CA2320515A1 - Therapeutic uses of keratinocyte growth factor-2 - Google Patents

Therapeutic uses of keratinocyte growth factor-2 Download PDF

Info

Publication number
CA2320515A1
CA2320515A1 CA002320515A CA2320515A CA2320515A1 CA 2320515 A1 CA2320515 A1 CA 2320515A1 CA 002320515 A CA002320515 A CA 002320515A CA 2320515 A CA2320515 A CA 2320515A CA 2320515 A1 CA2320515 A1 CA 2320515A1
Authority
CA
Canada
Prior art keywords
polypeptide
seq
kgf
amino acid
ser
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002320515A
Other languages
French (fr)
Inventor
Pablo Jimenez
Mark A. Rampy
Donna Mendrick
Deborah Russell
Arthur Louie
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.)
Human Genome Sciences Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2320515A1 publication Critical patent/CA2320515A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/50Fibroblast growth factors [FGF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/10Drugs for disorders of the urinary system of the bladder
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Abstract

The present invention relates to the administration of Keratinocyte Growth Factor-2 (KGF-2) to stimulate proliferation of platelets and to increase levels of fibrinogen, albumin, globulin and total serum protein. Further, the present invention relates to administering KGF-2 to protect or treat the bladder and prostate. Moreover, the present invention relates to administering KGF-2 to stimulate growth of nasal, oral, and esophageal mucosa, lacrimal glands, salivary glands and Goblet cells.

Description

Therapeutic Uses of Keratinocyte Growth Factor-2 Field of the Invention The present invention relates to the administration of Keratinocyte Growth Factor-2 (KGF-2} to increase levels of platelets, fibrinogen, albumin, globulin and total serum protein. Further, the present invention relates to administering KGF-2 to protect or treat the bladder and prostate. Moreover, the present invention relates to administering KGF-2 to stimulate growth of nasal, oral, and esophageal mucosa, lacrimal glands, salivary glands and Goblet cells.
Background of the Invention Thrombocytopenia is a condition in which there is an abnormally small number of platelets in the circulating blood (Stedman's Medical Dictionary, 26th edition, Marj ory Spraycar, Editor ( 1995). Thrombocytopenia results from various causes, but ultimately occurs when platelets are destroyed, sequestered in the body, or not produced. The differential diagnosis of thrombocytopenia is extensive and complex, and there is a significant overlap among disorders (Doyle B, and Porter D.L. A.A.C.N. Clin. Issues 8: 4b9-480 (1997).
Thrombocytopenia may be caused by a variety of mechanisms including, but not limited to, drug induced hypersensitivity, idiopathic thrombocytopenia purpura (ITP), posttransfusion purpura, neonatal thrombocytopenia, bone marrow deficiencies identified with metastatic tumors to the bone, aplastic anemia, myelofibrosis, acute and monocytic leukemia, microangiopathic hemolytic anemia which includes disseminated intravascular coagulation (DIC), thrombotic thrombocytopenic purpura (TTP), hemolytic-uremic syndrome, prosthetic valve hemolytic syndrome, cancer chemotherapy, Zieve's syndrome, sepsis, HELLP
preclamptic syndrome, megaloblastic anemia due to B21 and folic acid deficiency, infections such as peritonitis (without septicemia), congenital rubella syndrome, HIV-1 virus infections, Epstein-Barr infectious mononucleosis, rheumatoid-collagen diseases such as systemic lupus, hypertension of pregnancy associated with preclampsias, thyrotoxicosis and uremia (Clinical guide to laboratory tests. (3rd ed). Philadelphia, W.B. Saunders Company,1995; Clinical LaboratoryMedicine. Clinical application oflaboratory data. (6th ed.) St.
Louis, Mosby, 1995).
Fibrinogen is an abundant plasma glycoprotein that is synthesized in the liver. Thrombin sequentially cleaves fibrinopeptides A and B from the a and p chains of fibrinogen to produce fibrin monomer, which then polymerizes to form a fibrin clot which is the final major step in the coagulation process.
Mutations have been identified which alter the release of fibrinopeptides from the a and ~i chains of fibrinogen, the rate of polymerization of fibrin monomers, and the sites for fibrin cross-linking. This mutations lead to dysfibrinogenemias which are almost always inherited as autosomal dominant traits. Patients with afibrinogenemia, who have no detectable fibrinogen in plasma or platelets, may have infrequent, mild spontaneous bleeding episodes. (Harrison's Principles of Internal Medicine l lth edition Eugene Braunwald et. al., Editors (1987)).
Hypofibrinogenemia refers to a condition in which there is an abnormally low concentration of fibrinogen in the circulating blood plasma (Stedman's Medical Dictionary, 26th edition, Marjory Spraycar, Editor (1995)).
Hypofibrinogenemia may be caused by a variety of conditions or afflictions including, but not limited to, abnormal hepatic synthesis such as that associated with acute hepatitis or cirrhosis, and disseminated intravascular coagulation (DIC).
Albumin, the major serum protein, is considered to be responsible for tenance of normal serum colloid osmotic pressure, transport of certain hormones and maintaining an endogenous source of amino acids (Buehler, B.A.
Ann. Clin. Lab. Sci. 8: 283-286 (1978)). Hypoalbuminemia is a condition in which there is an abnormally low concentration of albumin in the circulating blood. The serum albumin level is one of several clinical parameters of the status of general health. There is a marked correlation between low albumin levels and the incidence of morbidity and mortality in hospitalized patients. Therefore, it is not surprising to find that hypoalbuminemia is a common finding among hospitalized patients. Hypoalbuminemia is known to be associated with delayed wound healing. The hypoalbuminemic state interferes with the normal functioning of the gastrointestinal tract. Qualitative changes in the albumin molecule which occur in renal disease may damage the nephron. Low serum albumin levels may adversely affect the coagulation system {Doweiko, J.P., and Nompleggi, D.J. J.P.E.N. J. Parenter. Enteral. Nutr. 15: 476-483 (1991)).
Hypoalbuminemia can be caused by a variety of ai~lictions or conditions including, but not limited to, hemorrhages, burns, exudates, rheumatic diseases, granulomatous processes, most bacterial infections, viral infections accompanied by tissue destruction, tissue necrosis, vasculitis, ulcerative bowel disease, serositis, subacute bacterial endocarditis, parasitic infestations, acute and chronic liver disease, amyloidosis, malnutrition, malignancy, congestive heart failure, constrictive pericarditis, cardiac valvular disease, nephrotic syndrome, trauma and crush injuries, gastrointestinal and lymphatic fistulae, and protein-losing gastroenteropathies (Clinical guide to laboratory tests. (3rd ed).
Philadelphia, W.B. Saunders Company, 1995; Clinical Laboratory Medicine. Clinical application of laboratory data. (6th ed.) St. Louis, Mosby, 1995).
Globulin is the name for a family of proteins precipitated from plasma or serum by half saturation with ammonium sulphate. Globulins may be fractionated by solubility, electrophoresis, ultracentrifugation, and other separation methods into many subgroups, the main subgroups being a-, (3-, and y-globulins. These differ with respect to associated lipids or carbohydrates and in their content of many physiologically important factors. Globulins include immunoglobulins in the (i, and y fractions, lipoproteins in the a and p fractions, gluco- or mucoproteins (orosomucoid, haptoglobulin), and metal binding and metal transporting proteins (such as transferrin, siderophilin, ceruloplasmin).
Other substances found in globulin fractions are: macroglobulin, plasminogen, prothrombin, euglobulin, antihemophilic globulin, fibrinogen, and cryoglobulin (Stedman's Medical Dictionary, 26th edition, Marjory Spraycar, Editor (1995)).

Certain reasonably predictable changes take place in plasma protein levels in response to acute illness. Hypoglobulinemia refers to an abnormally low concentration of globulin in the circulating plasma. Hypoglobulinemia may result from a variety of conditions or afflictions including, but not limited to, alpha-1 antityrpsin deficiencies, severe liver disease, estrogen therapy, megaloblastic anemia, hypogamrnaglobulinemia and aggammaglobulinemia (Clinical guide to laboratory tests. (3rd ed). Philadelphia, W.B. Saunders Company,1995; Clinical LaboratoryMedicine. Clinical application oflaboratory data. (6th ed.) St.
Louis, Mosby, 1995).
A decrease in total serum protein is associated with protein loss (protein-losing gastroenteropathies, acute burns, nephrotic syndrome) and decreased synthesis ofprotein (chronic liver disease, malabsorption syndrome, malnutrition, and agammaglobulinemia).
Hemorrhagic cystitis is a syndrome associated with certain disease states as well as exposure to drugs, viruses, and toxins. It manifests as diffuse bleeding ofthe endothelial lining ofthe bladder. Treatment includes intravesical, systemic, and nonphartnacologic therapies {West, N.J. Pharmacotherapy 17: 696-706 (1997)).
Loss of the ability to produce adequate amounts of saliva and tears is a maj or clinical problem at~ecting millions of people and there are few therapeutic options for these sufferers. Patients with xerostomia, or dry mouth, have difficulty swallowing, have painful cracks in their mouths, and experience a decrease in their ability to taste. This condition may be caused by Sjogren's syndrome, as a secondary event to radiation used with patients with head and neck tumors, and to drugs. Patients with Sjogren's syndrome sometimes have keratoconjunctivitis sicca or dry eye. This condition may be caused by damage to the lacrimal gland due to Sjogren's syndrome, sarcoidosis, aging, HIV
infection, burns, etc. Patients experience irritation, blurring of vision, burning, pain, and increased risk of infections. Millions of patients suffer with Sjogren's syndrome and therapy is sub-optimal.

As is the case with xerostomia, patients with keratoconjuactivitis sicca have few therapeutic options. At this time there are approximately 10 million patients in the US which require artificial tear preparations (temp, M.A, Adv.
Exp. Med. Biol. 438:791-803 (1998)). There are no treatments available at this time to stimulate replacement of the cells in the salivary and lacrimal gland cells.
Sinus infections usually occur in the setting of upper respiratory tract infections, allergies, or anatomic defects (within the sinuses or nasal septum) and may lead to symptoms of headache, facial pain, fever, and purulent rhinorrhea.
(Evens KL. Drugs 56( 1 ): 59-71 ( 1998)). The symptoms of nasal allergy and chronic ethmoid sinusitis overlap and treatment failure in allergic patients may suggest possible chronic sinusitis. Chronic ethmoidal sinusitis may be the leading cause of rhinorrhea and nasal obstruction in patients with perennial allergies.
(Bertrand et al., Acta Otorhinolaryngol Belg 51(4):227-237 (1997)). Acute sinusitis tends to occur in patients with a history of rhinitis, which may be allergic or non-allergic in origin. Patients with anatomic variants (Evens KL. Drugs 56(1): 59-71 (1998)) and cystic fibrosis (Brihaye et al., Acta Otorhinolaryngol Belg 51(4):323-337 (1997); Kamsey et al., J. Allergy Clin. Immunol.
90(3Pt2):547-552 (1992); Davidson et al., Laryngoscope 105(4Pt1):354-358 (1995); Jones etal'., Int. J. Pediatr. Otorhinolaryngol. 28(1):25-32 (1993)) are also at high risk for developing sinusitis. In particular, it is known that occlusion of the sinus ostia starts the cycle of events, which lead to and sustain sinusitis.
(Reilly J.S. Otolaryngology Head and Neck Surgery 103(5):856-862(1990)).
The goals of treatment for both acute and chronic sinusitis are to control the rhinitis, improve ventilation to the sinuses, and to improve the function of the sinuses for clearance of secretions. (Evens KL. Drugs 56(1): 59-71 (1998}).
Surgical treatment of the nasal sinuses is only considered when medical measures fail to control the sinusitis. The goals of treatment are identical:
to improve ventilation and to facilitate or restore sinus drainage. An additional goal is that surgical treatment may sometimes improve the penetration oftopical drugs into the sinus. Up to 250,000 procedures are performed each year in the United States. However, in the course of performing sinus surgery, the mucosal surface is stripped away or damaged. In cases where the surgery has been extensive, the underlying bone within the sinus may be ex~sed for periods of up to 6 months before the mucosa is fully reconstituted and recovery of ciliary density may require up to 2 years to achieve. Delayed re-epithelialization of the nasal sinuses following surgery is believed to be associated with increased risk of infection, scarring (subepithelial fibrosis) leading to recurrent disease, and cyst formation.
After 2 years of follow up approximately 2-5% of patients will have recurrent disease and up to 15% may require surgical revision. (Evans KL. Drugs SG( 1 ):
59-71 ( 1998)).
Thus, there is clearly a need in the art for therapeutic proteins capable of increasing levels of platelets, fibrinogen, albumin, globulin and total serum protein. Further, there is a need for therapies capable of treating or protecting against damage caused by cystitis. Moreover, there is a need for stimulating the proliferation of cells in the salivary and iacrimal glands; and stimulating re-epithelialization of the sinuses and the growth of nasal mucosa in the nasal sinuses after surgery.
Summary of the Invention The present invention provides isolated nucleic acid molecules comprising a polynucleotide encoding the keratinocyte growth factor (KGF-2) having the amino acid sequence is shown in Figure 1 [SEQ ID N0:2] or the amino acid sequence encoded by the cDNA clone deposited as ATCC Deposit Number 75977 on December 16, 1994. The nucleotide sequence determined by sequencing the deposited KGF-2 clone, which is shown in Figure 1 [SEQ ID
NO:1 ], contains an open reading frame encoding a polypeptide of 208 amino acid residues, including an initiation codon at positions 1-3, with a predicted leader sequence of about 35 or 36 amino acid residues, and a deduced molecular weight _'j-of about 23.4 kDa. The amino acid sequence of the mature KGF-2 is shown in Figure 1, amino acid residues about 36 or 37 to 208 [SEQ ID N0:2].
The polypeptide of the present invention has been identified as a member of the FGF family, more particularly the polypeptide has been identified as KGF-2 as a result of amino acid sequence homology with other members of the FGF
family.
In accordance with one aspect of the present invention, there are provided novel mature polypeptides which are KGF-2 as well as biologically active and diagnostically or therapeutically useful fragments, analogs and derivatives thereof. The polypeptides of the present invention are of human origin.
In accordance with another aspect of the present invention, there are provided isolated nucleic acid molecules encoding human KGF-2, including mRNAs, DNAs, cDNAs, genomic DNA, as well as antisense analogs thereof, and biologically active and diagnostically or therapeutically useful fragments thereof.
In accordance with another aspect of the present invention, there is provided a process for producing such polypeptides by recombinant techniques through the use of recombinant vectors, such as cloning and expression plasmids useful as reagents in the recombinant pzoducdon of KGF-2 proteins, as well as recombinant prokaryotic and/or eukaryotic host cells comprising a human KGF-2 nucleic acid sequence.
In accordance with yet a further aspect of the present invention, there is provided therapeutic methods such as increasing platelet levels by administering KGF-2 for the purpose of alleviating thrombocytopenia. KGF-2 can also be used to increase the levels of plasma fibrinogen which may be found in conditions of abnormal hepatic synthesis (such as that associated with acute hepatitis or cirrhosis) and disseminated intravascular coagulation. KGF-2 can further be used to increase levels of serum albumin in patients with hypoalbuminemia. KGF-2 can also be used to increase the levels of serum globulin found in patients with hypoglobulinemia. KGF-2 can also be used to increase the levels of total serum protein in patients with protein loss and decreased protein synthesis.

