner. and that is conjugated to biotin may also be used and can be detected after incubation with enzyme-linked streptavidin linked to AP or HRP An FGF-like polypeptide and an FOF-like binding partner may also be 30 immobilized by attachment to agarose beads, acrylic beads, or other types of such -55 inert substrates. The substrate-protein complex can be placed in a solution containing the complementary protein and the test compound; after incubation, the beads can be precipitated by centrifugation, and the amount of binding between an FGF-like polypeptide and its binding partner can be assessed using the methods 5 described above. Alternatively, the substrate-protein complex can be immobilized in a column and the test molecule and complementary protein passed over the column. Formation of a complex between an FGF-like polypeptide and its binding partner can then be assessed using any of the techniques set forth above, i.e., radiolabeling, antibody binding, or the like. 10 Another in vitro assay that is useful for identifying a test molecule which increases or decreases formation of a complex between an FGF-like binding protein and an FGF-like binding partner is a surface plasmon resonance detector system such as the Biacore assay system (Pharmacia, Piscataway, NJ). The Biacore system may be carried out using the manufacturers protocol. This essay 15 essentially involves covalent binding of either FGF-like polypeptide or an FGF like binding partner to a dextran-coated sensor chip that is located in a detector. The test compound and the other complementary protein can then be injected into the chamber containing the sensor chip either simultaneously or sequentially and the amount of complementary protein that binds can be assessed based on the 20 change in molecular mass which is physically associated with the dextran-coated side of the sensor chip; the change in molecular mass can be measured by the detector system. In some cases, it may be desirable to evaluate two or more test compounds together for their ability to increase or decrease formation of a complex between 25 an FGF-like polypeptide and an FGF-like binding partner complex. In these cases, the assays set forth above can be readily modified by adding such additional test compounds either simultaneous with, or subsequent to, the first test compound. The remaining steps in the assay are as set forth above. In vitro assays such as those described above may be used advantageously 30 to screen rapidly large numbers of compounds for effects on complex formation -56 by FGF-like polypeptide and FGF-like binding partner. The assays may be automated to screen compounds generated in phage display, synthetic peptide, and chemical synthesis libraries. Compounds which increase or decrease formation of a complex between 5 an FGF-like polypeptide and an FGF-like binding partner may also be screened in cell culture using cells and cell lines expressing either FGF-like polypeptide or FGF-like binding partner. Cells and eell lines may be obtained from any manual, but preferably will be from human or other primate,. canine, or rodent sources. The binding of an FGF-like polypeptide to cells expressing FGF-like 10 binding partner at the surface is evaluated in the prdsence.or absence of test molecules and the extent of binding may be determined by, for example, flow cytometry using a biotinylated antibody to an FGF-like binding partner. Cell culture assays may be used advantageously to further evaluate compounds that score positive in protein binding assays described above. 15 Cell cultures can also be used to screen the impact of a dug candidate. For example, drug candidates may decrease or increase expression of the FGF-like polypeptide gene. In certain embodiments, the amount of FGF-like polypeptide or an FGF-like polypeptide fragment that is produced may be measured after exposure of the cell culture to the drug candidate. In certain embodiments, one 20 may detect the actual impact of the drug, candidate on the cell culture. For example, overexpression of a particular gene may have a particular impact on the cell culture. In such cases, one may test a drug candidate's ability to increase or decrease expression of the gene or its ability to prevent or inhibit a particular impact on the cell culture. In other examples, production of a particular metabolic 25 product such as a fragment of a polypeptide, may result in, or be associated with, a disease or pathological condition. In such cases, one may test a drug candidate's ability to decrease production of such a metabolic product in a cell culture.
-57 Cell Source Identification Using FGF-like Polypeptide According to certain embodiments, it may be useful to be able to determine the source of a certain cell type. For example, it may be useful to 5 determine the origin of a disease or pathological condition that may aid in selecting appropriate therapy, FGF-like polypeptide is specifically expressed in the liver (and weakly expressed in the lung). In certain embodiments, nucleic acid encoding an FGF-like polypeptide can be used as a probe to identify liver-derived cells by screening the nucleic acids of the cells with such a probe. In other 10 embodiments, one may use the FGF-like polypeptide to mnake antibodies that are specific for FGF-like polypeptide. .Such antibodies can be used to test for the presence of FGF-like polypeptide in cells, and thus, as a means for determining whether such cells 'were derived from the liver. 15 FGF-like Polypeptide Compositions and Administration Therapeutic compositions of FGF-like polypeptides are within the scope of the present invention. Such compositions may comprise a therapeutically effective amount of an FGF-like polypeptide, fragment, variant or derivative in admixture with a pharmaceutically acceptable agent such as a pharmaceutically 20 acceptable carrier. The carrier material may be water for injection, preferably supplemented with other materials common in solutions for administration to mammals. Typically, an FGF-like polypeptide therapeutic compound will be administered in the form of a composition comprising purified polypeptide, fragment, variant, or derivative in conjunction with one or more physiologically 25 acceptable agents. Neutral buffered saline or saline mixed with serum albumin are exemplary appropriate carriers. Preferably, the product is formulated as a lyophilizate using appropriate excipients (eg., sucrose). Other standard pharmaceutically acceptable agents such as carriers, diluents, and recipients may be included as desired, Other exemplary compositions comprise Tris buffer of -58 about pH 7.0-8.5, or acetate buffer of about pH 4.0-5.5, which may further include sorbitol or a suitable substitute therefor. FGF-like polypeptide pharmaceutical compositions typically include a therapeutically or prophylactically effective amount of FGF-like protein in 5 admixture with one or .more pharmaceutically and physiologically acceptable formulation agents selected for suitability with the mode of administration. Suitable formulation materials or pharmaceutically acceptable agents include, but are not limited to, antioxidants, preservatives, coloring, flavoring and diluting agents, emulsifying agents, suspending agents, solvents, fillers, bulking agents, 10 buffers, delivery vehicles, dilnents, excipients and/or pharmaceutical adjuvants. For example, a suitable vehicle may be water for injection, physiological saline solution, or artificial cerebrospinal fluid, possibly supplemented with other materials common in compositions for parenteral administration. Neutral buffered saline or saline mixed with serum albumin are further exemplary vehicles. The 15 term "pharmaceutically acceptable carrier" or "physiologically acceptable carrier" as used herein refers to a formulation agent(s) suitable for accomplishing or enhancing the delivery of the FGF-like protein as a pharmaceutical composition. The primary solvent in a composition may be either aqueous or non aqueous in nature. In addition, the vehicle may contain other formulation 20 materials for modifying or maintaining the pH, osmolarity, viscosity, clarity, color, sterility, stability, rate of dissolution, or odor of the formulation. Similarly, the composition may contain additional formulation materials for modifying or maintaining the rate of release of FGF-like protein, or for promoting the absorption or penetration of FGF-like protein. 25 The FGF-like polypeptide compositions can be administered parentally. Alternatively, the compositions may be administered intravenously or subcutaneously. When systemically administered, the therapeutic compositions for use in this invention may be in the form of a pyrogen-free, parentally acceptable aqueous solution. The preparation of such pharmaceutically acceptable - 59 protein solutions, with due regard to pH, isotonicity, stability and the like, is within the skill of the art, Therapeutic formulations of FGF-like polypeptide compositions useful for practicing the present invention may be prepared for storage by mixing the 5 selected composition having the desired degree of purity with optional physiologically acceptable carriers, excipients, or stabilizers (Remington's Pharmaceutical Sciences (18th Ed., AR Gennaro, ed., Mack Publishing Company 1990) in the forn of a lyophilized cake or an aqueous solution. Acceptable cariers, excipients or stabilizers preferably are nontoxic to recipients 10 and are preferably inert at the dosages and concentrations employed, and preferably include buffers such as phosphate, citrate, or other organic acids; antioxidants such as ascorbic acid, low molecular weight polypeptides; proteins, such -as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, 15 arginine or lysine; monosaccharides, disaccharides, and other carbohydrates including glucose, nannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; salt-forning counterions such as sodium; and/or nonionic surfactants such as Tween, pluronics dr polyethylene glycol (PEG). 20 The optimal pharmaceutical formulation will be determined by one skilled in the art depending upon the intended route of administration, delivery format and desired dosage. See, e.g., Remington's Pharmaceutical Sciences, 1435-1712 (18th Ed., A.R. Gennaro, ed., Mack Publishing Company 1990). Such compositions may influence the physical state, stability, rate of in vivo release, and rate of in 25 vivo clearance of the present FGF-like protein. An effective amount of an FGF-like polypeptide composition to be employed therapeutically will depend, for example, upon the therapeutic objectives such as the indication for which the FGF-like polypeptide is being used, the route of administration, and the condition of the patient. Accordingly, it may 30 be necessary for the therapist to titer the dosage and modify the route of - 60 administration as required to obtain the optimal therapeutic effect A typical dosage may range from about 0.1 pg/kg to up to about 100 mg/kg or more, depending on the factors mentioned above. XI other embodiments, the dosage may range from I gg/kg up to about 100 mg/kg; or 5 sg/kg up to about 100 5 mg/kg; or 0.1 jig/kg up to about 100 mg/kg; or 1 g/kg up to about 100 mg/kg. Typically, a clinician will administer the composition until a dosage is reached that achieves the desired effect The composition may therefore be administered as a single dose or as two or more doses (which may or may not contain the same amount of FGF-like polypeptide) over time, or as a continuous infusion via 10 implantation device or catheter. As further studies are conducted, information will emerge regarding appropriate dosage levels for treatment of various conditions in various patients, and the ordinary skilled worker, considering the therapeutic context, the type of disorder under treatment, the age and general health of the recipient, will be able 15 to ascertain proper dosing. The FGF-like polypeptide composition to be used for in vivo parenteral administration typically must be sterile. This is readily accomplished by filtration through sterile filtration membranes. Where the composition is lyophilized, sterlization using these methods may be conducted either prior to, or following, 20 lyophilization and reconstitution- The composition for parenteral administration ordinarily will be stored in lyophilized form or in solution. Therapeutic compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle. 25 Effective administration forms, such as (1) slow-release formulations, (2) inhalant mists, or (3) orally active formulations are also envisioned. The FGF-like polypeptide pharmaceutical composition also may be formulated for parenteral administration. Such parenterally administered therapeutic compositions are typically in the for of a pyrogen-free, parenterally acceptable aqueous solution 30 comprising