_g_ KGF-2 can further be used to inhibit toxic effects on the prostate and bladder and to reduce the extent of ulceration caused by cystitis.
KGF-2 can also be used to stimulate the growth of sinus epithelia, nasal mucosa, lacrimal glands and salivary glands.
s Brief Description of the Figures Figures 1 A-1 C illustrate the cDNA and corresponding deduced amino acid sequence of the polypeptide of the present invention. The initial 35 or amino acid residues represent the putative leader sequence (underlined) . The standard one letter abbreviations for amino acids are used. Sequencing inaccuracies are a common problem when attempting to determine polynucleotide sequences. Sequencing was performed using a 373 Automated DNA sequencer (Applied Biosystems, Inc.). Sequencing accuracy is predicted to be greater than 97% accurate. [SEQ ID NO:1 ]
Figures 2A-2D are an illustration of a comparison of the amino acid sequence of the polypeptide of the present invention and other fibroblast growth factors. (SEQ ID NOS:13-22J
Figures 3A-3D show the full length mRNA and amino acid sequence for the KGF-2 gene. [SEQ ID NOS:23 and 24]
Figures 4A-4E show an analysis of the KGF-2 amino acid sequence.
Alpha, beta, turn and coil regions; hydrophilicity and hydrophobicity;
amphipathic regions; flexible regions; antigenic index and surface probability are shown. In the "Antigenic Index - Jameson-Wolf' graph, amino acid residues amino acid residues 41--109 in Figure 1 [SEQ ID N0:2J correspond to the shown highly antigenic regions of the KGF-2 protein. Hydrophobic regions (Hopp-Woods Plot) fall below the median line (negative values) while hydrophilic regions (Kyte-Doolittle Plot) are found above the median line (positive values, e.g. amino acid residues 41--109). The plot is over the entire 208 amino acid ORF.

Figure 5 shows the DNA sequence and the protein expressed from the pQE60-Cys37 construct [SEQ ID NOS:29 and 30]. The expressed KGF-2 protein contains the sequence from Cysteine at position 37 to Serine at position 208 with a 6X{His) tag attached to the N-terminus of the protein.
Figure 6 (A) shows the stimulation of normal primary epidermal keratinocyte proliferation by KGF-2. (B) shows the stimulation of normal primary epidermal keratinocyte proliferation by KGF-2 033. (C) shows the stimulation of normal primary epidermal keratinocyte proliferation by KGF-2 A28. Human normal primary epidermal keratinocytes were incubated with various concentrations of KGF-2, KGF-2 X33 or KGF-2 X28 for three days. For all three experiments alamarBlue was then added for 16 hr and the intensity of the red color converted from alamarBlue by the cells was measured by the difference between O.D. 570 nm and O.D. 600 nm. For each of the KGF-2 proteins a positive control with complete keratinocyte growth media (KGM), and a negative control with keratinocyte basal media (KBM) were included in the same assay plate.
Figure 7 (A) shows the stimulation of thymidine incorporation by KGF-2 and FGF7 in Baf3 cells transfected with FGFRIb and FGFR2. The effects of KGF-2 (right panel) and FGF7 (left panel) on the proliferation of Baf3 cells transfected with FGFRI iiib (open circle) or FGFR2iiib/KGFR (solid Circle were examined. Y-axis represents the amount of [3H]thymidine incorporation (cpm) into DNA of Baf3 cells. X-axis represents the final concentration of KGF-2 or FGF7 added to the tissue culture media. {B) shows the stimulation of thymidine incorporation by KGF-2A33 in Baf3 cells transfected with FGFR2iiib (C) shows the stimulation of thymidine incorporation by KGF-2 (white bar), KGF-2A33 (black bar) and KGF-228 (grey bar) in Baf3 cells transfected with FGFR2iiib.
Figure 8 shows a schematic representation of the pHE4-S expression vector {SEQ ID N0:147) and the subcloned KGF-2 cDNA coding sequence. The locations of the kanamycin resistance marker gene, the KGF-2 coding sequence, the oriC sequence, and the lacIq coding sequence are indicated.
Figure 9 shows the nucleotide sequence of the regulatory elements of the pHE promoter (SEQ ID N0:148). The two lac operator sequences, the Shine-Delgarno sequence (S/D), and the terminal HindIII and NdeI restriction sites (italicized) are indicated.
Figure 10 shows the DNA and protein sequence [SEQ ID NOS:38 and 39]
for the E.coli optimized full length KGF-2.
Figures 11 A and B show the DNA and protein sequences [SEQ ID
NOS:42, 43, 54, and 55] for the E.coli optimized mature KGF-2.
Figure 12 shows the DNA and the encoded protein sequence [SEQ ID
NOS:65 and 66] for the KGF-2 deletion construct comprising amino acids 36 to 208 of KGF-2.
Figure 13 shows the DNA and the encoded protein sequence [SEQ ID
NOS:67 and 68~ for the KGF-2 deletion construct comprising amino acids 63 to 208 of KGF-2.
Figure 14 shows the DNA and the encoded protein sequence [SEQ ID
NOS:69 and 70] for the KGF-2 deletion construct comprising amino acids 77 to 208 of KGF-2.
Figure 15 shows the DNA and the encoded protein sequence [SEQ ID
NOS:71 and 72] for the KGF-2 deletion construct comprising amino acids 93 to 208 of KGF-2.
Figure 16 shows the DNA and the encoded protein sequence [SEQ ID
NOS:73 and 74] for the KGF-2 deletion construct comprising amino acids 104 to 208 of KGF-2.
Figure 17 shows the DNA and the encoded protein sequence [SEQ ID
NOS:75 and 76] for the KGF-2 deletion construct comprising amino acids 123 to 208 of KGF-2.