FGF-like polypeptide in a pharmaceutically acceptable vehicle. The -61 FGF-like polypeptide pharmaceutical compositions also may include particulate preparations of polymeric compounds such as polylactic acid, polyglycolic acid, etc., or the introduction of FGF-like polypeptide into liposomes. Hyaluronic acid may also be used, and this may have the effect of promoting sustained duration in 5 the circulation. A particularly suitable vehicle for parenteral injection is sterile distilled water in which FGF-like polypeptide is formulated as a sterile, isotonic solution, properly preserved. Yet another preparation may involve the formulation of FGF like polypeptide with an agent, such as injectable microspheres, bio-erodible 10 particles or beads, or liposomes, that provides for the controlled or sustained release of the protein product which may then be delivered as a depot injection. Other suitable means for the introduction of-FGF-like polypeptide include implantable drug delivery devices that contain the FGF-like polypeptide. The preparations of the present invention may include other components, 15 for example parenterally acceptable preservatives, tonicity agents, cosolvents, wetting agents, completing agents, buffering agents, antimicrobials, antioxidants and surfactants, as are well known in the art. For example, suitable tonicity enhancing agents include alkali metal halides (preferably sodium or potassium chloride), mannitol, sorbitol and the like. Suitable preservatives include, but are 20 not limited to, benzalkoniun chloride, thimerosal, phenethyl alcohol, methylparaben, propylparaben, chlorbexidine, sorbic acid and the like. Hydrogen peroxide may also be used as preservative. Suitable cosolvents are for example glycerin, propylene glycol, and polyethylene glycol. Suitable complexing agents are for example caffeine, polyvinylpyrrolidone, beta-cyclodextrin, or 25 hydroxypropyl-beta-cyclodextrin. Suitable surfactants or wetting agents include sorbitan esters, polysorbates such as polysorbate 80, tromethamine, lecithin, cholesterol, tyloxapal and the like. The buffers can be conventional buffers such as borate, citrate, phosphate, bicarbonate, or Tris-HCI. The formulation components are present in concentrations that are 30 acceptable to the site of administration. For example, buffers are used to maintain - 62 the composition at physiological pH or at slightly lower pH, typically within a pH range of from about 5 to about 8. A pharmaceutical composition may be formulated for inhalation. For example, FOP-like polypeptide may be formulated as a dry powder for inhalation. 5 FGF-like polypeptide inhalation solutions may also be formulated in a liquefied propellant for aerosol delivery. In yet another formulation, solutions may be nebulized. It is also contemplated that certain formulations containing FGF-like polypeptide may be administered orally. FGF-like polypeptide that is 10 administered in this fashion. may be formulated with or without those carriers customarily used in-the compounding of solid dosage forms such as tablets and -capsules. For example; a capsule may be designed to release the active portion of the formulation at the point in the gastrointestinal tract when bioavailability is maximized and pre-systernic degradation is minimized. Additional agents may be 15 included to facilitate absorption of FGF-like polypeptide. Diluents, flavorings, low melting point waxes, vegetable oils, lubricants, suspending agents, tablet disintegrating agents, and binders may also be employed. Another preparation may involve an effective quantity of FGF-like polypeptide in a mixture with non-toxic excipients that are suitable for the 20 manufacture of tablets. By dissolving the tablets in sterile water, or other appropriate vehicle, solutions can be prepared in unit dose form. Suitable excipients include, but are not limited to, inert diluents, such as calcium carbonate, sodium carbonate or bicarbonate, lactose, or, calcium phosphate; or binding agents, such as starch, gelatin, or acacia; or lubricating agents such as 25 magnesium stearate, stearic acid, or talc. Additional FGF-like polypeptide formulations will be evident to those skilled in the art, including fonnulations involving FGF-like polypeptide in combination with one or more other therapeutic agents. Techniques for formulating a variety of other sustained- or controlled-delivery means, such as 30 liposome carriers, bio-erodible microparticles or porous beads and depot - 63 injections, are also known to those skilled in the art. See, for example, the Supersaxo et al. description of controlled release porous polymeric microparticles for the delivery of pharmaceutical compositions (See PCT Pub. No. WO 93/15722) the disclosure of which is hereby incorporated by reference. 5 Regardless of the manner of administration, the specific dose may be calculated according to body weight, body surface area, or organ size. Further refinement of the calculations necessary to detennine the appropriate dosage for treatment involving each'of the above frientioned formulations is routinely made by those of ordinary scill in the art and is within the ambit of tasks routinely 10 performed by them. Appropriate dosages may be ascertained through use of appropriate dose-response data.' the route of administration of the composition is in accord with known methods, for example, oral, injection or infusion by intravenous, intraperitoneal, intracerebral (intraparenchymal), intracerebroventricular, intramuscular, 15 intraocular, intraarterial, or intralesional routes, or by sustained release systems or implantation device which may optionally involve the use of a catheter. Where desired, the compositions may be administered continuously by infusion, bolus injection or by implantation device. One may further administer the present pharmaceutical compositions by 20 pulmonary administration, see, e.g., PCT Pub. WO 94/20069, which discloses pulmonary delivery of chemically modified proteins, herein incorporated by reference. For pulmonary delivery, the particle size should be suitable for delivery to the distal lung. For example, the particle size may be from 1 pm to 5 pm. However, larger particles may be used, for example, if each particle is fairly 25 porous. Alternatively or additionally, the composition may be administered locally via implantation into the affected area of a membrane, sponge, or other appropriate material on to which FGF-like polypeptide has been absorbed or encapsulated.
64 Where an implantation device is used, the device may be implanted into any suitable tissue or organ, and delivery of FOF-like polypeptide may be directly through the device via bolus, or via continuous administration, or via catheter using continuous infusion. 5 FGF-like polypeptide may be administered in a sustained release formulation or preparation. Suitable examples of sustained-release preparations include semipermeable polymer matrices in the form of shaped articles, for example, films, -or microcapsules. Sustained release matrices include polyesters, hydrogels, polylactides (U.S. Patent No. 3,773,919, EP Patent No. 58,481), 10 copolymers of L-glutamic acid and gamma ethyl-L-glutamate (Sidman et a, Biopolymers 22: 547-56 (1983)), poly (2-hydroxyethyl-methacrylate) (Langer et al., J. Biomed. Mater. Res. 15: 167-277 (1981) and Langer, Chem, Tech. 12: 98 105 (1982)), ethylene vinyl acetate (Langer et al., supra) or poly-D(-)-3 hydroxybutyric acid (EP Patent No. 133,988). Sustained-release compositions is also may include liposomes, which can be prepared by any of several methods known in the art (see, e.g., Epstein et al., Proc. NatL. Acad. Sci. U.S.A. 82:3688-92 (1985); EP Patent Nos. 36,676; 88,046; and 143,949). The FGF-like polypeptides, fragments thereof, variants, and derivatives, may be employed alone, together, or in combination with other pharmaceutical 20 compositions. The FGF-like polypeptides, fragments, variants, and derivatives may be used in combination with cytolines, growth factors, antibiotics, anti inflammatories, and/or chemotherapeutic agents as is appropriate for the indication being treated. In some -cases, it may be desirable to use FGF-like polypeptide 25 compositions in an ex vivo manner. Here, cells, tissues, or organs that have been removed from the patient ar6 exposed to FOP-like polypeptide compositions after which the cells, tissues and/or organs are subsequently implanted back into the patient. In other cases, an FGF-like polypeptide may be delivered through 30 implanting into patients certain cells that have been genetically engineered, using -63 methods such as those described herein, to express and secrete the polypeptides, fragments, variants, or derivatives. Such cells may be animal or human cells, and may be derived from the patient's own tissue or from another source, either human or non-buman. Optionally, the cells may be immortalized. However, in order to 5 decrease the chance of an immunological response, it is preferred that the cells be encapsulated to avoid infiltration of surrounding tissues. The encapsulation materials are typically biocompatible, semi-permeable polymeric enclosures or membranes that allow release of the protein product(s) but prevent destruction of the cells by the patient's immune system or by other detrimental factors from the .10 surrounding tissues. Methods used for membrane encapsulation of cells are familiar to the skilled artisan, and preparation of encapsulated cells and their implantation in patients may be accomplished without undue experimentation. See, e.g., US. Patent Nos. 4,892,538; 5,011,472; and 5,106,627. A system for encapsulating 15 living cells is described in PCT Pub. No. WO 91/10425 (Aebischer et al.). Techniques for formulating a variety of other' sustained or controlled delivery means, such as liposome carriers, bio-erodible particles or beads, are also known to those in the art, and are described. The cells, with or without encapsulation, may be implanted into suitable body tissues or organs of the patient. 20 As discussed above, it may be desirable to treat isolated cell populations such as stem cells, leukocytes, red blood cells, bone marrow, chondrocytes, neurons, pancreatic islets, liver cells and the like with one or more FGF-like polypeptides, variants, derivatives and/or fragments. This can be accomplished by exposing the isolated cells to the polypeptide, variant, derivative, or fragment 25 directly, where it is in a form that is permeable to or acts upon the cell membrane. Additional objects of the present invention relate to methods for both the in vitro production of therapeutic proteins by means of homologous recombination and for the production and delivery of therapeutic proteins by gene therapy. It is further envisioned that FGF-like protein may be produced by 3 a homologous recombination, or with recombinant production methods utilizing - 66 control elements introduced into cells already containing DNA encoding FGF-like polypeptide. For example, homologous recombination methods may be used to modify a cell that contains a normally transcriptionally silent FGF-like gene, or under expressed gene, and thereby produce a cell that expresses therapeutically S efficacious amounts of FGF-like polypeptide. Homologous recombination is a technique originally developed for targeting genes to induce or correct mutations in transcriptionally active genes (Kucherlapati, Prog. in Nucl. Acid Res. and Mol Biol. 36:301 (1989))- The basic technique was developed as a method for introducing specific mutations into specific regions of the mammalian genome 10 (Thomas et al., Cell 44:419-28 (1986); Thomas and Capecclii, Cell 51:503-12, (1987); Doctschman et al., Proc. Natd. Acad ScZ U.S.A. 85:8583-87 (1988)) or to correct specific mutations within defective genes (Doetschman et aL, Nature 330:576-78 (1987)). Exemplary homologous recombination techniques are described in U.S. Patent No. 5,272,071 (EP Patent No. 91 90 3051, EP Publication 15 No. 505 500; PCT/US90/07642, PCT Pub. No. WO 91/09955). Through homologous recombination, the DNA sequence to be inserted into the genome can be directed to a specific region of the gene of interest by attaching it to targeting DNA. The targeting DNA is a nucleotide sequence that is complementary (homologous) to a region of the genonic DNA. Small pieces of 20 targeting DNA that are complementary to a specific region of the genome are put in contact with the parental strand during the DNA replication process. It is a general property of DNA that has been inserted into a cell to hybridize, and therefore, recombine with other pieces of endogenous DNA through shared homologous regions. If this complementary strand is attached to an 25 oligonucleotide that contains a mutation or a different sequence or an additional nucleotide, it too is incorporated into the newly synthesized strand as a result of the recombination. As a result of the-proofreading function, it is possible for the new sequence of DNA to serve as the template. Thus, the transferred DNA is incorporated into the genome. 