Figure 18 shows the DNA and the encoded protein sequence [SEQ ID
NOS:77 and 78] for the KGF-2 deletion construct comprising amino acids 138 to 208 of KGF-2.
Figure 19 shows the DNA and the encoded protein sequence [SEQ ID
NOS:79 and 80] for the KGF-2 deletion construct comprising amino acids 36 to 153 of KGF-2.
Figure 20 shows the DNA and the encoded protein sequence [SEQ ID
NOS:81 and 82] for the KGF-2 deletion construct comprising amino acids 63 to 153 of KGF-2.
Figure 21 shows the DNA sequence for the KGF-2 Cysteine-37 to Serine mutant construct [SEQ ID N0:83].
Figure 22 shows the DNA sequence for the KGF-2 Cysteine-37/Cysteine-106 to Serine mutant construct [S,EQ iD N0:84].
Figure 23 shows the proliferation of bladder epithelium following ip or sc administration of KGF-2 .0,33.
Figure 24 shows the proliferation of prostatic epithelial cells after systemic administration of KGF-2 A33.
Figure 25 shows the effect of KGF-2 X33 on bladder wall ulceration in a cyclophosphamide-induced hemorrhagic cystitis model in the rat.
Figure 26 shows the effect of KGF-2 X33 on bladder wall thickness in a cyclophosphamide-induced cystitis rat model.
Figure 27 provides an overview of the study design to determine whether KGF-2 X33 induces proliferation of normal epithelia in rats when administered systemically using SC and IP routes.
Figure 28. Normal Sprague Dawley rats were injected daily with KGF-2 033 (5 mg/kg; HG03411-E2) or buffer and sacrificed one day after the final injection. A blinded observer counted the proliferating cells in ten randomly chosen fields per animals at a 10 x magnification. SC administration of KGF-2 X33 elicited a significant proliferation after one day which then returned to normal by 2 days. KGF-2 033 given ip stimulated proliferation from 1-3 days but only the results from days 1 and 3 were statistically significant.
Figure 29. Normal Sprague Dawley rats were injected daily with KGF-2 033 (5 mg/kg; HG03411-E2) or buffer and sacrificed one day after the final injection. A blinded observer counted the proliferating cells in ten randomly chosen fields per animal at a 10 x magnification. KGF-2 X33 given ip stimulated proliferation over the entire study period while sc administration of KGF-2 did not increase the proliferation at any time point.
Figure 30. Normal Sprague Dawley rats were injected daily with KGF-2 D33 (5 mg/kg; HG03411-E2) or buffer and sacrificed one day after the final injection. A blinded observer counted the proliferating cells in one cross-section per animal at a 10 x magnification. KGF-2 X33 given sc elicited a significant increase in proliferation after 1, 2, and 3 days of daily administration. When KGF-2 X33 was given ip, proliferation was seen after 2 and 3 days only.
Figure 31 demonstrates KGF-2 X33 induced proliferation ~n normal rat lung.
Figure 32 shows a morphometric assessment of the number of BrdU
positive cells in the parotid gland revealed dramatic proliferation over the seven day study period. Note that BrdU incorporation returned to normal after a seven day rest period. Measurements were performed in a blinded manner.
Figure 33 shows the weight of the parotid glands from animals treated daily with KGF-2 or buffer are expressed as mg per g of body weight. Only after seven daily intravenous injections of KGF-2 did the organ weight significantly increase and this normalized after a seven day rest period.
Figure 34 shows the proliferative effect of KGF-2 on the submandibular gland was apparent after one injection (day 1). It remained active over three doses but after seven daily injections was no longer inducing a significant increase in proliferation. All measurements were performed in a blinded manner.
Figure 35 shows the weight of the submandibular glands obtained from rats treated daily with KGF-2 showed a significant increase in size after two injections. This enlargement peaked after seven daily injections. After one week of recovery the weight of the submandibular gland from rats treated with KGF-2 had began to normalize but was still significant larger than tissue taken from buffer treated control rats.
Figure 36 shows the proliferation of the lacrimal gland after 1, 2 and 3 daily i.v. injections of KGF-233. Animals were used in groups of 6, and the proliferation of the tissue was assessed by a blinded observer using a computerized morphometry unit. The proliferation results are expressed as the percentage of a region of interest that has proliferating cells (% ROI). The error bars reflect the SEM. Statistical analysis was performed using StatView. A
factorial ANOVA was performed followed by a Scheffe post-hoc test and statistical significant was defined as p< 0.05.
Figure 37 shows the effectof KGF-2 intravenous injection at days 1, 2, 3 and 7 on the cornea. The results express the replicating epithelia as number of cells per mm of tissue. An increase in the number of proliferating cells following KGF-2 treatment on days 1 and 2 was observed. However, only the results from day 2 attained statistical significance.
Figure 38 shows the results of KGF-2 intravenous injection at days 1, 2, 3 and 7 an the conjunctiva. The results express the replicating epithelia as number of cells per mm of tissue. An increase in the number of proliferating cells following KGF-2 treatment on days l, 2 and 3 was observed. These results were statistically significant.
Figures 39 (A) and (B) show the results of KGF-2033 daily intravenous injections on pilocarpine-induced salivation. Figure 39(A) shows KGF-2033 treatment induces a large increase in saliva production in the treated animals.
Figure 39(B) shows that the concentration of amylase in saliva is decreased following the daily KGF-2033 injections.
Figure 40 shows the results of KGF-2 intravenous injection at days 1, 2, 3, 7 and 14 on the maxillary sinus in normal rats. There was an increase in the total number of proliferating cells following KGF-2 treatment on Days 1, 2, 3 WO 99/41282 PC1'/US99/03018 and 7. All days, except Day 14, attained statistical significance. By day 14, the number of proliferating cells returned to normal.
Figure 41 shows the results of KGF-2L~33 intravenous injection at days 1, 2, 3, 7 and 14 on the nasal septum in normal rats. There was an increase in the S total number of proliferating cells following KGF-2 treatment on Days 1, 2, and 7. All days, except Day 14, attained statistical significance. By day 14, the number of proliferating cells returned to normal.
Figure 42 shows the effect of KGF-233 on the number of goblet cells in the conjunctiva in male rats intravenously injected with either KGF-2033 or buffer at days 1, 2, 3, and 7.
Figure 43 shows the effect of intravenous KGF-2 on lung proliferation.
Male SD rats received buffer or KGF-233 i.v. (1 or 5 mg/kg) on day 1. Animals were injected with BrdU (100 mg/kg, i.p.) 2 hours prior to euthanasia. Animals were sacrificed at 6, 24, or 48 hour time points. The average number of BrdU
positive cells per field was counted in 5 random fields at 20X. Statistical significance is achieved using the unpaired t-test where p<0.05 when compared to the buffer control.
Figure 44 shows the effect of nebulized KGF-2 on proliferation in normal rat lungs. Male Lewis rats inhaled nebulized buffer or KGF-233 (56 or 12 mg/rat). Twenty four hours post-nebulization, animals received BrdU ( l OOmg/kg, i.p.) 2 hours prior to euthanasia. The average number of positively stained cells in 5 random microscopic fields (20X) per lobe was the BrdU cell count.
Figure 45 shows the of prophylactic KGF-2033 on body weights in a bleomycin-induced lung injury rat model. Male rats were given intratracheal doses of KGF-2033 (0.5, 1 and S mg/kg) of buffer. Treatments were given on day 0 and day 1. Bleomycin was given on day 3. Animals were weighed on alternate days until the 14'" day. Statistical analysis was done using an unpaired t-test. *compared with the no treatment group; **compared to the KGF-2 buffer group. Figure 46 shows the effect of prophylactic KGF-233 on fibrosis score in bleomycin-induced lung injury rat model. Male Lewis rats (n=5) were given intratracheal doses of of KGF-233 0.5 mg/ml, l mg/ml, 5 mg/ml or buffer while under anesthesia. Treatrnents were given on day 0 and day 1. Bleomycin was administered on day 3. On the final day of the experiment (day 14), animals were injected with BrdU (100 mg/kg, i.p.) 2 hours prior to euthanasia. The scoring was graded 0 = normal, 1-3 = mild, 4-b = moderate, and 7-9 = severe at 1 OX. Statistical analysis was performed with a student's paired t-test.
*compared to buffer treatment group, * * compared to no treatment group.
Figure 47 shows the effect of prophylactic KGF-2 X33 on bleomycin-induced lung injury rat model. Male Lewis rats (n=5) were given intratracheal doses of of KGF-2 0.5 mg/ml, l mg/ml, 5 mg/ml or buffer while under anesthesia.
Treatments were given on day 0 and day 1. Bleomycin was administered on day 3. On the final day of the experiment (day 14), animals were injected with BrdU
(100 mg/kg, i.p.) 2 hours prior to euthanasia. The number of BrdU positive cells per field was counted in 10 random fields at 20X. Statistical analysis was performed with a student's paired t-test. *compared to Buffer treatment gmup * * compared to no trea~nent group.
Figure 48 shows the effect of KGF-233 on bladder capacity in cyclophosphamide-induced cystitis rat model. Male SD rats (200g) received KGF-2033 in doses of 0.1 mg/kg, 0.3 mg/kg, 1.0 mg/kg or 3.0 mg/kg intravenously. On day 1, 200 mg/kg of cyclophosphamide (CP) was administered i.p. Animals were sacrificed on day 4. After urine was removed from the bladder, it was filled with formalin until it leaked out of the urethra. The volume of formalin injected into the bladders was recorded as the bladder capacity. (* compared to CP only control; j' compared to buffer control).
Figure 49 shows the effect of KGF-2033 on bladder wet weight in cyclophosphamide-induced cystitis rat model. Male SD rats (200g) received KGF-2033 in doses of 0.1 mg/kg, 0.3 mg/kg, 1.0 mg/kg or 3.0 mglkg intravenously. On day 1, 200 mg/kg of cyclophosphamide (CP) was administered i.p. Animals were sacrificed on day 4. Bladders were fixed in fonmalin, harvested, weighed and then placed in tissue cassettes for histological analysis. (* compared to CP only control; ~ competed to buffer control).
Figure 50 shows the effect of KGF-2033 and mesna on bladder ulceration of cyclophosphamide-induced cystitis. Male S.D. rats (200g) received either buffer or 5.0 mg/kg KGF-233 intravenously on day 0. On day 1, 200 mg/kg of cyclophosphamide (CP) i.p and Mesna (20 ~g/g or 40 ~g/g, i.v.). Animals were sacrificed on day 3 and bladders were fixed internally with fonmalin before tissue harvest. Saline was used as a normal contml. Statistical analysis was performed using the unpaired t-test where significance is found if p < 0.05. * Compared to CP oniy; * *compared to buffer.
Figure 51 shows the effect of KGF-2033 and mesna on percent normal urothelium of cyclophosphamide-induced cystitis. Male S.D. rats (200g) received either buffer or 5.0 mglkg KGF-2033 intravenously on day 0. On day 1, 200 mg/kg of cyclophosphamide (CP} i.p and Mesna (20 ~,g/g or 40 ~glg, i.v.).
Animals were sacrificed on day 3 and bladders were fixed internally with formalin before tissue harvest. Saline was used as a normal control.
Statistical analysis was performed using the unpaired t-test where significance is found if p <_ 0.05. *Compared to CP only; **compared to buffer.
Figure 52 shows the effect of KGF-2033 and mesna on bladder wall thickness of cyclophosphamide-induced cystitis. Male S.D. rats (200g) received either buffer or 5.0 mg/kg KGF-233 intravenously on day 0. On day I, 200 mg/kg of cyclophosphamide (CP) i.p and Mesna {20 pg/g or 40 ~g/g, i.v.).
Animals were sacrificed on day 3 and bladders were fixed internally with formalin before tissue harvest. Saline was used as a normal control.
Statistical analysis was performed using the unpaired t-test where significance is found if p <_ 0.05. *Compared to CP only; **compared to buffer.
Figure 53 shows the synergistic effect of KGF-2A33 and mesna on bladder capacity of cyclophosphamide-induced cystitis. Male SD rats (350-400 g) (n=7) received, on day 0, either buffer or 5.0 mg/kg KGF-2033 intravenously and on day 1, Mesna (40 ~g/g, i.v.) or both treatments on the respective administration days. Cyclophosphamide (300 mglkg, i.p.) was administered on day 1 to all treatment groups with the exception of the saline control. One group was added as a CP control with no treatment. On day 3, animals received BrdU
( 100mg/kg, i.p.) 2 hours prior to euthanasia. After urine was removed from the bladder, it was filled with formalin until it leaked out of the urethra. The volume of formalin injected into the bladder was recorded as the bladder capacity.
*Compared to CP only control; ~ compared to buffer control.
Figure 54 shows the synergistic effect of KGF-233 and mesna on bladder wet weight of cyclophosphamide-induced cystitis. Male SD rats (350-400 g) (n=7) received, on day 0, either buffer or 5.0 mg/kg KGF-233 intravenously and on day 1, Mesna (40 p,g/g, i.v.) or both treatments on the respective administration days. Cyclophosphamide (300 mg/kg, i.p.) was administered on day 1 to all treatment groups with the exception of the saline control. One group was added as a CP control with no treatment. On day 3, animals received BrdU (100mg/kg, i.p.) 2 hours prior to euthanasia. Bladders were fixed with 10% neutral-buffered formalin, weighed and placed in tissue cassettes for histological analysis. *Compared to CP only control; j' compared to buffer control.
Detailed Description In accordance with an aspect of the present invention, there is provided an isolated nucleic acid (polynucleotide) which encodes for the polypeptide having the deduced amino acid sequence of Figure 1 (SEQ ID N0:2) or for the polypeptide encoded by the cDNA of the clone deposited as ATCC Deposit No.
75977 on December 16, 1994 at the American Type Culture Collection, Patent Depository, 10801 University Blvd, Manassas, VA 20110-2209.