30 Attached to these pieces of targeting DNA are regions of DNA that may -67 interact with the expression of a FGF-like protein. For example, a promoter/enhancer element, a suppresser, or an exogenous transcription modulatory element is inserted in the genome of the intended host cell in proximity and orientation sufficient to influence the transcription of DNA 5 encoding the desired FGF-like protein. The control element controls a portion of the DNA present in the host cell genome. Thus, the expression of FGF-like protein may be achieved not by transfection of DNA that encodes the FGF-like gene itself, but rat-her by the use of targeting DNA (containing regions of homology with the endogenons gene of interest) coupled with DNA regulatory o segments that provide the endogenous gene sequence with recognizable signals for transcription of a FGF-like protein. In an exemplary method, expression of a desired targeted gene in a cell (i.e., a desired endogenous cellular gene) is altered by the introduction, by homologous' recombination into the cellular genorne at a preselected site, of DNA 15 which includes at least a regulatory sequence, an exon and a splice donor site. These components are introduced into the chromosomal (genomic) DNA in such a manner that this, in effect, results in production of a new transcription unit (in which the regulatory sequence, the exon, and the splice donor site present in the DNA construct are operatively linked to the endogenous gene). As a result of 20 introduction of these components into the chromosomal DNA, the expression of the desired endogenous gene is altered. Altered gene expression, as used herein, encompasses activating (or causing to be expressed) a gene which is normally silent (unexpressed) in the cell as obtained, increasing expression of a gene which may include expressing a gene 25 that is not expressed at physiologically significant levels in the cell as obtained, changing the pattern of regulation or induction such that it is different than occurs in the cell as obtained, and reducing (including eliminating) expression of a gene which is expressed in the cell as obtained. The present invention further relates to DNA constructs useful in the 30 method of altering ~expression of a target gene. In certain embodiments, the - 68 exemplary DNA constructs comprise: (a) a targeting sequence, (b) a regulatory sequence, (c) an exon, and (d) an unpaired splice-donor site. The targeting sequence in the DNA construct directs the integration' of elements (a)-(d) into a target gene in a cell such that the elements (b)-(d) are operatively linked to 5 sequences of the endogenous target gene. In another embodiment, the DNA constructs comprise: (a) a targeting sequence, (b) a-regulatory sequence, (c) an exon, (d) a splice-donor'site, (e) an intron, and (f) a splice-acceptor site, wherein the targeting sequence directs the integration of elements (a)-(f) such that the elements of (b)-(f) are operatively linked to the endogenous gene. The targeting 10 sequence is homologous to the preselected site in the cellular chromosomal DNA with which homologous recombination is to occur. In the construct, the exon is. generally 3' of the regulatory sequence and the splice-donor site is 3' of the exon. If the sequence of a particular gene is known, such as the nucleic acid sequence of FGF-like polypeptide presented herein, a piece of DNA that is 15 complementary to a selected region of the gene can be synthesized or otherwise obtained, such as by appropriate restriction of the native DNA at specific recognition sites bounding the region of interest. This piece serves as- a targeting sequence upon insertion into the cell and will hybridize to its homologous region within the genome. -If this hybridization occurs during DNA r'plication, this piece 20 of DNA, and any additional sequence attached thereto, will act as an Okazaki fragment and will be'backstitched into the newly synthesized daughter strand of DNA. The present invention, therefore, includes nucleotides encoding a FGF-like molecule, which nucleotides may be used as targeting sequences. FGF-like polypeptide cell therapy -- for example, implantation of cells 25 producing FGF-like polypeptide - is also contemplated. This embodiment would involve implanting into the cells of a patient a construct capable of synthesizing and secreting a biologically active form of FGF-like polypeptide. Such FGF-like polypeptide-producing cells may be cells that are natural producers of FGF-like polypeptide or may be recombinant cells whose ability to produce FGF-like 30 polypeptide. has been augmented by transformation with a gene encoding the - 69 desired FGF-like molecule or with a gene augmenting the expression of FGF-like polypeptide. Such a modification may be accomplished by means of a vector suitable for delivering the gene as well as promoting its expression and secretion. In order to minimize a potential immunological reaction in a patient that is being 5 administered an FGF-like protein or polypeptide of a foreign species, it is preferred that the natural cells producing FGF-like polypeptide be of human origin and produce human FGF-like polypeptide. Likewise, it is preferred that the recombinant cells producing FGF-like polypeptide are transformed with an expression vector containing a gene encoding a human FGF-like molecule. 10 Implanted cells may be encapsulated to avoid infiltration of surrounding tissue. Human or non-human animal cells may be implanted in patients in biocompatible, semipermeable polymeric enclosures. or membranes -that allow release of FGF-like polypeptide, but that prevent destruction of the cells by the patient's immune system. or by other detrimental factors from the surrounding 15 tissue. Alternatively, the patient's own cells, transformed to produce FGF-like polypeptide gr vivo, could be implanted directly into the patient without Such encapsulation. Techiiques for the encapsulation of living cells are known in the art, and the preparation of the encapsulated cells and their implantation in a patient may be 20 accomplished without undue experimentation. For example, Baetge et al. (PCT Pub. No. WO 95/05452, the disclosure of which is hereby incorporated -by reference) describe membrane capsules containing genetically engineered cells for the effective delivery of biologically active rmolecules. The capsules are bioconpatible and are easily retrievable. The capsules encapsulate cells 25 transfected with recombinant DNA molecules comprising DNA sequences coding for biologically active molecules operatively linked to promoters that are not subject to down regulation in vivo upon implantation into a mammalian host. The devices provide for the delivery of the molecules from living cells to specific sites within a recipient. In addition, see U.S. Patent Nos. 4,892,538; 5,011,472; and 30 5,106,627. A system for encapsulating living cells is described in PCT Pub. No.
-70 WO 91/10425 (Aebischer et al.), See also, PCT Pub. No. WO 91/10470 (Aebischer et al.); Winn et al., Exper: Neurol. 113:322-29 (1991); Aebischer et al., Exper. Neural, 111:269-75 (1991); and Tresco et al., ASAIO 38:17-23 (1992). In vivo and -in vitro gene therapy delivery of FGF-like polypeptide is also 5 envisioned. In vivo gene therapy may be accomplished by introducing the gene encoding FGF-like polypeptide into cells via local injection of a polynucleotide molecule or other appropriate delivery vectors (Hefti, J Neurobiology 25:1418-35 (1994)). For example, a polynucleotide molecule encoding FGF-like protein may be contained in an adeno-associated virus vector for delivery to the targeted cells 10 (see, e.g., Johnson, PCT Pub. No. WO 95/34670; PCT App. No. PCTIUS95/07178). The recombinant adeno-associated virus (AAV) genome typically contains' AAV inverted tenninal repeats flaaking a DNA sequence encoding FGF-like polypeptide operably linked to functional promoter and polyadenylation sequences. 15 Alternative viral vectors include, but are not limited to, retrovins, adenovimus, herpes simplex virus, and papilloma virus vectors. U.S. Patent No. 5,672,344 describes an in vivo viral-mediated gene transfer system involving a recombinant neurotrophic HSV-1 vector. U.S. Patent No. 5,399,346 provides examples of a process for providing a, patient with a therapeutic protein by the 20 delivery of human cells which have been treated in vitro to insert a DNA segment encoding a therapeutic protein.' Additional methods and materials for the practice of gene therapy techniques are described in U.S. Patent No. 5,631,236 (involving adenoviral vectors); U.S. Patent No. 5,672,510 (involving retroviral vectors); and U.S. Patent 5,635,399 evolvingg retroviral vectors expressing cytokines). 25. Nonviral delivery methods include liposome-mediated transfer, naked DNA delivery. (direct injection), receptor-mediated transfer (ligand-DNA complex), electroporation, calcium phosphate precipitation, and microparticle bombardment (e.g., gene gun). Gene therapy materials and methods may also include inducible promoters, tissue-specific enhancer-promoters, DNA sequences 30 designed for site-specific integration, DNA sequences capable of providing a -71 selective advantage over the parent cell, labels to identify transformed cells, negative selection systems and expression control systems (safety measures), cell specific binding agents (for cell targeting), cell-specific internalization factors, transcription factors to enhance expression by a vector as well -as methods of 5 vector manufacture. Such additional methods and materials for the practice of gene therapy techniques are described in U.S. Patent No. 4,970,154 (electroporation techniques); PCT Pub. No. WO 96/40958 (nuclear ligands); U.S. Patent No. 5,679,559 (concerning a lipoprotein-containing system for gene delivery); U.S. Patent No. 5,676,954 (involving liposome caniers); U.S. Patent i0 No. 5,593,875 concerningg methods for calcium phosphate. transfection); and U.S. Patent No. 4,945,050 (wherein biologically active particles are propelled at cells at a speed whereby the particles penetrate the surface of the cells and become incorporated into the interior of the cells). Expression control techniques include chemical induced regulation (see, e.g., PCT Pub. Nos. WO 96/41865 and WO 15 97/31899), the use of a progesterone antagonist in a modified steroid hormone receptor system '(see, e.g., U.S. Patent No. 5,364,791), ecdysone control systems (see, e.g., PCT Pub. No. WO 96/37609), and positive tetracycline-controllable transactivators (see, e.g., U.S. Patent No. 5,589,362; U.S. Patent No. 5,650,298; and U.S. Patent No. 5,654,168). 20 It is also contemplated that FGF-like polypeptide gene therapy or cell therapy can further include the delivery of a second protein. For example, the host cell may be modified to express and release both FOE-like polypeptide and a second protein, for example insulin-like growth factor I (IGF-1). Alternatively, both FGF-like polypeptide and a second protein nay be expressed in and released 25 from separate cells. Such cells may be separately introduced into the patient or the cells may be contained in a single implantable device, such as the encapsulating membrane described above. One manner in which gene therapy can be applied is to use the FGF-like gene (either genomic DNA, cDNA, and/or synthetic DNA encoding a FGF-like 3 0 polypeptide, or a fragment, variant, or derivative thereof) which may be operably 72 linked to a constitutive or inducible promoter to form a "gene therapy DNA construct." The promoter may be homologous or heterologous to the endogenous FGF-like gene, provided that it is active in the cell or tissue type into which the construct will be inserted. Other components of the gene therapy DNA construct 5 may optionally include: DNA molecules designed for site-specific integration (e.g., endogenous flanking sequences useful for homologous recombination), a tissue-specific promoter, enhancers, silencers, DNA molecules 'capable of providing a selective advantage over the parent cell, DNA molecules useful as labels to identify transformed cells, negative selection systems, cell specific 10 binding agents (as, for example, for cell targeting), cell-specific internalization factors, and transcription factors to enhance expression by a vector as well as factors to enable vector manufacture. This gene therapy DNA construct cap then be introduced into the patient's cells (either ex vivo or in vivo). One means for introducing the gene therapy DNA 15 construct is via viral vectors. Suitable viral vectors typically used in gene therapy for delivery of gene