Nucleic Acid Molecules Unless otherwise indicated, all nucleotide sequences determined by sequencing a DNA molecule herein were determined using an automated DNA
sequencer (such as the Model 373 from Applied Biosystems, Inc.), and all amino S acid sequences of polypeptides encoded by DNA molecules determined herein were predicted by translation of a DNA sequence determined as above.
Therefore, as is known in the art for any DNA sequence determined by this automated approach, any nucleotide sequence determined herein may contain some errors. Nucleotide sequences determined by automation are typically at least about 90% identical, more typically at least about 95% to at least about 99.9% identical to the actual nucleotide sequence of the sequenced DNA
molecule. The actual sequence can be more precisely determined by other approaches including manual DNA sequencing methods well known in the art.
As is also known in the art, a single insertion or deletion in a determined nucleotide sequence compared to the actual sequence will cause a frame shift in translation of the nucleotide sequence such that the predicted amino acid sequence encoded by a detenmined nucleotide sequence will be completely different from the amino acid sequence actually encoded by the sequenced DNA
molecule, beginning at the point of such an insertion or deletion.
Unless otherwise indicated, each "nucleotide sequence" set forth herein is presented as a sequence of deoxyribonucleotides (abbreviated A, G , C and T).
However, by "nucleotide sequence" of a nucleic acid molecule or polynucleotide is intended, for a DNA molecule or polynucleotide, a sequence of deoxyribonucleotides, and for an RNA molecule or polynucleotide, the corresponding sequence of ribonucleotides (A, G, C and U), where each thymidine deoxyribonucleotide (T) in the specified deoxyribonucleotide sequence is replaced by the ribonucleotide uridine (U). For instance, reference to an RNA
molecule having the sequence of SEQ ID NO:1 set forth using deoxyribonucleotide abbreviations is intended to indicate an RNA molecule having a sequence in which each deoxyribonucleotide A, G or C of SEQ ID NO: l has been replaced by the corresponding ribonucleotide A, G or C, and each deoxyribonucleotide T has been replaced by a ribonucleotide U.
By "isolated" nucleic acid molecules) is intended a nucleic acid molecule, DNA or RNA, which has been removed from its native environment For example, recombinant DNA molecules contained in a vector are considered isolated for the purposes of the present invention. Further examples of isolated DNA molecules include recombinant DNA molecules maintained in heterologous host cells or purified (partially or substantially) DNA molecules in solution.
Isolated RNA molecules include in vivo or in vitro RNA transcripts of the DNA
molecules of the present invention. Isolated nucleic acid molecules according to the present invention further include such molecules produced synthetically.
Isolated nucleic acid molecules of the present invention include DNA
molecules comprising an open reading frame (ORF) with an initiation codon at positions 1-3 of the nucleotide sequence shown in Figure 1 (SEQ ID NO:1 ); DNA
molecules comprising the coding sequence for the mature KGF-2 protein shown in Figure 1 (last 172 or 173 amino acids) (SEQ ID N0:2); and DNA molecules which comprise a sequence substantially different from those described above but which, due to the degeneracy of the genetic code, still encode the KGF-2 protein.
Of course, the genetic code is well known in the art. Thus, it would be routine for one skilled in the art to generate the degenerate variants described above.
A polynucleotide encoding a polypeptide of the present invention may be obtained from a human prostate and fetal lung. A fragment of the cDNA
encoding the polypeptide was initially isolated from a library derived from a human normal prostate. The open reading frame encoding the full length protein was subsequently isolated from a randomly primed human fetal lung cDNA
library. It is structurally related to the FGF family. It contains an open reading frame encoding a protein of 208 amino acid residues of which approximately the first 35 or 36 amino acid residues are the putative leader sequence such that the mature protein comprises 173 or 172 amino acids. The protein exhibits the highest degree of homology to human keratinocyte growth factor with 45%
identity and 82% similarity over a 206 amino acid stretch. It is also important that sequences that are conserved through the FGF family are found to be conserved in the protein of the present invention.
In addition, results from nested PCR of KGF-2 cDNA from libraries showed that there were potential alternative spliced forms of KGF-2.
Specifically, using primers flanking the N-terminus of the open reading frame of KGF-2, PCR products of 0.2 kb and 0.4 kb were obtained from various cDNA
libraries. A 0.2 kb size was the expected product for KGF-2 while the 0.4 kb size may result from an alternatively spliced form of KGF-2. The 0.4 kb product was observed in libraries from stomach cancer, adult testis, duodenum and pancreas.
The polynucleotide of the present invention may be in the form of RNA
or in the form of DNA, which DNA includes cDNA, genomic DNA, and synthetic DNA. The DNA may be doublestranded or single-stranded, and if single stranded may be the coding strand or non-coding (anti-sense) strand.
The coding sequence which encodes the mature polypeptide may be identical to the coding sequence shown in Figure 1 (SEQ ID NO: I ) or that of the deposited clone or may be a different coding sequence which coding sequence, as a result of the redundancy or degeneracy of the genetic code, encodes the same mature polypeptide as the DNA of Figure 1 (SEQ ID NO:1) or the deposited cDNA.
The polynucleotide which encodes for the predicted mature polypeptide of Figure 1 (SEQ ID N0:2) or for the predicted mature polypeptide encoded by the deposited cDNA may include: only the coding sequence for the mature polypeptide; the coding sequence for the mature polypeptide and additional coding sequence such as a leader or secretary sequence or a proprotein sequence;
the coding sequence for the mature polypeptide (and optionally additional coding sequence) and non-coding sequence, such as intron or non-coding sequence 5' and/or 3' of the coding sequence for the predicted mature polypeptide. In WO 99141282 PC"T/US99/03018 addition, a full length mRNA has been obtained which contains 5' and 3' untranslated regions of the gene (Figure 3 (SEQ ID NO:23)).
As one of ordinary skill would appreciate, due to the possibilities of sequencing errors discussed above, as well as the variability of cleavage sites for leaders in different known proteins, the actual KGF-2 polypeptide encoded by the deposited cDNA comprises about 208 amino acids, but may be anywhere in the range of 200-220 amino acids; and the actual leader sequence of this protein is about 35 or 36 amino acids, but may be anywhere in the range of about 30 to about 40 amino acids.
Thus, the term "polynucleotide encoding a polypeptide" encompasses a polynucleotide which includes only coding sequence for the polypeptide as well as a polynucleotide which includes additional coding and/or non-coding sequence.
The present invention further relates to variants of the hereinabove i 5 described polynucleoddes which encode for fragments, analogs and derivatives of the polypeptide having the deduced amino acid sequence of Figure 1 (SEQ ID
N0:2) or the polypeptide encoded by the cDNA of the deposited clone. The variant of the polynucleotide may be a naturally occurring allelic variant of the polynucleotide or a nonnaturally occurring variant of the polynucleotide.
Thus, the present invention includes polynucleotides encoding the same predicted mature polypeptide as shown in Figure 1 (SEQ ID N0:2) or the same predicted mature polypeptide encoded by the cDNA of the deposited clone as well as variants of such polynucleotides which variants encode for a fragment, derivative or analog of the polypeptide of Figure 1 (SEQ ID N0:2) or the polypeptide encoded by the cDNA of the deposited clone. Such nucleotide variants include deletion variants, substitution variants and addition or insertion variants.
The present invention includes polynucleotides encoding mimetic peptides of KGF-2 which can be used as therapeutic peptides. Mimetic KGF-2 peptides are short peptides which mimic the biological activity of the KGF-2 protein by WO 99!41282 PCTNS99/03018 binding to and activating the cognate receptors of KGF-2. Mimetic KGF-2 peptides can also bind to and inhibit the cognate receptors of KGF-2. KGF-2 receptors include, but are not limited to, FGFR2iiib and FGFRliiib. Such mimetic peptides are obtained from methods such as, but not limited to, phage display or combinatorial chemistry. For example the method disclosed by Wrighton et al. Science 273:458-463 { 1996) to generate mimetic KGF-2 peptides.
As hereinabove indicated, the polynucleotide may have a coding sequence which is a naturally occurring allelic variant of the coding sequence shown in Figure 1 (SEQ ID NO:1) or of the coding sequence of the deposited clone. As known in the art, an allelic variant is an alternate form of a polynucleotide sequence which may have a substitution, deletion or addition of one or more nucleotides, which does not substantially alter the function of the encode polypeptide.
The present invention also includes polynucleotides, wherein the coding sequence for the mature polypeptide may be fused in the same reading frame to a polynucleotide sequence which aids in expression and secretion of a polypeptide from a host cell, for example, a leader sequence which functions as a secretory sequence for controlling transport of a polypeptide from the cell. The polypeptide having a leader sequence is a preprotein and may have the leader sequence cleaved by the host cell to form the mature form of the polypeptide. The polynucleotides may also encode for proprotein which is the mature protein plus additional 5' amino acid residues. A mature protein having a prosequence is a proprotein and is an inactive form of the protein. Once the prosequence is cleaved an active mature protein remains.
Thus, for example, the polynucleotide of the present invention may encode for a matiu~e protein, or for a pmtein having a prosequence or for a protein having both prosequence and a presequence (leader sequence).
The polynucleotides of the present invention may also have the coding sequence fiised in frame to a marker sequence which allows for purification of the polypeptide of the present invention. The marker sequence may be a hexahistidine tag supplied by a pQE-9 vector to provide for purification of the mature polypeptide fused to the marker in the case of a bacterial host, or, for example, the marker sequence may be a hemagglutinin (HA) tag when a mammalian host, e.g. COS-7 cells, is used. The HA tag corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson, I. et al. Cell 37:767 ( 1984)).
The term "gene" means the segment of DNA involved in producing a polypeptide chain; it includes regions preceding and following the coding region (leader and trailer) as well as intervening sequences (introns) between individual coding segments (exons).
Fragments of the full length gene of the present invention may be used as a hybridization probe for a cDNA library to isolate the full length cDNA and to isolate other cDNAS which have a high sequence similarity to the gene or similar biological activity. Probes of this type preferably have at least 30 bases and may contain, for example, 50 or more bases. The probe may also be used to identify a cDNA clone corresponding to a full length transcript and a genomic clone or clones that contain the complete gene including regulatory and promotor regions, exons, and introns. An example of a screen comprises isolating the coding region of the gene by using the known DNA sequence to synthesize an oligonucleodde probe. Labeled oligonucleotides having a sequence complementary to that of the gene of the present invention are used to screen a library of human cDNA, genomic DNA or cDNA to determine which members of the library the probe hybridizes to.
Further embodiments of the invention include isolated nucleic acid molecules comprising a polynucleotide having a nucleotide sequence at least 90%
identical, and more preferably at least 95%, 96%, 97%, 98% or 99% identical to (a) a nucleotide sequence encoding the full-length KGF-2 polypeptide having the complete amino acid sequence in Figure 1 (SEQ ID N0:2), including the predicted leader sequence; (b) a nucleotide sequence encoding the mature KGF-2 polypeptide (full-length polypeptide with the leader removed) having the amino acid sequence at positions about 36 or 37 to 208 in Figure 1 (SEQ ID N0:2);
(c) a nucleotide sequence encoding the full-length KGF-2 polypeptide having the complete amino acid sequence including the leader encoded by the cDNA clone contained in ATCC Deposit No. 75977; {d) a nucleotide sequence encoding the mature KGF-2 polypeptide having the amino acid sequence encoded by the cDNA clone contained in ATCC Deposit No. 75977; (e) a nucleotide sequence encoding any of the KGF-2 analogs or deletion mutants described below; or (fj a nucleotide sequence complementary to any of the nucleotide sequences in (a), (b), (c),(d), or (e).
By a polynucleotide having a nucleotide sequence at least, for example, 95% "identical" to a reference nucleotide sequence of the present invention, it is intended that the nucleotide sequence of the polynucleotide is identical to the reference sequence except that the polynucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequence encoding the KGF-2 polypeptide. In other words, to obtain a polynucleotide having a nucleotide sequence at least 95% identical to a reference nucleotide sequence, up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence. The query sequence may be an entire sequence shown of SEQ ID NO:1, the ORF (open reading frame), or any fragment specified as described herein.
As a practical matter, whether any particular nucleic acid molecule or polypeptide is at least 90%, 95%, 96%, 97%, 98% or 99% identical to a nucleotide sequence of the presence invention can be determined conventionally using known computer programs. A preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence, also referred to as a global sequence alignment, can be determined using the FASTDB computer program based on the algorithm of Brutlag et al. (Comp. App. Biosci. (1990) 6:237-245.) In a sequence alignment the query and subject sequences are both DNA sequences. An RNA sequence can be compared by converting U's to T's. The result of said global sequence alignment is in percent identity. Preferred parameters used in a FASTDB
alignment of DNA sequences to calculate percent identify are: Matrix=Unitary, k-tuple=4, Mismatch Penalty=l, Joining Penalty=30, Randomization Group Length=0, Cutoff Score=1, Gap Penalty=5, Gap Size Penalty 0.05, Window Size=500 or the length of the subject nucleotide sequence, whichever is shorter.
If the subject sequence is shorter than the query sequence because of 5' or 3' deletions, not because of internal deletions, a manual correction must be made to the results. This is because the FASTDB program does not account for 5' and 3' truncations ofthe subject sequence when calculating percent identity. For subject sequences truncated at the S' or 3' ends, relative to the query sequence, the percent identity is corrected by calculating the number of bases of the query sequence that are S' and 3' of the subject sequence, which are not I S matchedlaligned, as a percent of the total bases of the query sequence.
Whether a nucleotide is matched/aligned is determined by results of the FASTDB
sequence alignment. This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score. This corrected score is what is used for the purposes of the present invention. Only bases outside the 5' and 3' bases of the subject sequence, as displayed by the FASTDB alignment, which are not matched/aligned with the query sequence, are calculated for the purposes of manually adjusting the percent identity score.
For example, a 90 base subject sequence is aligned to a 100 base query sequence to determine percent identity. The deletions occur at the 5' end of the subject sequence and therefore, the FASTDB alignment does not show a matched/alignment of the first 10 bases at 5' end. The 10 unpaired bases represent 10% of the sequence (number of bases at the 5' and 3' ends not matched/total number of bases in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 bases were perfectly matched the final percent identity would be 90%. In another example, a 90 base subject sequence is compared with a 100 base query sequence. This time the deletions are internal deletions so that there are no bases on the 5' or 3' of the subject sequence which are not matched/aligned with the query. In this case the percent identity calculated by FASTDB is not manually corrected. Once again, only bases 5' and 3' of the subject sequence which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention.
The KGF-2 variants may contain alterations in the coding regions, non-coding regions, or both. Especially preferred are polynucleotide variants containing alterations which produce silent substitutions, additions, or deletions, but do not alter the properties or activities of the encoded polypeptide.
Nucleotide variants produced by silent substitutions due to the degeneracy of the genetic code are preferred. Moreover, variants in which 5-10,1-5, or 1-2 amino acids are substituted, deleted, or added in any combination are also preferred.
KGF-2 polynucleotide variants can be produced for a variety of reasons, e.g., to optimize colon expression for a particular host (change colons in the human mRNA to those preferred by a bacterial host such as E. coli).
Alternatively, whether any particular nucleic acid molecule is at least 90%, 95%, 9b%, 97%, 98% or 99% identical to, for instance, the nucleotide sequence shown in Figure 1 (SEQ ID NO:1 ) or to the nucleotides sequence of the deposited cDNA clone may also be determined conventionally using known computer programs such as the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, WI 53711. Bestfit uses the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2: 482-489 ( 1981 ), to find the best segment of homology between two sequences. When using Bestfit or any other sequence alignment program to determine whether a particular sequence is, for instance, 95% identical to a reference sequence according to the present invention, the parameters are set, of course, such that the WO 99/41282 PG"TIUS99103018 percentage of identity is calculated over the full length of the reference nucleotide sequence and that gaps in homology of up to 5% of the total number of nucleotides in the reference sequence are allowed.
The present application is directed to nucleic acid molecules at least 90%, S 95%, 96%, 97%, 98% or 99% identical to the nucleic acid sequence shown in Figure 1 [SEQ ID NO:1 ] or to the nucleic acid sequence of the deposited cDNA, irrespective of whether they encode a polypeptide having KGF-2 activity. This is because even where a particular nucleic acid molecule does not encode a polypeptide having KGF-2 activity, one of skill in the art would still know how to use the nucleic acid molecule, for instance, as a hybridization probe or a polymerase chain reaction (PCR) primer. Uses of the nucleic acid molecules of the present invention that do not encode a polypeptide having KGF-2 activity include, inter alia, (1) isolating the KGF-2 gene or allelic variants thereof in a cDNA library; (2) in situ hybridization (e.g., "FISH") to metaphase chromosomal spreads to provide precise chromosoltlal location of the KGF-2 gene, as described in Verma et al., Human Chromosomes: A Manual ofBasic Technigues, Pergamon Press, New York (1988); and Northern Blot analysis for detecting KGF-2 mRNA
expression in specific tissues.
Preferred, however, are nucleic acid molecules having sequences at least 90%, 95%, 96%, 97%, 98% or 99% identical to the nucleic acid sequence shown in Figure 1 [SEQ ID NO: I ] or to the nucleic acid sequence of the deposited cDNA which do, in fact, encode a polypeptide having KGF-2 protein activity. By "a polypeptide having KGF-2 activity" is intended polypeptides exhibiting activity similar, but not necessarily identical, to an activity of the wild-type KGF-2 protein of the invention or an activity that is enhanced over that of the wild-type KGF-2 protein (either the full-length protein or, preferably, the mature protein), as measured in a particular biological assay.
Assays of KGF-2 activity are disclosed, for example, in the Examples below. These assays can be used to measure KGF-2 activity of partially purified or purified native or recombinant protein.

KGF-2 stimulates the proliferation of epidermal keratinocytes but not mesenchymal cells such as fibroblasts. Thus, "a polypeptide having KGF-2 protein activity" includes polypeptides that exhibit the KGF-2 activity in the keratinocyte proliferation assay set forth in the Examples below and will bind to the FGF receptor isoforms 1-iiib and 2-iiib. Although the degree of activity need not be identical to that of the KGF-2 protein, preferably, "a polypeptide having KGF-2 protein activity" will exhibit substantially similar activity as compared to the KGF-2 protein (i.e., the candidate polypeptide will exhibit greater activity or not more than about tenfold less and, preferably, not more than about twofold less activity relative to the reference KGF-2 protein).
Of course, due to the degeneracy of the genetic code, one of ordinary skill in the art will immediately recognize that a large number of the nucleic acid molecules having a sequence at least 90%, 95%, 96%, 97%, 98%, or 99%
identical to the nucleic acid sequence of the deposited cDNA or the nucleic acid sequence shown in Figure 1 [SEQ ID NO:1 J will encode a polypeptide "having KGF-2 protein activity" In fact, since degenerate variants of these nucleotide sequences all encode the same polypeptide, this will be clear to the skilled artisan even without performing the above described comparison assay. It will be further recognized in the art that, for such nucleic acid molecules that are not degenerate variants, a reasonable number will also encode a polypeptide having KGF-2 protein activity. This is because the skilled artisan is fully aware of amino acid substitutions that are either less likely or not likely to significantly effect protein function (e.g., replacing one aliphatic amino acid with a second aliphatic amino acid).
For example, guidance concerning how to make phenotypically silent amino acid substitutions is provided in Bowie, J. U. et al., "Deciphering the Message in Protein Sequences: Tolerance to Amino Acid Substitutions," Science 247:1306-1310 (1990), wherein the authors indicate that there are two main approaches far studying the tolerance of an amino acid sequence to change. The first method relies on the process of evolution, in which mutations are either accepted or rejected by natural selection. The second approach uses genetic engineering to introduce amino acid changes at specific positions of a cloned gene and selections or screens to identify sequences that maintain functionality.
As the authors state, these studies have revealed that proteins are surprisingly tolerant of amino acid substitutions. The authors further indicate which amino acid changes are likely to be permissive at a certain position of the protein. For example, most buried amino acid residues require nonpolar side chains, whereas few features of surface side chains are generally conserved. Other such phenotypically silent substitutions are described in Bowie, J.U. et al., supra, and the references cited therein.
The present invention further relates to polynucleotides which hybridize to the hereinabove-described sequences if there is at least 70%, preferably at least 90%, and more preferably at least 95% and still more preferably 96%, 97%, 9$%, 99% identity between the sequences. The present invention particularly relates to polynucleotides which hybridize under stringent conditions to the hereinabove-described polynucleotides. As herein used, the term "stringent conditions"
means hybridization will occur only if there is at least 95% and preferably at least 97%
identity between the sequences. The polynucleotides which hybridize to the hereinabove described polynucleotides in a preferred embodiment encode polypeptides which either retain substantially the same biological function or activity as the mature polypeptide encoded by the cDNAs of Figure 1 (SEQ ID
NO:1) or the deposited cDNA(s).
An example of "stringent hybridization conditions" includes overnight incubation at 42°C in a solution comprising: 50% formamide, 5x SSC (750 mM
NaCI, 75mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), Sx Denhardt's solution, 10% dextran sulfate, and 20 ~tg/ml denatured, sheared salmon sperm DNA, followed by washing the filters in 0. lx SSC at about 65 °C.
Alternatively, the polynucleotide may have at least 20 bases, preferably bases, and more preferably at least 50 bases which hybridize to a 30 polynucleotide of the present invention and which has an identity thereto, as hereinabove described, and which may or may not retain activity. For example, such polynucleotides may be employed as probes for the polynucleotide of SEQ
ID NO:1, for example, for recovery of the polynucleotide or as a diagnostic probe or as a PCR primer.
Of course, polynucleotides hybridizing to a larger portion of the reference polynucleotide (e.g., the deposited cDNA clone), for instance, a portion 50, 75, I00, 125, 150, 175, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, 600, 625, 650, 675, 700, 725, or 750 nt in length, or even to the entire length of the reference polynucleotide, are also useful as probes according to the present invention, as are polynucleotides corresponding to most, if not all, of the nucleotide sequence of the deposited cDNA or the nucleotide sequence as shown in Figure 1 [SEQ ID NO:1 ]. By a portion of a polynucleotide of "at least 20 nt in length," for example, is intended 20 or more contiguous nucleotides from the nucleotide sequence of the reference polynucleotide (e.g., the deposited cDNA or the nucleotide sequence as shown in Figure 1 [SEQ ID
NO:1]}. As indicated, such portions are useful diagnostically either as a probe according to conventional DNA hybridization techniques or as primers for amplification of a target sequence by the polymerase chain reaction (PCR), as described, for instance, in Molecular Cloning, A Laboratory Manual, 2nd.
edition, edited by Sambrook, J., Fritsch, E. F. and Maniatis, T., (1989), Cold Spring Harbor Laboratory Press, the entire disclosure of which is hereby incorporated herein by reference.
Since a KGF-2 cDNA clone has been deposited and its determined nucleotide sequence is provided in Figure 1 [SEQ ID NO:1], generating polynucleotides which hybridize to a portion of the KGF-2 cDNA molecule would be routine to the skilled artisan. For example, restriction endonuclease cleavage or shearing by sonication of the KGF-2 cDNA clone could easily be used to generate DNA portions of various sizes which are polynucleotides that hybridize to a portion of the KGF-2 cDNA molecule. Alternatively, the hybridizing polynucleotides of the present invention could be generated synthetically according to known techniques. Of course, a polynucleotide which hybridizes only to a poly A sequence (such as the 3' terminal poly(A) tract of the KGF-2 cDNA shown in Figure 1 [SEQ ID NO:1 ]), or to a complementary stretch of T (or U) resides, would not be included in a polynucleotide of the invention used to hybridize to a portion of a nucleic acid of the invention, since such a polynucleotide would hybridize to any nucleic acid molecule containing a poly (A) stretch or the complement thereof (e.g., practically any double-stranded cDNA clone).
The invention further provides isolated nucleic acid molecules comprising a polynucleotide encoding an epitope-bearing portion of the KGF-2 protein. In particular, isolated nucleic acid molecules are provided encoding polypeptides comprising the following amino acid residues in Figure 1 (SEQ ID N0:2), which the present inventors have determined are antigenic regions of the KGF-2 protein:
1. G1y41-Asn7l: GQDMVSPEATNSSSSSFSSPSSAGRHVRSYN [SEQ ID
N0:25];
2. Lys91-Ser109: KIEKNGKVSGTKKENCPYS [SEQ ID N0:26];
3. Asn135-Tyr164: NKKGKLYGSKEFNNDCKLKERIEENGYNTY [SEQ ID
NO 27]; and 4. Asn181-A1a199: NGKGAPRRGQKTRRKNTSA [SEQ ID N0:28].
Also, there are two additonal shorter predicted antigenic areas, G1n74-Arg78 of Figure 1 (SEQ ID N0:2) and G1n170-G1n175 of Figure 1 (SEQ ID N0:2).
Methods for generating such epitope-bearing portions of KGF-2 are described in detail below.
The deposits) referred to herein will be maintained under the terms of the Budapest Treaty on the International Recognition of the Deposit of Micro-organisms for purposes of Patent Procedure. These deposits are provided merely as convenience to those of skill in the art and are not an admission that a deposit is required under 35 U.S.C. ~ 1I2. The sequence of the polynucleotides contained in the deposited materials, as well as the amino acid sequence of the polypeptides encoded thereby, are incorporated herein by reference and are controlling in the event of any conflict with any description of sequences herein.
A license may be required to make, use or sell the deposited materials, and no such license is hereby granted.
KGF 2 Polypeptides and Fragments The present invention further relates to a polypeptide which has the deduced amino acid sequence of Figure 1 (SEQ ID N0:2) or which has the amino acid sequence encoded by the deposited cDNA, as well as fragments, analogs and derivatives of such polypeptide.
As one of ordinary skill would appreciate, due to the possibilities of sequencing errors discussed above, as well as the variability of cleavage sites for leaders in different known proteins, the actual KGF-2 polypeptide encoded by the deposited cDNA comprises about 208 amino acids, but may be anywhere in the range of 200-220 amino acids; and the actual leader sequence of this protein is about 35 or 36 amino acids, but may be anywhere in the range of about 30 to about 40 amino acids.
The terms "fragment," "derivative" and "analog" when referring to the polypeptide, of Figure 1 (SEQ ID N0:2) or that encoded by the deposited cDNA, means a polypeptide which retains essentially the same biological function or activity as such polypeptide. Thus, an analog includes a protein which can be activated by cleavage of the protein portion to produce an active mature polypeptide.
The polypeptide of the present invention may be a recombinant polypeptide, a natural polypeptide or a synthetic polypeptide, preferably a recombinant polypeptide.
2S The fragment, derivative or analog of the poiypeptide of Figure 1 (SEQ
ID N0:2) or that encoded by the deposited cDNA may be (i) one in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which the mature polypeptide is fused with another compound, such as a compound to increase the half life of the polypeptide (for example, polyethylene glycol), or (iv) one in which the additional amino acids are fused to the mature polypeptide, such as a leader or secretary sequence or a sequence which is employed for purification of the mature polypeptide or a protein sequence. Such fragments, derivatives and analogs are deemed to be within the scope of those skilled in the art from the teachings herein.
The terms "peptide" and "oligopeptide" are considered synonymous (as is commonly recognized) and each term can be used interchangeably as the context requires to indicate a chain of at least to amino acids coupled by peptidyl linkages. The word "polypeptide" is used herein for chains containing more than ten amino acid residues. All oligopeptide and polypeptide formulas or sequences herein are written from left to right and in the direction from amino terlninus to carboxy terminus.
It will be recognized in the art that some amino acid sequences of the KGF-2 polypeptide can be varied without significant effect of the structure or function of the protein. If such differences in sequence are contemplated, it should be remembered that there will be critical areas on the protein which determine activity. In general, it is possible to replace residues which form the tertiary structure, provided that residues performing a similar function are used.
In other instances, the type of residue may be completely unimportant if the alteration occurs at a non-critical region of the protein.
Thus, the invention further includes variations of the KGF-2 polypeptide which show substantial KGF-2 polypeptide activity or which include regions of KGF-2 protein such as the protein portions discussed below. Such mutants include deletions, insertions, inversions, repeats, and type substitutions (for example, substituting one hydrophilic residue for another, but not strongly hydrophilic for strongly hydrophobic as a rule). Small changes or such "neutral"
amino acid substitutions will generally have little effect on activity.
Typically seen as conservative substitutions are the replacements, one for another, among the aliphatic amino acids Ala, Val, Leu and Ile; interchange ofthe hydroxyl residues Ser and Thr, exchange of the acidic residues Asp and Glu, substitution between the amide residues Asn and Gln, exchange of the basic residues Lys and Arg and replacements among the aromatic residues Phe, Tyr.
As indicated in detail above, further guidance concerning which amino acid changes are likely to be phenotypically silent (i.e., are not likely to have a significant deleterious effect on a function) can be found in Bowie, J.U., et al., "Deciphering the Message in Protein Sequences: Tolerance to Amino Acid Substitutions," Science 247:1306-1310 (1990).
The present invention includes mimetic peptides of KGF-2 which can be used as therapeutic peptides. Mimetic KGF-2 peptides are short peptides which mimic the biological activity of the KGF-2 protein by binding to and activating the cognate receptors of KGF-2. Mimetic KGF-2 peptides can also bind to and inhibit the cognate receptors of KGF-2. KGF-2 receptors include, but are not limited to, FGFR2iiib and FGFRI iiib. Such mimetic peptides are obtained from methods such as, but not limited to, phage display or combinatorial chemistry.
For example, the method disclosed by Wrighton et al. Science 273:458-463 (1996) can be used to generate mimetic KGF-2 peptides.
The polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity.
The polypeptides of the present invention are preferably in an isolated form. By "isolated polypeptide" is intended a polypeptide removed from its native environment. Thus, a polypeptide produced and/or contained within a recombinant host cell is considered isolated for purposes of the present invention.
Also intended are polypeptides that have been purified, partially or substantially, from a recombinant host cell or a native source.