therapy DNA constructs include, without limitation, adenovirus, adeno-associated virus, herpes simplex virus, lentivirus, papilloma virus, and retrovirus vectors. Some of these vectors, such as retroviral vectors, will deliver the gene therapy DNA construct to the chromosomal DNA of the 20 patient's cells, and the gene therapy DNA construct can integrate into the chromosomal DNA; other vectors will function as episomes and the gene therapy DNA construct will remain in the cytoplasm. Another means to increase endogenous FGF-like polypeptide expression in a cell via gene therapy is to insert one or more enhancer elements into the FGF 25 like polypeptide promoter, where the enhancer elements can serve to increase transcriptional activity of the FGF-like polypeptide gene. The enhancer elements used will be selected based on the tissue in which one desires to activate the gene - enhancer elements known to confer promoter activation in that tissue will be selected. For example, if a gene encoding an FGF-like polypeptide is to be 30 "turned on" in T-cells, the ick promoter enhancer element may be used. Here, the -73 ftnctional portion of the transcriptional element to be added may be inserted into a fragment of DNA containing the FGF-like polypeptide promoter (and optionally, inserted into a vector and/or 5' and/or 3' flanking sequences, etc.) using standard cloning techniques. This construct, known as a "homologous recombination 5 construct," can then be introduced into the desired cells either ax vivo or in vivo, Gene therapy can be used to decrease FGF-like polypeptide expression by modifying the nucleotide sequence of the eidogenous promoter. Such modification is typically accomplished via homologous recombination methods. For example, a DNA molecule containing all or a portion of the promoter of the 10 FGF-like gene selected for inactivation can be engineered to remove and/or replace pieces of the promoter that regulate transcription. Here, for example, the TATA box and/or the binding site of a transcriptional activator of the promoter may be deleted using standard molecular biology techniqnes; such deletion can inhibit promoter activity thereby repressing transcription of the corresponding is FGF-like gene, Deletion of the TATA box or transcription activator binding site in the promoter. may be accomplished by generating a DNA construct comprising all or the relevant portion of the FGF-like polypeptide promoter (from the same or a related species as the FGF-like gene to be regulated) in which one or more of the TATA box and/or transcriptional activator binding site nucleotides are mutated -20 via substitution, deletion and/or insertion of one or more nucleotides such that the TATA box and/or activator binding site has decreased activity or is rendered completely inactive. This construct, which also will typically contain at least about 500 bases of DNA that correspond to the native (endogenous) 5' and 3' DNA sequences adjacent to the promoter segment that has been modified, may be 25 introdiiced into the appropriate cells (either ex vivo or in vivo) either directly or via a viral vector as described above. Typically, integration of the construct into the genomic DNA of the cells will be via homologous recombination, where the 5' and 3' DNA sequences in the promoter construct can serve to help integrate the modified promoter region via hybridization to the endogenous chromosomal 3o DNA.
-74 Other gene therapy methods may also be employed where it is desirable to inhibit the activity of one or more FGF-like polypeptides. For example, antisense DNA or RNA molecules, which have a sequence that is complementary to at least a portion of the selected FGF-like polypeptide gene can be introduced into the 5 cell. Typically, each such antisense molecule will be complementary to the start site (5' end) of each selected FGF-like gene. When the antisense molecule then hybridizes to the corresponding FGF-like mRNA, translation of this mnRNA is prevented. Alternatively, gene therapy may be employed to create a dominant 10 negative inhibitor of one or more FGF-like polypeptides. In this situation, the DNA 'encoding a mutant full length or truncated polypeptide of each selected FGF-like polypeptide can be prepared and introduced into-the cells of a patient using either viral or non-viral methods as described above. Each such mutant is typically designed to compete with endogenous polypeptide in its biological role. 15 Uses of FGF-like Nucleic Acids and Polypeptides Nucleic acid molecules of the invention may be used to map the locations of the FGF-like gene and related genes on chromosomes. Mapping may be done by techniques known in the art, such as PCR amplification and- in situ 20 hybridization. The nucleic acid molecules are also used as anti-sense inhibitors of FGF like polypeptide expression. Such inhibition may be effected by nucleic acid molecules that are complementary to and hybridize to expression control sequences (triple helix fbmation) or to FGF-like mRNA. Anti-sense probes may 25 be designed by available techniques sing the sequence of the FGF-like genes disclosed herein. Anti-sense inhibitors provide information relating to the decrease or absence of an FGF-likepolypeptide in a cell or organism. Hybridization probes may be prepared using an FGF-like gene sequence as provided herein to screen cDNA, genomic or synthetic DNA libraries for related 30 sequences. Regions of the DNA and/or amino acid sequence of FOE-like -75 polypeptide that exhibit significant identity to known sequences are readily determined using sequence alignment algorithms disclosed above and those regions may be used to design probes for screening. The nucleic acid molecules of the invention may be used for gene therapy. 5 Nucleic acid molecules that express EGF-like polypeptide in vivo provide information relating to the effects of the polypeptide in cells or organisms. FGF-like nucleic acid molecules, fragments, variants, and/or derivatives that do not themselves encode biologically active polypeptides may be useful as hybridization probes in diagnostic assays to .test, either qualitatively or 10 quantitatively, for the presence of FGF-like DNA or corresponding RNA in mammalian tissue or bodily fluid samples. FGF-like polypeptides, fragments, variants, and/or derivatives may be used to prevent or treat cirhosis or other toxic insult of the liver; inflammatory bowel disease, mucositis, Crohn's disease, or other gastrointestinal abnornality 15 diabetes; obesity; nerodegenerative diseases; wounds; damage to the corneal epithelium, lens, or retinal tissue; damage to renal tubules as a result of acute tabular necrosis; bematopoictic cell reconstitution following chemotherapy; wasting syndromes (for example, cancer associated cachexia), multiple sclerosis, myopathies; short stature, delayed maturation, excessive growth (for example, 20 acromegaly), premature maturation; alopecia; diseases or abnormalities of androgen target organs; infantile respiratory distress syndrome, bronchopuhnonary dysplasia, acute respiratory distress syndrome, or other lung abnormalities; tumors of the eye or other tissues; atherosclerosis; hypercholesterolemnia; diabetes; obesity; strke; osteoporosis; osteoarthritis; degenerative joint disease; muscle 25 atrophy; sarcopenia; decreased lean body mass; baldness; wrinkles; increased fatigue; decreased stamina; decreased cardiac function; immune system dysfunction; cancer, Parkinson's disease; senile dementia; Alzheiner's disease; and decreased cognitive function. FGF-like polypeptide fragments, variants, and/or derivatives, whether 3o biologically active or not, are useful for preparing antibodies that bind to an FGF- -76 like polypeptide.. The antibodies may be used for in vivo and in vitro diagnostic purposes, including, but not limited to, use in labeled fonn to detect the presence of FOF-like polypeptide in a body fluid or cell sample. The antibodies may also be used to prevent or treat conditions that may be associated with an increase in 5 FGF-like polypeptide expression or activity. The antibodies may bind to an FGF like polypeptide so as to diminish or block at least one activity characteristic of an FGF-like polypeptide, or may bind to a polypeptide to increase an activity. A deposit of cDNA encoding FGF-like polypeptide has been made with the American Type Culture Collection, 10801 University Boulevard, Manassas, 10 VA 20110-2209 on September 3, 1999 and having accession No. PTA-626. The following examples are intended for illustration purposes only, and should not be constmed as limiting the scope of the invention in any way, Example 1: Cloning of the Murne FGF-like Polypvetide Gene 15 Generally, materials and methods as described in Sambrook et al. supra were used to clone and analyze the gene encoding rat FOF-like polypeptide. Sequences encoding the murine FGF-like polypeptide were isolated from a mouse regenerating liver cDNA library by screening the library in a kPGF signal trap system (U.S. Patent App. No.'09/026,958). This primary screening technique 20 enriched for clones encoding signal peptide-containing secreted proteins. A primary library (Tmrll) was constructed in the kFGF vector as follows. Regenerating mouse liver was removed 24 hours after partial hepatectomy, and poly A+ RNA prepared using a commercially available RNA extraction kit and mRNA purification kit (Pharmacia Biotech). A cDNA library was prepared using 25 the. SuperScript t " Plasmid System for cDNA Synthesis and Plasmid Cloning (Gibco BRL) with some modifications. First strand reactions were performed using 3 pLg of poly A+ RNA and 500 gg of the primer 5G'--G-A-A-G-G-A-A-A A-A-A-G-C-G-G-C-C-G-C-A-A-C-A-N-N-N-N-N-N-N-N-N-3' (SEQ ID NO: 34). Following second strand synthesis, the cDNA was ligated to a Sal I adapter, 30 digested with Not I, and then size fractionated by agarose gel electrophoresis.
Fractionated cDNA, ranging from 0.2 to 0-8 kb in size, was purified using a Qiagen gel extraction kit and then ligated to the kFGF vector as follows. In a 20 al reaction, 50 ptg of vector DNA (previously digested with Sal I and Not I) was mixed with 20 gg of the purified eDNA, IX ligase buffer, and 1 ml of T4 DNA 5 ligase at 16"C for 20 hours. The product of this ligation was precipitated and introduced into E. coli by electroporation, after which transformed bacterial cells were grown in 5 ml SOC medium at 37C for 1 hour and then frozen at -- 0*C in 10% glycerol. This constituted the primary Tmrll library. Plasmid DNA from the primary Tmrll 10 cDNA library was prepared from pools of 50,000 colonies grown on LB/agar plates using standard procedures. Ten pools were prepared and plasmid DNA was isolated from the pools using Qiagen maxi prep kits. The plasmid DNA that was recovered' was subsequently introduced into NIH 3T3 cells by calcium phosphate transfection. In each reaction 100 ng of is plasmid DNA from each pool was used to transfect approximately 2 x 10 cells in one 35 mm plate. After 24 hours, the cells were then split into five 100 mm plates, grown in normal medium for one day, and then grown in low serum medium for 13 days. Approximately 4000 total colonies were obtained following growth in the low serum media. 20 Signal peptide-enriched regenerating cDNA molecules were recovered from the transfected cells as follows. Cells were released from the plates by the addition of 2 ml trypsin-EDTA and incubation at 374C for 5 minutes, followed by gentle swirling. Released cells weri transferred to 50 ml conical tubes with 2 ml of fetal calf serum and centrifuged at 1000 rpm for 5 minutes to pellet the cells. 25 Cell pellets of no more than 1 gram were lysed with 20 ml of TRIZOL reagent (BRL), homogenized for 30 seconds, and then.extracted with 4 nl of chloroform. The tubes were centrifuged at 4000 rpm for 30 minutes and the aqueous phase was transferred to a new tube. RNA was precipitated by adding 10 nil isopropanol, mixing, and then centrifuging for 30 minutes at 4200 rpm. The RNA pellet was 30 washed with 10 ml of 70/a ethanol, briefly dried, and .then the pellet was - 78 resuspended in 0.5 ml TE buffer. Poly A+ RNA was prepared by using a commercially available mRNA purification kit (Pharmacia). After eluting poly A+ RNA from the column in 750 pl of TE buffer, the sample was then ethanol precipitated in two 1.5 ml microcentrifuge tubes by adding 40 pl of sample buffer - and 1 ml of ethanol and incubating overnight at -70*C, The cDNA inse-ts of positive clones were rescued by RT-PCR as follows, First strand synthesis~ was. performed using the SuperScriptTm preamplification system (BRL). A mixture containing 15 ptg of poly A+ RNA from selected-3T3 colonies and 15 l (2 pM) of vector primer (5'-A-A-T-C-C-G-A-T-G-C-C-C-A 3.0 C-Q-T-T-G-C-A-G-T-A-3'; SEQ ID NO: 35) was prepared and then incubated at 70*C for 10 minutes followed by equilibration at 50*C. A premixture containing 2.5 1 II 1OX buffer, 2.5 yl 25 mM MgCl 2 , 1.3 pl 10 mM dNTFs, and 2.5 pl 0.1 M dithiotbreitol was then added to the poly At RNA/primer mixture, after which 1.2 .Il of reverse transcriptase was added and the reaction incubated at 50*C for I 15 hour. The reaction was stopped by heating at 70*C for 15 minutes. Following first strand synthesis, RNA was digested with 1 pl RNase H at 37*C for 20 minutes. PCR was perftnned using Pfu polymerase (Perkin Elmer) as follows. In a total vohime of 100 pI, 2 pl of the first strand reaction was added to IX Pfu buffer, 0.5 pM each of the amplification primers (5'-A-A-A-A-T-C-T 20 T-A-G-A-C-C-G-A-C-G-A-C-T-G-T-G-T-T-T-3'; SEQ ID NO: 36; and. 5'-G-A G-T-C-T-C-C-G-C-A-G-C-C-T-T-T-T-G-A-G-G-3'; SEQ ID NO: 37), 0.2 mM dNTPs. 5% DMS,'and 2.5 units- of Pfu polymerase. The amplification reaction was performed at 95*C for 1 minute for 1 cycle; 95*C for 30 seconds, 66*C for 45 seconds, and 72 0 C for 30 cycles; and 72*C for 10 minutes for I cycle. PCR 25 products were purified by phenol/ chloroform extraction followed by ethanol precipitation, and then were digested with Not I and Sal I. Small digestion products and PCR primers were removed from the reaction using SizeSep 400 Spun columns. Clones identified in the'primary screen were subsequently analyzed in a 30 secondary secretion assay. This secondary screening technique utilized a vector - 79 containing a truncated placental alkaline phosphatase (FLAP), in which the native signal peptide had been removed, as a secretion reporter gene. Heterologous cDNA fragments were tested for the presence of signal peptide secretion sequences by inserting individual sequences immediately upstream of the 5 truncated PLAP gene and then transfecting COS7 cells with the test constructs. Inserted cDNA sequences encoding a signal peptide, when inserted in frame with the PLAP reporter sequence, lead to the formation of a fusion protein that can be secreted from the transfected cells. The PLAP reporter construct was generated as follows. .Human placenta 10. RNA was subjected to RT/PCR amplification under standard conditions to generate a DNA fragment encoding a truncated human PLAP protein. The RNA was transcribed by reverse transcriptase using oligo d(T) as priner, and then PCR amplified with PLAP specific primers to produce a DNA product which encodes a PLAP sequence corresponding to amino acids 22 to 536 (Millan, J. BiAL Chem. 15 261(7):3112-15 (1986)). The PLAP amplification primers (5'-A-C-T-G-G-C-G-G C-C-G-C-A-G-G-C-A-T-C-A-T-C-CC--A-GT-T-G-A-G-G-A-G-3'; SEQ ID NO: 38; and 5'-A-C-T-G-G-T-C-A-C-T-C-G-A-G-G-G-T-A-C-C-T-T-A-G-C-T-A-G C-C-C-C-C-G-G-G-3'; SEQ ID NO; 39) were designed to contain Not I and Kpn I restriction sites, respectively, in order to facilitate the ligation of the PLAP 20 fragment into the pcDNA3.1 vector (generating the vector pcDNA3.1/PLAP). The TmrlIl secondary library was constructed in pcDNA3.1/PLAP as follows. The cDNA inserts from clones in the primary Tril1 library were recovered from 3T3 cell colonies using the PCR amplification prjimers and conditions noted above. Recovered PCR products were then ligated into the 25 pcDNA3./PILAP vector which had been digested with the Xho I and Not I restriction enzymes. Ligation products were then transformed into the E. Coli strain DH10B to generate the secondary Tmrl1 library. To assay heterologous cDNA fragments for the presence of signal peptide secretion sequences individual colonies were first selected from a low density 3 0 plating of the secondary Trl 1 library on agar plates and placed into the wells of a - 80 standard 96-well plate containing bacterial growth medium. The- cultures were then grown to saturatin and plasmid DNA was prepared from each culture by standard procedures. Plasmid DNA prepared as- such was used to transfect COS7 cells as described below. 5 COS7 cells were seeded in 96-well flat-bottom plates at a concentration of 6000 cells per well in 200 p of a growth medium consisting of DMEM-HG, 10% fetaf bovine serum and IX PSG (penicillin,. streptomycin and gentamycin). Following seeding with COS7 cells, the plates were incubated for 24 hours at 37*C in a CO 2 incubator. The cells in each well were subsequently transfected 10 with 500. ng of plasmid DNA recovered from selected secondary Tmrll library clones using Superfect reagent (Qiagen). Transfection reactions were allowed to proceed for two hours, after which the transfected cells were washed with 200 pl of phenol red-free, serm-free DMEM'and then incubated with 100 pl of.phenol red-fiee, serum-free DMEM and glutamate for 24 hours at 37*C in a CO 2 is incubator. Following incubation, 100 pt of a solution containing 200 pM 4 methylumbelli-feryl phosphate (Molecular Probes) in IM diethanolamine, 10 mM homo-arginine, 1 mM MgC 2 , and I mg/nil BSA was added to each well, and incubation continued for 1 hour at 37*C. The product of the alkaline phosphatase reaction was then read in a fluorometer at 360/460 nm. 20 A single clone (tmrl1-00001-e9), yielding an increased fluorescence readout in the PLAY assay and possessing a computer-predicted signal peptide sequence and homology to the FGF family, was identified in the secondary Tmrll library screen. This clone was subsequently used as a probe to isolate a full length cDNA for murine FGF-like polypeptide from a mouse liver cDNA library. 25 A mouse liver full-length cDNA library was constructed using standard techniques. Essentially, oligo d(T) was used to prime first strand synthesis from mRNA isolated from regenerating mouse liver and full-length cDNA sequences were then.cloned into the pSPORT (Gibco BRL) vector using the Superscript Plasmid System for cDNA Synthesis and Plasmid Cloning (Gibco BRL).
-81 In a primary screen of the mouse liver cDNA library, 50,000 colonies were plated on LB/Ampicillin plates and then transferred to nitrocellulose filters. The filters were screened with a mixture of probes derived from clones identified in the kFGF signal trap and secretion assay screenings. This pool of probes included s a 339 bp Not I-Xba I fragment isolated'from the tmrll-00001-e9 clone, The filters were first prehybridized for 2 hours at 42 0 C in a hybridization solution consisting of 50% formamide, 5X Denhardt's; 5X SSC, 0.5% SDS, and .100. pg/ml salmon sperm DNA. Following prehybridization, the filters.were incubated with the "P dCTP labeled probes overnight at 42'C in fresh hybridization solution. The filters 10 were then. washed with 0.1X SSC/0.1% SDS at 65 0 C and were subsequently analyzed by autoradiography- overnight at -80"C., From this initial screen, 92 positive clones were identified. The positive clones isolated in the primary screen were pooled and rescreened with the 339 bp tmrll-00001-e9 fragment alone. In this secondary 1- screening, 6000 colonies derived from the clone pool were plated on LB/Ampicillin plates and then transferred to nitrocellulose filters. The hybridization conditions used in the secondary screen were the same as those used for the primary screen. Three individual clones (lE, 1E-4, and 1E-6) were identified in the secondary screen. 20 Restridtion digestion of the three individual clones identified in the secondary screen indicated that each contained a 1.6 kb insert. Primers corresponding to the 5' (5'-T-G-G-A-A-T-G-A-T-G-A-G-A-T-C-T-A-G-A-G 3'; SEQ ID NO;: 7) and 3' (5-C-T-A-G-A-T-T-C-A-G-G-A-A-G-A-G-T-C-A-3'; SEQ ID NO: 8) ends of the coding sequence encoded by this insert were designed 25 following partial sequence analysis of clone 1E. These primers were used in a polymerase chain reaction (PCR) amplification of clone lE plasmid DNA. The amplification reaction was performed at 944C for 1 minute for I cycle; 94"C for 15 seconds and 65"C for 1.5 minutes for 35 cycles; and 72*C for 10 minutes for 1 cycle. A PCR product of approximately 650 bp was obtained following 30 amplification.
-82 Following purification from a 1% agarose gel using a Qiagen gel extraction kit, the PCR product was sequenced. Sequence analysis of this amplification product indicated that the cDNA clones from which the PCR primers were derived contained a gene comprising a 630 bp open reading frame 5 encoding a protein of 210 amino acids. Figure 1 illustrates the nucleotide sequence of the urine FGFelike gene (SEQ ID NO: 3) and the deduced amino acid sequence of murine FGF-like protein (SEQ ID NO: 4). Subsequent sequence analysis of clone IE also established that the open reading frame of the cDNA clones identified in .the secondary screening was interrupted by two intron 1x0 sequences., Computer analysis using the FASTA program of the Swissprot database indicated that this open reading frame encoded a polypeptide that is most closely related (39% identical) to FGF-6 (Figures 3A-3D). Computer analysis, using the SIGNALP program (Center for Biological Sequence Analysis, The Technical University of Denmark), also indicated that the 15 murine FGF-like polypeptide possessed a potential signal peptide at its amino terminus (M-E-W-M-R-S-R-V-G-T-L-G-L-W-V-R-L-L-L-A-V-F-L-L-G-V-Y-Q A; SEQ ID NO: 40; as underlined in Figure 1). The initial translation product of murine FGF-like polypeptide has a calculated molecular weight of 23,237, not including possible post-translational modifications. After removal of the 20 predicted 29 amino acid signal peptide sequence, the remaining predicted mature protein of 181 amino acids has a calculated molecular weight of 19,876. No predicted N-linked glycosylation sites were identified in. the protein and the protein does not possess any dibasic protease processing sites. 25 Example 2: Cloning of the Human FGF-like Polypeptide Gene Generally, materials and methods as described in Sambrook et al. supra -were used to clone and analyze the gene encoding human FGF-like polypeptide. Sequences encoding the human FGF-like polypeptide were isolated by screening a human cDNA library with a probe derived from the murine FGF-like 30 polypeptide gene. A 460 bp probe was generated by PCR amplification of urine -83 FGF-like polypeptide cDNA using the following primers: 5'-T-G-G-A-A-T-G-Q A-T-G-A-G-A-T-C-T-A-G-A-G-3'; SEQ ID NO: 7) and 3' (5'-C-A-T-T-G-C-G G-C-C-G-C-T-C-A-A-G-A-T-G-C-A-A-A-A-C-G-C-A-G-T-G-3'; SEQ ID NO: 9) in reactions containing "P-dCTI. The amplification reaction was performed at 5 - 94"C for I minute for 1 cycle; 94 0 C for 15 seconds and 651C for 1.5 minutes for 35 cycles; and 72"C for 10 minutes for 1 cycle The 460 bp murine probe was used to isolate a full-length cDNA for human FGF-like polypeptide from a human liver cDNA library. A human liver l'll-length cDNA library was constructed using standard techniques. Essentially, 10 oligo d() was used to prima first strand synthesis from mRNA obtained from Clontech (Palo Alto, CA) and full-length cDNA sequences were then cloned into the pSPORT (Gibco BRL) vector using the Superscript Plasmid System for cDNA Synthesis and Plasmid Cloning (Gibco BRL). In a primary screen of the human liver cDNA library, 550,000 colonies 15 were plated on LB/Ampicillin plates and then transferred to nitrocellulose. filters. The filters were first prehybridized for 30 minutes at 60*C in ExpressHyb solution (Clontech), and then were incubated with the nP-dCTP labeled urine FGF-like cDNA probe ovemight at 60"C in fresh ExpressHyb solution. Following hybridization, the filters were washed twice for 30 minutes at room temperature in 20 2X SSC/0.1% SDS, twice for 30 minutes at 60*C in IX SSC/0.1% SDS, twice for 30 minutes at 65*C in 0.1XSSC/0.1% SDS, and then the filters were analyzed by autoradiography ovemight at -80"C. Positive clones identified in the primary screen were then rescreened, and four independent clones were recovered following the secondary screen. Plasmid DNA for these four clones was prepared 25 and sequenced Sequence analysis indicated that the four cloeies contained inserts of either 1.2 kb or 1.8 kb. The 1.2 kb cDNA insert contained a gene comprising a 627 bp open reading frame encoding a protein of 209 amim acids. Figures 2A-2B illustrate the nucleotide sequence of the humau FGF-like gene (SEQ ID NO: 1) 30 and the deduced amino acid sequence of human FPGF-like protein (SEQ ID NO: -- 84 2). While the 1.8 kb cDNA insert contained the same open reading frame as encoded by the 1.2 kb insert, the open reading frane of this insert was additionally interrupted by an intron corresponding in location to the second intron found in some of the urine cDNA clones isolated in Example 1. 5 Figures 3A-3D illustrate the amino acid sequence alignment of human FGF-like protein, urine FGF-like protein, and other members of the FGF family. The human FGF-like polypeptide is 76% identical to the murine FGF-like protein. Computer analysis, using the SIGNALP program (Center for Biological Sequence Analysis, The Technical University of Denmark). indicated that the human FGF 10 like polypeptide also possessed a potential signal peptide at its amino terminus (M-D-S-D-E-G-F-E-H-S-GL-W-V-S-V-L-A-G-L-L-L-G-A-C-Q-A; SEQ ID NO: 41; as underlined in Figures 2A-2B). The initial translation product of human FGF-like polypeptide has a calculated molecular weight of 22,284, not including possible post-translational modifications. After removal of the is predicted 28 amino acid signal peptide sequence, the remaining predicted mature protein of 181 amino acids has a calculated rolecular weight of 19,395. No predicted N-linked glycosylation sites were identified in the protein and the protein does not possess any dibasic protease processing sites. 20 Example 3: FGF-like mR4NA Expression Expression of murine FGF-like tnRNA was' examined on a murine multiple tissue Northem blot (Clontech) using a "P-dCTP labeled murine FGF like cDNA probe. The probe consisted of a 391 bp fragment isolated from the tnr11-00001-e9 clone by restriction digestion with Xba I and Not L The blot was 25 first prehybridized for 2 hours at 424C in a hybridization- solution consisting of 50% formamide, 5X Denhardt's, 6X SSC, 0.5% SDS, and 100 fig/mI salmon iperm DNA. Following prehybridization, the filters were incubated with the 3