The polypeptides of the present invention include the polypeptide of SEQ
ID N0:2 (in particular the mature polypeptide) as well as polypeptides which have at least 90%, 95%, 96%, 97%, 98%, 99% similarity (more preferably at least 90%, 95%, 96%, 97%, 98%, 99% identity) to the polypeptide of SEQ ID N0:2 and also include portions of such polypeptides with such portion of the polypeptide (such as the deletion mutants described below) generally containing at least 30 amino acids and more preferably at least 50 amino acids.
As known in the art "similarity" between two polypeptides is determined by comparing the amino acid sequence and its conserved amino acid substitutes of one polypeptide to the sequence of a second polypeptide.
By "% similarity" for two polypeptides is intended a similarity score produced by comparing the amino acid sequences of the two polypeptides using the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, WI 53711) and the default settings for determining similarity.
Bestfit uses the local homology algorithm of Smith and Waterrnan (Advances in Applied Mathematics 2: 482-489,1981 ) to find the best segment of similarity between two sequences.
By a polypeptide having an amino acid sequence at least, for example, 95% "identical" to a reference amino acid sequence of a KGF-2 polypeptide is intended that the amino acid sequence of the polypeptide is identical to the reference sequence except that the polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the reference amino acid of the KGF-2 polypeptide. In other words, to obtain a polypeptide having an amino acid sequence at least 95% identical to a reference amino acid sequence, up to 5%
of the amino acid residues in the reference sequence may be deleted or substituted with another amino acid, or a number of amino acids up to 5% of the total amino acid residues in the reference sequence may be inserted into the reference sequence. These alterations of the reference sequence may occur at the amino or carboxy terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence.
By a polypeptide having an amino acid sequence at least, for example, 95% "identical" to a query amino acid sequence of the present invention, it is intended that the amino acid sequence of the subject polypeptide is identical to the query sequence except that the subject polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the query amino acid sequence. In other words, to obtain a polypeptide having an amino acid sequence at least 95% identical to a query amino acid sequence, up to 5% of the amino acid residues in the subject sequence may be inserted, deleted, (indels) or substituted with another amino acid. These alterations of the reference sequence may occur at the amino or carboxy terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence. .
As a practical matter, whether any particular polypeptide is at least 90%, 95%, 96%, 97%, 98% or 99% identical to, for instance, the amino acid sequences shown in SEQ ID N0:2 or to the amino acid sequence encoded by deposited DNA clone can be determined conventionally using known computer programs.
A preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence, also referred to as a global sequence alignment, can be determined using the FASTDB
computer program based on the algorithm of Brutlag et al. (Comp. App. Biosci.
(1990) 6:237-245). In a sequence alignmentthe query and subject sequences are either both nucleotide sequences or both amino acid sequences. The result of said global sequence alignment is in percent identity. Preferred parameters used in a FASTDB amino acid alignment are: Matrix=PAM 0, k-tuple=2, Mismatch Penalty=1, Joining Penalty=20, Randomization Group Length; Cutoff Score=1, Window Size=sequence length, Gap Penalty=5, Gap Size Penalty=0.05, Window Size=500 or the length of the subject amino acid sequence, whichever is shorter.
Ifthe subject sequence is shorter than the query sequence due to N- or C-terminal deletions, not because of internal deletions, a manual correction must be made to the results. This is because the FASTDB program does not account for N- and C-terminal truncations of the subject sequence when calculating global percent identity. For subject sequences truncated at the N- and C-termini, relative to the query sequence, the percent identity is corrected by calculating the number of residues of the query sequence that are N- and C-terminal of the subject sequence, which are not matched/aligned with a corresponding subject residue, as a percent of the total bases of the query sequence. Whether a residue is matched/aligned is determined by results of the FASTDB sequence alignment. This percentage is then subtracted from the percent identity, calculated by the above FASTDB
program using the specified parameters, to arrive at a final percent identity score.
This final percent identity score is what is used for the purposes of the present invention. Only residues to the N- and C-termini of the subject sequence, whip are not matched/aligned with the query sequence, are considered for the purposes of manually adjusting the percent identity score. That is, only query residue positions outside the farthest N- and C-terminal residues of the subject sequence.
For example, a 90 amino acid residue subject sequence is aligned with a 100 residue query sequence to determine percent identity. The deletion occurs at the N-terminus of the subject sequence and therefore, the FASTDB alignment does not show a matching/alignment of the first 10 residues at the N-terminus.
The 10 unpaired residues represent 10% of the sequence (number of residues at the N- and C- termini not matched/total number of residues in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 residues were perfectly matched the final percent identity would be 90%. In another example, a 90 residue subject sequence is compared with a 100 residue query sequence. This time the deletions are internal deletions so there are no residues at the N- or C-termini of the subject sequence which are not matched/aligned with the query. In this case the percent identity calculated by FASTDB is not manually corrected. Once again, only residue positions outside the N- and C-terminal ends of the subject sequence, as displayed in the FASTDB alignment, which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention.
Alternatively, whether any particular polypeptide is at least 90%, 95%, 96%, 97%, 98% or 99% identical to, for instance, the amino acid sequence shown in Figure 1 [SEQ ID N0:2] or to the amino acid sequence encoded by deposited cDNA clone may also be determined conventionally using known computer programs such the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, WI 53711. When using Bestfit or any other sequence alignment program to determine whether a particular sequence is, for instance, 95% identical to a reference sequence according to the present invention, the parameters are set, of course, such that the percentage of identity is calculated over the full length of the reference amino acid sequence and that gaps in homology of up to 5% of the total number of amino acid residues in the reference sequence are allowed.
As described in detail below, the polypeptides of the present invention can be used to raise polyclonal and monoclonal antibodies, which are useful in diagnostic assays for detecting KGF-2 protein expression as described below or as agonists and antagonists capable of enhancing or inhibiting KGF-2 protein function. Further, such polypeptides can be used in the yeast two-hybrid system to "capture" KGF-2 protein binding proteins which are also candidate agonist and antagonist according to the present invention. The yeast two hybrid system is described in Fields and Song, Nature 340: 245-246 ( 1989).
In another aspect, the invention provides a peptide or polypeptide comprising an epitope-bearing portion of a polypeptide of the invention. The epitope of this polypeptide portion is an immunogenic or antigenic epitope of a polypeptide of the invention. An "immunogenic epitope" is defined as a part of a protein that elicits an antibody response when the whole protein is the immunogen. These immunogenic epitopes are believed to be confined to a few loci on the molecule. On the other hand, a region of a protein molecule to which an antibody can bind is defined as an "antigenic epitope." The number of immunogenic epitopes of a protein generally is less than the number of antigenic epitopes. See, for instance, Geysen et al., Proc. Natl. Acad. Sci. USA 81:3998-4002 (1983).
As to the selection of peptides or polypeptides bearing an antigenic epitope (i.e., that contain a region of a protein molecule to which an antibody can bind), it is well known in that art that relatively short synthetic peptides that mimic part of a protein sequence are routinely capable of eliciting an antiserum that reacts with the partially mimicked protein. See, for instance, Sutcliffe, J. G., Shinnick, T. M., Green, N. and Learner, R. A. (1983) Antibodies that react with predetermined sites on proteins. Science 219:660-666. Peptides capable of eliciting protein-reactive sera are frequently represented in the primary sequence of a protein, can be characterized by a set of simple chemical rules, and are confined neither to immunodominant regions of intact proteins (i.e., immunogenic epitopes) nor to the amino or carboxyl terminals. Peptides that are extremely hydrophobic and those of six or fewer residues generally are ineffective at inducing antibodies that bind to the mimicked protein; longer, soluble peptides, especially those containing proline residues, usually are effective. Sutcliffe et al., supra, at 661. For instance, 18 of 20 peptides designed according to these guidelines, containing 8-39 residues covering 75% of the sequence of the influenza virus hemagglutinin HAl polypeptide chain, induced antibodies that reacted with the HAl protein or intact virus; and 12/12 peptides from the MuLV
polymerase and 18/18 from the rabies glycoprotein induced antibodies that precipitated the respective proteins.
Antigenic epitope-bearing peptides and polypeptides of the invention are therefore useful to raise antibodies, including monoclonal antibodies, that bind specifically to a polypeptide of the invention. Thus, a high proportion of hybridomas obtained by fusion of spleen cells from donors immunized with an antigen epitope-bearing peptide generally secrete antibody reactive with the native protein. Sutcliffe et al., supra, at 663. The antibodies raised by antigenic epitope-bearing peptides or polypeptides are useful to detect the mimicked protein, and antibodies to different peptides may be used for tracking the fate of various regions of a protein precursor which undergoes post-translational processing. The peptides and anti-peptide antibodies may be used in a variety of qualitative or quantitative assays for the mimicked protein, for instance in competition assays since it has been shown that even short peptides (e.g., about 9 amino acids) can bind and displace the larger peptides in inununoprecipitation assays. See, for instance, Wilson et al., Cell 37:767-778 (1984) at 777. The anti-peptide antibodies of the invention also are useful for purification of the mimicked protein, for instance, by adsorption chromatography using methods well known in the art.
Antigenic epitope-bearing peptides and polypeptides of the invention designed according to the above guidelines preferably contain a sequence of at least seven, more preferably at least nine and most preferably between about I

to about 30 amino acids contained within the amino acid sequence of a polypeptide of the invention. However, peptides or polypeptides comprising a larger portion of an anuno acid sequence of a polypeptide of the invention, containing about 30, 40, 50, 60, 70, 80, 90,100, or 150 amino acids, or any length up to and including the entire amino acid sequence of a polypeptide of the invention, also are considered epitope-bearing peptides or polypeptides of the invention and also are useful for inducing antibodies that react with the mimicked protein. Preferably, the amino acid sequence of the epitope-bearing peptide is selected to provide substantial solubility in aqueous solvents (i.e., the sequence includes relatively hydrophilic residues and highly hydrophobic sequences are preferably avoided); and sequences containing proline residues are particularly preferred.
Non-limiting examples of antigenic polypeptides or peptides that can be used to generate KGF-2-specific antibodies include the following:
1. G1y41-Asn7l: GQDMVSPEATNSSSSSFSSPSSAGRHVRSYN [SEQ ID
N0:25];
2. Lys91-Ser109: KIEKNGKVSGTKKENCPYS [SEQ ID N0:26];
3. Asn135-Tyrl64: NKKGKLYGSKEFNNDCKLKERIEENGYNTY [SEQ ID
NO: 27]; and 4. Asnl 81-A1a199: NGKGAPRRGQKTRRKNTSA [SEQ ID N0:28].
Also, there are two additional shorter predicted antigenic areas, G1n74-Arg78 of Figure 1 (SEQ ID N0:2) and G1n170-G1n175 of Figure 1 {SEQ ID N0:2).
The epitope-bearing peptides and polypeptides of the invention may be produced by any conventional means for making peptides or polypeptides including recombinant means using nucleic acid molecules of the invention. For instance, a short epitope-bearing amino acid sequence may be fused to a larger polypeptide which acts as a Garner during recombinant production and purification, as well as during immunization to produce anti-peptide antibodies -Epitope-bearing peptides also may be synthesized using known methods of chemical synthesis. For instance, Houghten has described a simple method for synthesis of large numbers of peptides, such as 10-20 mg of 248 different 13 residue peptides representing single amino acid variants of a segment of the polypeptide which were prepared and characterized (by ELISA-type binding studies) in less than four weeks. Houghten, R. A. (1985) General method for the rapid solid-phase synthesis of large numbers of peptides: specificity of antigen-antibody interaction at the level of individual amino acids. Proc.
Natl.
Acad. Sci. USA 82:5131-5135. This "Simultaneous Multiple Peptide Synthesis (SMPS)" process is further described in U.S. Patent No. 4,631,211 to Houghten et al. (1986). In this procedure the individual resins for the solid-phase synthesis ofvarious peptides are contained in separate solvent-permeable packets, enabling the optimal use of the many identical repetitive steps involved in solid-phase methods. A completely manual procedure allows 500-1000 or more syntheses to - be conducted simultaneously. Houghten et al., supra, at 5134.