P
dCTP labeled urine FGF-like cDNA probe overnight at 42CC in fresh hybridization solution. The filters were then washed with 0.1X SSC/0. 1% SDS at 30 65"C and were subsequently analyzed by autoradiography overnight at -80 0
C.
-85 Two transcripts, of approximately 135 kb and 1.8 kb, were detected in murine liver (Figure 4A). The 1.35 kb transcript showed the predominant expression. Expression of human FGF-like mRNA was examined on a Human RNA Master BlotTh (Clontech) and a human multiple tissue Northern blot (Clontech) B.- using a "P-dCTP labeled human FGF-like cDNA probe. The probe consisted of a 660 bp PCR product derived from the hmnan FGF-like protein coding region using the primers: 5'-C-T-A-C-TA-A-A-G-C-T-T-C-C-A-C-C-A-TG-G-A-C-T C-G-G-A-C-G-A-G-A-C-C-G-3'; SEQ I) NO: 12; and 5'A-T-T-C-A-T-G-C-G G-C-C-G-C-G-G-A-A-G-C-G-T-A-G-C-T-G--G-G-C-T-T-C-3'; SEQ ID NO: 10 13, and the human cDNA clone described above as a template. The amplification reaction was performed at 94*C for 1 minute for I Cycle; 94*C for 15 seconds, 60*C for 15 seconds, and 72*C fbr 1 minute for 35 cycles; and 72*C for 10 minutes for 1 cycle, The blots were first prehybridized for I hour at 65"C'in ExpressHyb solution (Clontech), and then were incubated with the "P-dCTP 5 labeled human FGF-like cDNA probe overnight at 65*C in fresh ExpressfIyb solution. Following hybridization, the filters were washed twice for 30 minutes at room temperature in 2X SSC/0.1% SDS, twice for 30 minutes at 65*C in 0.lX SSC/ 0.1% SDS, and then the filters were. analyzed by autoradiography overnight at -80CC. Two transcripts, of approximately 1.2 kb and 2 kb, were detected in 20 human liver on the multiple tissue Northern blot 9 (Figure 4B). Strong expression in adult liver and weak expression in lung and fetal liver was detected on the Human RNA Master BlotTA (Figure 4C). Example 4: FGF-like mRNA in situ analysis 25 In siru hybridization was performed with a 648 bp antisense RNA probe spanning the coding region of murine FGF-like polypeptide. The probe was transcribed from a linearized pCR2.1 TOPO plasmid containing the FGF-like cDNA insert using T7 RNA polymerase and 33 P-UTP. A panel of normal embryonic and adult mouse tissues were fixed in 4% 30 parafomaldehyde, embedded in paraffin, and iectioned at 5 gm. Pior to in situ - 86 hybridization, tissues were permeabilized with 0.2M HCI, followed by digestion with Proteinase K, and acetylation with triethanolamine and acetic anhydyide. Sections were hybridized with the "P-labeled riboprobe -overnight at 55 0 C,'then subjected to a high stringency wash in 0.1 X SSC at 60*C. Slides were dipped in 5 Kodak NTB2. emulsion, exposed at 4 0 C for 2-3 weeks, developed, and counterstained with hematoxylin and cosin. Sections were examined with darkfield and standard illunmination to allow simultaneous evaluation of tissue morphology and hybridization signal. The strongest overall expression was noted in the pancreas with a strong 1o signal being detected over the islets and a lower, more diffuse signal over the acinar portion of the pancreas. The liver displayed a moderate level of diffise signal, indicative of moderate hepatocellular expression of FGF-like polypeptide. Significant signal was also present over spermatogonia within the seminiferous tubules in the testis and over cells in the thymic medulla. A low level of diffuse 15 signal was detected in kidney, spleen, pituitary, and white and brown adipose tissue. Example 5: Production of FGF-like Polypeptides in Mammalian Cells Both the' human and mnuine FGF-like polypeptides were expressed as 20 fusion proteins with a human immunoglobulin IgG heavy chain Fc region at their. carboxyl terminus. Template DNA sequences encoding human or marine FGF like polypeptide were amplified by PCR using primers corresponding to the 5' and 3'ends of the sequence (Table 11). The resulting PCR products corresponded to the coding region of either the human or urine FGF-like polypeptide, minus the 25 translation teribination codon.- In addition, the primers were designed to incorporate a Hind IIM restriction.endonuclease site at the 5' end of the PCX product and a Not I site at the 3' end of the product. Table II 30 Primers Used in Recombinant Protein Expression -87 Murine 5' 5' -CTACTAAAGCTTCCACCATGGAATGGATGAGATCTAG-3' (SEQ ID NO: 10) Murine 3' V'ATTCATGCGGCCGCGGACGCATAGCTGGGGCTr-3' (SEQ ID NO, 11) Human 5' 5' -CTACTAAAGCTTCCACCATrGACTCGACGA3ACCG-3' (SEQ ID NO: 12) Human 3' 5'-ATTCATGCGGCCGCGGAAGCGTAGCTGGGGCTTC- 3 ' (SEQ ID NO: 13) The human and murine FGF-like protein-Fc fusion constructs were generated by first cleaving the PCR products with Hind MI and Not I and then ligating the fragments in frame to a DNA fragment encoding the H1c chain of human immunoglobulin IgG. The FGF-like protein-Fc insert was then lighted lo into the pCEP4 mammalian expression vector (Invitrogen). These ligations were transformed into the K cohl strain DH1O by ele'troporation and transformants selected for ampicillin resistance. Following sequence analysis of selected transformants, large-scale plasmid stocks were prepared for tissue culture transfection. Plasmid DNA for selected ampicillin resistant colonies was prepared is and sequenced to confirm that the clone contained the desired insert. To conduct functional studies on FGF-like protein, human or mine FGF like protein-Fc fusion expression constructs 'were introduced into 293-EBNA (Invitrogen) cells using SuperFect-r transfection reagent (Qiagen). The conditioned medium was harvested 48 hours after transient transfection. Western 20 blot analysis, using an anti-humai Fc antibody, confirmed that the conditioned media contained human or marine FGF-like/Fe fusion polypeptides. Conditioned medium was purified by affinity chromatography as described below. The medium was first passed through a 0.2 ym fiten Protein A colums (Pharmacia) were equilibrated with Ix unoPure Gentle Binding Buffer (Pierce, 25 Rockford, IL), and then loaded with the filtered medium. The column was washed with ImimunoPure Gentle Binding Buffer until the absorbance at 280 mm reached a baseline. FGF-like/Fc protein was eluted from the column with ImmunoPure Gentle Binding Buffer. Fractions containing FGF-like/Fc protein were pooled, dialyzed in Tris-buffered saline (TBS) followed by Phosphate 30 buffered *saline (PBS), and stored at 4 0 C. Gel electrophoretic analysis confirmed - 88 that the pooled factions contained a purified protein of the expected molecular weight of about 60 kD. Example 6: Production and Analysis of Transgenic Mice Overexpressing FGF ~ like. Polvetide Transgenic mice overexpressing mouse FGF-like polypeptide (SEQ ID NO: 4) from the human apolipoprotein E promoter were generated as previously described (see Simonet et al., 1997, Cell 89:309-19). Seven mice (four males and three females) which were transgenic for the urine FGF-like gene (SEQ ID NO: 10 3) and five non-transgenie littennates (two males and thee females) underwent necropsy and pathological analysis at &8 weeks of age. All of the mice were injected with 50 mg/kg of BrdU one hour prior to harvest, then were radiographed and sacrificed. Body and selected organ weights were measured, blood was drawn for hematology and serum chemistries, and organs were harvested for 15 histological analysis and BrdU labeling. 1l of the transgenic mice were under 20 gm body weight while all of the non-transgenic mice were over 20 gin body weight (p<0.0001). In addition, traUsgcnic mice. bad statistically significantly lower liver (p-0.001 1) and spleen weights (p=0.00 3 9 ) and a higher thynic (p=0.01 18) weight (as a percent of body 20 weight) than their non-transgenic counterparts. For the transgenic mice, body weight was 67% of wild type, Liver, spleen, and thymus weights, as a percentage of body weight, were, respectively, 85%, 63% and 170% of wild type. All female transgenic mice also had small uteri and ovidncts and ovaries that lacked corpora lutea and exhibited little follicular development. In summary, transgenic mice had 25 a phenotype that is best characterized as stunted growth with small livers, spleens, and poorly developed ovaries, and enlarged (probably not involuted) thymuses. These changes are most consistent with overexpression of FGF-like polypeptide in the transgenics causing inhibited or delayed maturation in comparison with their age-matched non-transgenid littermates. 30 -89 Examule 7: Analysis of One-Year-Old FGF-like Polypentide Transgenic Mice Necropsy and pathological analysis was performed on one-year-old FGF like polypeptide transgenics (five males and five females) and non-transgenic (three males and two females) litterniates that had been produced in the 5 experiments described in Example 6. EGF-like polypeptide transgenics continued to demonstrate an abnormal phenotype generally characterized as inhibited or delayed maturation. These observed effects included reduced body weight (48% of wild type) and, in the females, poorly developed ovaries with lack of significant follicular development. Liver, spleen, and tbymus weights as a percentage of 10 body weight had normalized to that found in the non-transgenic litternates. Several of the one-year-old non-trausgenic control mice were found to be obese, and at least one of the controls exhibited changes consistent with the development of type II diabetes. However, none of the one-year-old transgenic mice were obese or showed any evidence of developing diabetes. Thus, it appears that FGF 15 like polypeptide transgenics do not develop at least some of the age related changes commonly seen in mice as they age and, indeed, that the FGF-like gene of this invention may help retard the aging process. These findings are significant and support the conclusion that the FGF-like polynucleotides and polypeptides of the present invention may be useful for the 20 treatment or diagnosis of age-related diseases,.disorders, or conditions. By way of illustration, such diseases, disorders or conditions may include, without limitation, atherosclerosis, hypercholesterolemia, diabetes, obesity, stroke, osteoporosis, osteoarthritis, degenerative joint disease, muscle atrophy, sarcopenia, decreased lean body mass, baldness, wrinkles, increased fatigue,, decreased stamina, 25 decreased cardiac function, immune system dysfunction, cancer, Parkinsbn's disease, senile dementia, Alzheimer's disease, and decreased cognitive function. More generally, the molecules of the present invention may be applicable for enhancing or increasing life-span.
-90 While the present invention has been described in terms of the preferred embodiments, it is understood that variations and modifications will occur to those skilled in the art. Therefore, it is intended that the appended claims cover all such equivalent variations that come within the scope of the invention as claimed. 5 Throughout the specification and claims, unless the context requires otherwise, the word "comprise" or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers. Each document, reference, patent application or patent cited in this text is 10 expressly incorporated herein in their entirety by reference, which means that it should be read and considered by the reader as part of this text. That the document, reference, patent application or patent cited in this text is not repeated in this text is merely for reasons of conciseness. Reference to cited material or information contained in the text should not be 15 understood as a concession that the material or information was part of the common general knowledge or was known in Australia or any other country,

Claims (22)

1. A recombinant host cell comprising an isolated nucleic acid molecule having a nucleotide sequence that encodes the polypeptide of SEQ ID NO: 5.
2. The recombinant host cell of claim 1 that is a eukaryotic cell. 5
3. The recombinant host cell of claim 1 that is a prokaryotic cell.
4. A process of producing a polypeptide encoded by an isolated nucleic acid molecule having a nucleotide sequence that encodes the polypeptide of SEQ ID NO: 5 comprising, culturing a recombinant host cell comprising the nucleic acid molecule having a nucleotide sequence that encodes the polypeptide of SEQ ID NO: 5 under suitable 10 conditions to express the polypeptide.
5. The process of claim 4, further comprising recovering the polypeptide from the culture.
6. The process of claim 4, wherein the nucleic acid molecule comprises promoter DNA other than the promoter DNA for a native FGF-like gene operatively linked to the is nucleic acid molecule.
7. A vector comprising an isolated nucleic acid molecule having a nucleotide sequence that encodes the polypeptide of SEQ ID NO: 5.
8. A recombinant host cell comprising the vector of claim 7.
9. The recombinant host cell of claim 8 that is a eukaryotic cell. 20
10. The recombinant host cell of claim 8 that is a prokaryotic cell,
11. A process for determining whether a compound inhibits FOP-like polypeptide production comprising exposing a recombinant host cell according to claim 1 to the compound, and measuring FGF-like polypeptide production in said cell.
12. A method of identifying a modulator of FGF-like polypeptide activity comprising: 25 (a) incubating a polypeptide comprising the amino acid sequence of SEQ ID NO: 2, the amino acid sequence of SEQ ID NO: 5, or an amino acid sequence that is at least 95% identical to the amino acid sequence of either SEQ ID NO: 2 or SEQ ID NO: 5, with a test molecule, under conditions that permit interaction of the polypeptide with the test molecule; and 30 (b) measuring FGF-like polypeptide activity to identify a test molecule that modulates FGF-like polypeptide activity.
13. A method of identifying a modulator of FGF-like polypeptide activity comprising: - 92 (a) transforming a host cell with a vector comprising the nucleotide sequence of SEQ ID NO: 1, the DNA insert in ATCC Deposit No. PTA-626, or a nucleotide sequence that is 95% identical to the nucleotide sequence of either SEQ ID NO: 1 or the DNA insert in ATCC Deposit No. PTA-626; 5 (b) incubating the host cell with a test molecule, under conditions that permit interaction of the polypeptide encoded by the nucleotide sequence of either SEQ ID NO: 1 or the DNA insert in ATCC Deposit No. PTA-626 with the test molecule; and (c) measuring FOF-like polypeptide activity to identify a test molecule that modulates FGF-like polypeptide activity. 10
14, The method of either of claims 12 or 13, wherein the test molecule that modulates FPGF-like polypeptide activity is an FGF-like polypeptide agonist.
15. The method of either of claims 12 or 13, wherein the test molecule that modulates FGF-like polypeptide activity is an FGF-like polypeptide antagonist.
16. The method of either of claims 12 or 13, wherein the test molecule specifically 15 binds the FGF-like polypeptide.
17. The method of claim 16, wherein the test molecule binds to the polypeptide with a KD of at least about Ix 10-6 to I x 10-10 M.
18. The method of either of claims 12 or 13, wherein the test molecule indirectly interacts with the FGF-like polypeptide. 20
19. The method of claim 12, wherein test molecule that modulates FGF-like polypeptide activity is identified in a cell-based binding assay, membrane-binding assay, solution-phase assay, or immunoassay.
20. The method of claim 13, wherein the host cell is a mammalian cell.
21. The method of claim 13, wherein the host cell is in a non-human mammal. 25
22. The method of claim 21, wherein the non-human mammal is a mouse.
AU2008201780A 1999-09-07 2008-04-22 Fibroblast growth factor-like polypeptides Ceased AU2008201780B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2008201780A AU2008201780B2 (en) 1999-09-07 2008-04-22 Fibroblast growth factor-like polypeptides