WO 99/41282 PCTlUS99/03018 Epitope-bearing peptides and polypepddes of the invention are used to induce antibodies according to methods well known in the art. See, for instance, Sutcliffe et al., supra; Wilson et al., supra; Chow, M. et al., Proc. Natl.
Acad. Sci. .
USA 82:910-914; and Bittle, F. J. et al., J. Gen. virol. 66:2347-2354 (1985).
Generally, animals may be immunized with free peptide; however, anti-peptide antibody titer may be boosted by coupling of the peptide to a macromolecular carrier, such as keyhole limpet hemacyanin {KLH) or tetanus toxoid. For instance, peptides containing cysteine may be coupled to carrier using a linker such as m-maleimidobenzoyl-N-hydroxysuccinimide ester (MBS), while other peptides may be coupled to carrier using a more general linking agent such as glutaraldehyde. Animals such as rabbits, rats and mice are immunized with either free or carrier-coupled peptides, for instance, by intraperitoneal andlor intradermal injection of emulsions containing about 100 pg peptide or Garner protein and Freund's adjuvant. Several booster injections may be needed, for instance, at intervals of about two weeks, to provide a useful titer of anti-peptide antibody which can be detected, for example, by ELISA assay using free peptide adsorbed to a solid surface. The titer of anti-peptide antibodies in serum from an immunized animal may be increased by selection of anti-peptide antibodies, for instance, by adsorption to the peptide on a solid support and elution of the selected antibodies according to methods well known in the art.
Immunogenic epitope-bearing peptides of the invention, i.e., those parts of a protein that elicit an antibody response when the whole protein is the immunogen, are identified according to methods known in the art. For instance, Geysen et al., supra, discloses a procedure for rapid concurrent synthesis on solid supports of hundreds of peptides of sufficient purity to react in an enzyme-linked immunosorbent assay. Interaction of synthesized peptides with antibodies is then easily detected without removing them from the support. In this manner a peptide bearing an immunogenic epitope of a desired protein may be identified routinely by one of ordinary skill in the art. For instance, the immunologically important epitope in the coat protein of foot-and-mouth disease virus was located by Geysen et al. with a resolution of seven amino acids by synthesis of an overlapping set of all 208 possible hexapeptides covering the entire 213 amino acid sequence of the protein. Then, a complete replacement set of peptides in which all 20 amino acids were substituted in turn at every position within the epitope were synthesized, and the particular amino acids conferring specificity for the reaction with antibody were determined. Thus, peptide analogs of the epitope-bearing peptides of the invention can be made routinely by this method. U.S. Patent No.
4,708,781 to Geysen ( 1987) further describes this method of identifying a peptide bearing an immunogenic epitope of a desired protein.
Further still, U.S. Patent No. 5,194,392 to Geysen (1990) describes a general method of detecting or determining the sequence of monomers (amino acids or other compounds) which is a topological equivalent of the epitope (i.e., a "mimotope") which is complementary to a particular paratope (antigen binding site) of an antibody of interest. More generally, U.S. Patent No. 4,433,092 to Geysen (1989) describes a method of detecting or determining a sequence of monomers which is a topographical equivalent of a ligand which is complementary to the ligand binding site of a particular receptor of interest.
Similarly, U.S. Patent No. 5,480,971 to Houghten, R. A, et al. (1996) on Peralkylated Oligopeptide Mixtures discloses linear C,-C~-alkyl peralkylated oligopeptides and sets and libraries of such peptides, as well as methods for using such oligopeptide sets and libraries for determining the sequence of a peralkylated oligopeptide that preferentially binds to an acceptor molecule of interest.
Thus, non-peptide analogs of the epitope-bearing peptides of the invention also can be made routinely by these methods.
As one of skill in the art will appreciate, KGF-2 polypeptides of the present invention and the epitope-bearing fraginents thereof described above can be combined with parts of the constant domain of immunoglobulins (IgG), resulting in chimeric polypeptides. These fusion proteins facilitate purification and show an increased half life in vivo. This has been shown, e.g., for chimeric proteins consisting of the first two domains of the human CD4-polypeptide and various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins (EPA 394,827; Traunecker et al., Nature 331:84-86 (1988)). Fusion proteins that have a disulfide-linked dimeric structure due to the IgG part can also be more efficient in binding and neutralizing other molecules than the monomeric KGF-2 protein or protein fragment alone (Fountoulakis et al., JBiochem 270:3958-39b4 (1995)).
In accordance with the present invention, novel variants of KGF-2 are also described. These can be produced by deleting or substituting one or more amino acids of KGF-2. Natural mutations are called allelic variations. Allelic variations can be silent (no change in the encoded polypeptide) or may have altered amino acid sequence. -In order to attempt to improve or alter the characteristics of native KGF-2, protein engineering may be employed. Recombinant DNA technology known to those skilled in the art can be used to create novel polypeptides. Muteins and deletions can show, e.g., enhanced activity or increased stability. In addition, they could be purified in higher yield and show better solubility at least under certain purification and storage conditions. Set forth below are examples of mutations that can be constructed.
Amino terminal and carboxy terminal deletions Various members of the FGF family have been modified using recombinant DNA technology. Positively charged molecules have been substituted or deleted in both aFGF and bFGF that are important for heparin binding. The modified molecules resulted in reduced heparin binding activity.
Accordingly, it is known that the amount of modified molecule sequestered by heparin in a patient would be reduced, increasing the potency as more FGF
would reach the appropriate receptor. (EP 0 298 723).
Native KGF-2 is relatively unstable in the aqueous state and it undergoes chemical and physical degradation resulting in loss of biological activity during processing and storage. Native KGF-2 is also prone to aggregation in aqueous solution, at elevated temperatures and it becomes inactivated under acidic conditions.
In order to improve or alter one or more characteristics of native KGF-2, protein engineering may be employed. Ron et al., J. Biol. Chem., 268(4):
2984-2988 (1993) reported modified KGF prnteins that had heparin binding activity even if the 3, 8, or 27 amino terminal amino acid residues were missing.
The deletion of 3 and 8 amino acids had full activity. More deletions of KGF
have been described in PCT/IB95/00971. The deletion ofcarboxyterminal amino acids can enhance the activity of proteins. One example is interferon gamma that shows up to ten times higher activity by deleting ten amino acid residues from the carboxy terminus of the protein (Dtibeli et al., J. of Biotechnology 7:199-216 (1988)). Thus, one aspect of the invention is to provide polypeptide analogs of KGF-2 and nucleotide sequences encoding such analogs that exhibit enhanced stability (e.g., when exposed to typical pH, thermal conditions or other storage conditions) relative to the native KGF-2 polypeptide.
Particularly preferred KGF-2 polypeptides are shown below (numbering starts with the first amino acid in the protein (Met) (Figure 1 (SEQ ID N0:2))):

Thr (residue 36) -- Ser (residue Asn (51 )-- Ser (208) 208) Cys (37) -Ser (208) Ser (52) -- Ser (208) Gln (38) -- Ser (208) Ser (53) -- Ser (208) Ala (39) -- Ser (208) Ser (54) -- Ser (208) Leu (40) -- Ser (208) Ser (55) -- Ser (208) Gly (41) -- Ser (208) Ser (56) -- Ser (208) Gln (42) -- Ser (208) Phe (57) -- Ser (208) Asp (43) -- Ser (208) Ser (59) -- Ser (208) Met (44) -- Ser (208) Ser(62) -- Ser (208) Val (45) -- Ser (208) Ala(63) -- Ser (208) Ser (46) -- Ser (208) Gly (64) -- Ser (208) Pro (47) -- Ser (208) Arg (65) -- Ser (208) Glu (48) - Ser (208) Val (67) -- Ser (208) Ala (49} -- (Ser (208) Ser (69) -- 5er (208) Thr (50) -- Ser (208) Val (77) -- Ser (208) Arg (80) - Ser (208) Met(1), Thr (36), or Cys (37) -- His (207) Met ( 1 ), Thr (36), or Cys (37) -- Val (206) Met (1), Thr (36), or Cys (37) -- Val (205) Met( 1 ), 'Thr (36), or Cys (37) -- Met (204) Met(I), Thr (36), or Cys (37) -- Pro (203) Met(1), 'Thr (36), or Cys(37) -- Leu (202) Met( 1 ), Thr (36), or Cys (37) -- Phe (201 ) Met(1), Thr (36), or Cys (37) -- His (200) Met(1), Thr (36), or Cys (37) -- Ala (199) Met (1), Thr (36), or Cys (37) -- Ser (198) Met (1), Thr (36), or Cys (37) -- Thr (197) Met(1), Thr (36), or Cys (37) --Asn (196) Met(1), Thr (36), or Cys (37) -- Lys (195) Met (1), Thr (36), or Cys (37) -- Arg (194) Met{1), Thr (36), or Cys (37) -- Arg (I93) Met{1), Tlu (36), or Cys (37) -- Thr (192) Met(1), Thr (36), or Cys (37) - Lys (191) Met(1), Thr (36), or Cys (37) - Arg (188) Met(1), Thr (36), or Cys (37) - Arg (187) Met(1), 'Thr (36), or Cys (37) - Lys (183) Preferred embodiments include the N-terminal deletions Ala (63) -- Ser (208) (KGF-2028) (SEQ ID N0:68) and Ser (69) -- Ser (208) (KGF-233) (SEQ
ID N0:96). Other preferred N-terminal and C-terminal deletion mutants are described in Examples 13 and 16 (c) of the specification and include: Ala {39) --Ser (208) (SEQ ID N0:116); Pro (47) -- Ser (208) of Figure 1 (SEQ ID N0:2);
Val (77) -- Ser (208) (SEQ ID N0:70); Glu (93) -- Ser (208) (SEQ ID N0:72);
Glu (104) -- Ser (208) (SEQ ID N0:74); Val (123) - Ser (208) (SEQ ID N0:76);
and Gly (138) -- Ser (208) (SEQ ID N0:78). Other preferred C-terminal deletion mutants include: Met (1 ), Thr (36), or Cys (37) -- Lys (153) of Figure 1 (SEQ
ID
N0:2).
Also included by the present invention are deletion mutants having amino acids deleted from both the N- terminus and the C-terminus. Such mutants include all combinations of the N-terminal deletion mutants and C-terminal deletion mutants described above, e.g., Ala (39) -- His (200) of Figure 1 (SEQ
ID
N0:2), Met (44) -- Arg (193) of Figure 1 (SEQ ID N0:2), Ala (63) -- Lys (I53) of Figure 1 (SEQ ID N0:2), Ser (69) - Lys ( 153) of Figure 1 (SEQ ID N0:2), etc.
etc. etc . . . . Those combinations can be made using recombinant techniques known to those skilled in the art.
Thus, in one aspect, N-terminal deletion mutants are provided by the present invention. Such mutants include those comprising the amino acid sequence shown in Figure 1 (SEQ ID N0:2) except for a deletion of at Least the first 38 N-terminal amino acid residues (i.e., a deletion of at least Met (1) -- GIn (38)) but not more than the first 147 N-terminal amino acid residues of Figure (SEQ ID N0:2). Alternatively, the deletion will include at least the first 38 N-terminal amino acid residues (i.e., a deletion of at least Met (1) -- Gln (38)) but not more than the first 137 N-terminal amino acid residues of Figure 1 (SEQ ID
N0:2). Alternatively, the deletion will include at least the first 46 N-terminal amino acid residues but not more than the first 137 N-terminal amino acid residues of Figure 1 (SEQ ID N0:2). Alternatively, the deletion will include at least the first 62 N-terminal amino acid residues but not more than the first N-terminal amino acid residues of Figure 1 (SEQ ID N0:2). Alternatively, the deletion will include at least the first 68 N-terminal amino acid residues but not more than the first 137 N-terminal amino acid residues of Figure 1 (SEQ ID
N0:2). Alternatively, the deletion will include at Least the first 76 N-terminal amino acid residues but not more than the first 137 N-terminal amino acid residues of Figure 1 (SEQ ID N0:2). Alternatively, the deletion will include at least the first 92 N-terminal amino acid residues but not more than the first N-terminal amino acid residues of Figure 1 (SEQ ID N0:2). Alternatively, the deletion will include at least the first 103 N-terminal amino acid residues but not more than the first 137 N-terminal amino acid residues of Figure 1 (SEQ ID
N0:2). Alternatively, the deletion will include at least the first 122 N-terminal amino acid residues but not more than the first 137 N-terminal amino acid residues of Figure 1 (SEQ ID N0:2}.
In addition to the ranges of N-terminal deletion mutants described above, the present invention is also directed to all combinations of the above described ranges, e.g., deletions of at least the first 62 N-terminal amino acid residues but not more than the first 68 N-terminal amino acid residues of Figure 1 (SEQ ID
N0:2); deletions of at least the first 62 N-terminal amino acid residues but not more than the first 76 N-terminal amino acid residues of Figure 1 (SEQ ID
N0:2); deletions of at least the first 62 N-terminal amino acid residues but not more than the first 92 N-terminal amino acid residues of Figure 1 (SEQ ID
N0:2); deletions of at least the first 62 N-terminal amino acid residues but not more than the first 103 N-terminal amino acid residues of Figure 1 (SEQ ID
N0:2); deletions of at least the first 68 N-terminal amino acid residues but not more than the first 76 N-terminal amino acid residues of Figure 1 (SEQ ID
N0:2}; deletions of at least the first 68 N-terminal amino acid residues but not more than the first 92 N-terminal amino acid residues of Figure 1 (SEQ ID
N0:2); deletions of at least the first 68 N-terminal amino acid residues but not more than the first 103 N-terminal amino acid residues of Figure 1 (SEQ ID
N0:2}; deletions of at least the first 46 N-terminal amino acid residues but not more than the first 62 N-terminal amino acid residues of Figure 1 (SEQ ID
N0:2); deletions of at least the first 46 N-terminal amino acid residues but not more than the first 68 N-terminal amino acid residues of Figure 1 (SEQ ID
N0:2); deletions of at least the first 46 N-terminal amino acid residues but not more than the first 76 N-terminal amino acid residues of Figure 1 {SEQ ID
N0:2); etc. etc. etc. . . .
In another aspect, C-terminal deletion mutants are provided by the present invention. Preferably, the N-terminal amino acid residue of said C-terminal deletion mutants is amino acid residue 1 (Met), 36 (Thr}, or 37 (Cys) of Figure 1 (SEQ ID N0:2). Such mutants include those comprising the amino acid sequence shown in Figure 1 (SEQ ID N0:2) except for a deletion of at least the last C-terminal amino acid residue (Ser (208)) but not more than the last 55 C-terminal amino acid residues (i.e., a deletion of amino acid residues Glu (154) -Ser (208}) of Figure 1 (SEQ ID N0:2). Alternatively, the deletion will include at least the last C-terminal amino acid residue but not more than the last 65 C-terminal amino acid residues of Figure 1 (SEQ ID N0:2). Alternatively, the deletion will include at least the last 10 C-terminal amino acid residues but not more than the last 55 C-terminal amino acid residues of Figure 1 {SEQ ID
N0:2).
Alternatively, the deletion will include at least the last 20 C-terminal amino acid residues but not more than the last 55 C-terminal amino acid residues of Figure 1 (SEQ ID N0:2). Alternatively, the deletion will include at least the last 30 C-terminal amino acid residues but not more than the last 55 C-terminal amino acid residues of Figure 1 (SEQ ID N0:2). Alternatively, the deletion will include at least the last 40 C-terminal amino acid residues but not more than the last 55 C-terminal amino acid residues of Figure 1 {SEQ ID N0:2). Alternatively, the deletion will include at least the last 50 C-terminal amino acid residues but not 1 S more than the last 55 C-terminal amino acid residues of Figure 1 {SEQ ID
N0:2).
In addition to the ranges of C-terminal deletion mutants described above, the present invention is also directed to all combinations of the above described ranges, e.g., deletions of at least the last C-terminal amino acid residue but not more than the last 10 C-terminal amino acid residues of Figure 1 (SEQ ID
N0:2);
deletions of at least the last C-terminal amino acid residue but not more than the last 20 C-terminal amino acid residues of Figure 1 (SEQ ID N0:2); deletions of at least the last C-terminal amino acid residue but not more than the last 30 C-terminal amino acid residues of Figure 1 (SEQ ID N0:2); deletions of at least the last C-terminal amino acid residue but not more than the last 40 C-terminal amino _ acid residues of Figure 1 (SEQ ID N0:2); deletions of at least the Iast IO C-terminal amino acid residues but not more than the last 20 C-terminal amino acid residues of Figure 1 (SEQ ID N0:2); deletions of at least the last 10 C-terminal amino acid residues but not more than the last 30 C-terminal amino acid residues of Figure 1 (SEQ ID N0:2); deletions of at least the last 10 C-terminal amino acid residues but not more than the last 40 C-terminal amino acid residues of Figure 1 (SEQ ID N0:2); deletions of at least the last 20 C-terminal amino acid residues but not more than the last 30 C-terminal amino acid residues of Figure 1 (SEQ
ID
N0:2); etc. etc. etc. . . . .
In yet another aspect, also included by the present invention are deletion mutants having amino acids deleted from both the N- terminal and C-terminal residues. Such mutants include all combinations of the N-terminal deletion mutants and C-terminal deletion mutants described above. Such mutants include those comprising the amino acid sequence shown in Figure 1 (SEQ ID N0:2) except for a deletion of at least the first 46 N-terminal amino acid residues but not more than the first 137 N-terminal amino acid residues of Figure 1 (SEQ ID
N0:2) and a deletion of at least the last C-terminal amino acid residue but not more than the last 55 C-terminal amino acid residues of Figure 1 (SEQ ID
N0:2).
Alternatively, a deletion can include at least the first 62, 68, 76, 92, 103, or 122 N-terminal amino acids but not more than the first 137 N-terminal amino acid residues of Figure 1 (SEQ ID N0:2) and a deletion of at least the last 10, 20, 30, 40, or 50 C-terminal amino acid residues but not more than the last 55 C-terminal amino acid residues of Figure 1 (SEQ ID N0:2). Further included are all combinations of the above described ranges.
Substitution of amino acids A further aspect of the present invention also includes the substitution of amino acids. Native mature KGF-2 contains 44 charged residues, 32 of which carry a positive charge. Depending on the location of such residues in the protein's three dimensional structure, substitution of one or more of these clustered residues with amino acids carrying a negative charge or a neutral charge ~ may alter the electrostatic interactions of adjacent residues and may be useful to achieve increased stability and reduced aggregation of the protein.
Aggregation of proteins cannot only result in a loss of activity but be problematic when preparing pharmaceutical formulations, because they can be immunogenic (Pinckard etal., Clin. Exp. Immunol. 2:331-340 (1967), Robbins etal., Diabetes 36: 838-845 (1987), Cleland et al., Crit. Rev. Therapeutic Drug Carrier Systems 10: 307-377 ( 1993)). Any modification should give consideration to minimizing charge repulsion in the tertiary structure of the protein molecule. Thus, of special interest are substitutions of charged amino acid with another charge and with neutral or negatively charged amino acids. The latter results in proteins with a reduced positive charge to improve the characteristics of KGF-2. Such improvements include increased stability and reduced aggregation of the analog as compared to the native KGF-2 protein.
The replacement of amino acids can also change the selectivity of binding to cell surface receptors. Ostade et al., Nature 361: 266-26$ (1993), described certain TNF alpha mutations resulting in selective binding of TNF alpha to only one of the two known TNF receptors.
KGF-2 molecules may include one or more amino acid substitutions, deletions or additions, either from natural mutation or human manipulation.
Examples of some preferred mutations are: Ala (49) Gln, Asn (51 ) Ala, Ser (54) Val, Ala (63) Pro, Gly (64) Glu, Val (67) Thr, Trp (79) Val, Arg (80) Lys, Lys (87) Arg, Tyr (88) Trp, Phe (89) Tyr, Lys (91) Arg, Ser (99) Lys, Lys (102) Gln, Lys 103(Glu), Glu (104) Met, Asn (105) Lys, Pro (107) Asn, Ser (109) Asn, Leu {111) Met, Thr (114) Arg, Glu(117) Ala, Val (120) Ile, Val (123) Ile, Ala (125) Gly, Ile (126) Val, Asn (127) Glu, Asn (127) Gln, Tyr (130) Phe, Met (134) Thr, Lys (136) Glu, Lys (137) Glu, Gly (142) Ala, Ser (143) Lys, Phe (146) Ser, Asn (148) Glu, Lys (151) Asn, Leu (152) Phe, Glu (154) Gly, Glu (154) Asp, Arg {155) Leu, Glu (157) Leu, Gly (160) His, Phe (167) Ala, Asn (168) Lys, Gln (170) Thr, Arg (174) Gly, Tyr (177) Phe, Gly (182) Gln, Ala (185) Val, Ala (185) Leu, Ala (185) Ile, Arg (187) Gln (190) Lys, Lys (195) Glu, Thr (197} Lys, Ser ( 198) Thr, Arg ( 194) Glu, Arg ( 194) Gln, Lys ( 191 ) Glu, Lys ( 191 ) Gln, Arg (188) Glu, Arg (188) Gln, Lys (183) Glu.
By the designation, for example, Ala (49) Gln is intended that the Ala at position 49 of Figure 1 (SEQ ID N0:2) is replaced by Gln.

Changes are preferably of minor nature, such as conservative amino acid substitutions that do not significantly affect the folding or activity of the protein.
Examples of conservative amino acid substitutions known to those skilled in the art are set forth below:
Aromatic: phenylalanine tryptophan tyrosine Hydrophobic: leucine isoleucine valine Polar: glutamine asparagine Basic: arginine lysine histidine Acidic: aspartic acid glutamic acid Small: alanine serine threonine methionine glycine Of course, the number of amino acid substitutions a skilled artisan would make depends on many factors, including those described above. Generally speaking, the number of substitutions for any given KGF-2 polypeptide will not be more than 50, 40, 30, 20,10, 5, or 3, depending on the objective. For example, a number of substitutions that can be made in the C-terminus of KGF-2 . to improve stability are described above and in Example 22.
Amino acids in KGF-2 that are essential for function can be identified by methods well known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis (Cunningham and Wells, Science 244 :1081-1085 { 1989). The latter procedure introduces single alanine mutations at every residue in the molecule. The resulting mutant molecules are then tested for biological activity such as receptor binding or in vitro and in vivo proliferative activity.
(See, e.g., Examples 10 and 11 ). Sites that are critical for ligand-receptor binding can also be determined by structural analysis such as crystallization, nuclear magnetic resonance or photoaffinity labeling. (See for example: Smith et al., J.
Mol. Biol., 224: 899-904 (1992); and de Vos etal. Science, 255: 306-312 (1992).) Another aspect of the present invention substitutions of serine for cysteine at amino acid positions 3 7 and 106 and 150. An uneven number of cysteins means that at least one cysteine residue is available for intermolecular crosslinks or bonds that can cause the protein to adopt an undesirable tertiary structure.
Novel KGF-2 proteins that have one or more cysteine replaced by serine or e.g.
alanine are generally purified at a higher yield of soluble, correctly folded protein.
Although not proven, it is believed that the cysteine residue at position 106 is important for function. This cysteine residue is highly conserved among all other FGF family members.
A further aspect of the present invention are fusions of KGF2 with other proteins or fragments thereof such as fusions or hybrids with other FGF
proteins, e.g. KGF (FGF-7), bFGF, aFGF, FGF-5, FGF-6, etc. Such a hybrid has been reported for KGF (FGF-7). In the published PCT application no. 90/0877T a chimeric protein has been produced consisting of the first 40 amino acid residues of KGF and the C-terminal portion of aFGF. The chimera has been reported to target keratinocytes like KGF, but lacked susceptibility to heparin, a characteristic of aFGF but not KGF. Fusions with parts of the constant domain of immunoglobulins (IgG) show often an increased half life time in vivo. This has been shown, e.g., for chimeric proteins consisting of the first two domains of the human CD4-polypeptide with various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins (European Patent application, Publication No. 394 827, Traunecker et al., Nature 331, 84-86 ( 1988). Fusion proteins that have a disulfide-linked dimeric structure can also be more efficient in binding monomeric molecules alone (Fountoulakis et al., J.
of Biochemistry, 270: 3958-3964, {1995)).

Antigeniclhydrophilic parts of KGF 2 As demonstrated in Figure 4A-4E, there are 4 major highly hydrophilic regions in the KGF-2 protein. Amino acid residues G1y41 - Asn 71, Lys91 - Ser 109, Asn135 - Tyr 164 and Asn 181 - Ala 199 [SEQ ID NOS:25-28]. There are two additional shorter predicted antigenic areas, GIn 74 - Arg 78 of Figure 1 (SEQ ID N0:2) and Gln 170 - Gln 175 of Figure 1 (SEQ ID N0:2). Hydrophilic parts are known to be mainly at the outside (surface) of proteins and, therefore, available for antibodies recognizing these regions. Those regions are also likely to be involved in the binding of KGF-2 to its receptor(s). Synthetic peptides derived from these areas can interfere with the binding of KGF-2 to its receptors) and, therefore, block the function of the protein. Synthetic peptides from hydrophilic parts of the protein may also be agonistic, i.e. mimic the function of KGF-2.
Thus, the present invention is further directed to isolated polypeptides comprising a hydrophilic region of KGF-2 wherein said polypeptide is not more than 150 amino acids in length, preferably not more than 100, 75, or 50 amino acids in length, which comprise one or more of the above described KGF-2 hydrophilic regions.
Chemical modifications The KGF wild type and analogs may be further modified to contain additional chemical moieties not normally part of the protein. Those derivatized moieties may improve the solubility, the biological half life or absorption of the protein. The moieties may also reduce or eliminate any desirable side effects of the proteins and the like. an overview for those moieties can be found in REMINGTONS PHfARMACEUTICAL SCIENCES,18th ed., Mack Publishing Co., Easton, PA (1990). Polyethylene glycol (PEG) is one such chemical moiety which has been used for the preparation of therapeutic proteins. The attachment of PEG to proteins has been shown to protect against proteolysis, Sada et al., J.
Fermentation Bioengineering 71:137-139 ( 1991 ). Various methods are available for the attachment of certain PEG moieties. For review, see: Abuchowski et al., in Enzymes as Drugs. (Holcerberg and Roberts, eds.) pp. 367- 383 (1981). Many published patents describe derivatives of PEG and processes how to prepare them, e.g., Ono et aI.U.S. Patent No. 5,342,940; Nitecki et aI.U.S. Patent No.
5,089,261; Delgado et aI.U.S. Patent No. 5,349,052. Generally, PEG molecules are connected to the protein via a reactive group found on the protein. Amino groups, e.g. on lysines or the amino terminus of the protein are convenient for this attachment among others.
The entire disclosure of each document cited in this section on "Polypeptides and Peptides" is hereby incorporated herein by reference.
Vectors and Host Cells The present invention also relates to vectors which include the isolated DNA molecules of the present invention, host cells which are genetically engineered with the recombinant vectors, and the production of KGF-2 polypeptides or fragments thereof by recombinant techniques.
Fragments or portions of the polypeptides of the present invention may be employed for producing the corresponding full-length polypeptide by peptide synthesis; therefore, the fragments may be employed as intermediates for producing the full-length polypeptides. Fragments or portions of the polynucleotides of the present invention may be used to synthesize full-length polynucleotides of the present invention. T;he present invention also relates to vectors which include polynucleotides of the present invention, host cells which are genetically engineered with vectors of the invention and the production of polypeptides of the invention by recombinant techniques.
Host cells are genetically engineered (transduced or transformed or transfected) with the vectors of this invention which may be, for example, a cloning vector or an expression vector. The vector may be, for example, in the form of a plasmid, a viral particle, a phage, etc. The engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transformants or amplifying the KGF-2 genes. The culture conditions, such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
The polynucleotides of the present invention may be employed for producing polypeptides by recombinant techniques. Thus, for example, the poIynucleotide may be included in any one of a variety of expression vectors for expressing a polypeptide. Such vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., derivatives of SV40; bacterial plasmids;
phage DNA; baculovirus; yeast plasmids; vectors derived from combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, fowl pox virus, and pseudorabies. However, any other vector may be used as long as it is replicable and viable in the host.
The appropriate DNA sequence may be inserted into the vector by a variety of procedures. In general, the DNA sequence is inserted into an appropriate restriction endonuclease sites) by procedures known in the art.
Such procedures and others are deemed to be within the scope of those skilled in the art.
The DNA sequence in the expression vector is operatively linked to an appropriate expression control sequences) (promoter) to direct cDNA synthesis.
As representative examples of such promoters, there may be mentioned: LTR or SV40 promoter, the E. coli. lac or trp, the phage lambda PL promoter and other promoters known to control expression of genes in prokaryotic or eukaryotic cells or their viruses. The expression vector also contains a ribosome binding site for translation initiation and a transcription terminator. The vector may also include appropriate sequences for amplifying expression.

In addition, the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in E. coli.
The vector containing the appropriate DNA sequence as hereinabove described, as well as an appropriate promoter or control sequence, may be employed to transform an appropriate host to permit the host to express the protein.
As representative examples of appropriate hosts, there may be mentioned:
bacterial cells, such as E. coli, Streptomyces, Salmonella typhimurium; fungal cells, such as yeast; insect cells such as Drosophila S2 and Spodoptera Sf9;
animal cells such as CHO, COS or Bowes melanoma; adenoviruses; plant cells, etc. The selection of an appropriate host is deemed to be within the scope of those skilled in the art from the teachings herein.
More particularly, the present invention also includes recombinant constructs comprising one or more of the sequences as broadly described above.
The constructs comprise a vector, such as a plasmid or viral vector, into which a sequence of the invention has been inserted, in a forward or