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US09391861 1999-09-07
US09644052 2000-08-23
AU2004242465A AU2004242465C1 (en) 1999-09-07 2004-12-24 Fibroblast Growth Factor-Like Polypeptides
AU2008201780A AU2008201780B2 (en) 1999-09-07 2008-04-22 Fibroblast growth factor-like polypeptides

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2004242465A Division AU2004242465C1 (en) 1999-09-07 2004-12-24 Fibroblast Growth Factor-Like Polypeptides

Publications (2)

Publication Number Publication Date
AU2008201780A1 AU2008201780A1 (en) 2008-05-15
AU2008201780B2 true AU2008201780B2 (en) 2012-05-17

Family

ID=39409746

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2008201780A Ceased AU2008201780B2 (en) 1999-09-07 2008-04-22 Fibroblast growth factor-like polypeptides

Country Status (1)

Country Link
AU (1) AU2008201780B2 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101917854B1 (en) * 2017-08-24 2018-11-12 한국콜마주식회사 Peptides having capacity of binding to cell receptor and cosmetic composition comprising the same

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU3755900A (en) * 1999-03-15 2000-10-04 Chiron Corporation Use of recombinant gene delivery vectors for treating or preventing diseases of the eye

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU3755900A (en) * 1999-03-15 2000-10-04 Chiron Corporation Use of recombinant gene delivery vectors for treating or preventing diseases of the eye

Also Published As

Publication number Publication date
AU2008201780A1 (en) 2008-05-15

Similar Documents

Publication Publication Date Title
EP1218509B1 (en) Fibroblast growth factor-like polypeptides
US7727742B2 (en) Nucleic acid molecules encoding fibroblast growth factor-like polypeptides
EP1268793A2 (en) Fibroblast growth factor receptor-like molecules and uses thereof
AU2008201780B2 (en) Fibroblast growth factor-like polypeptides
AU2004242465C1 (en) Fibroblast Growth Factor-Like Polypeptides

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired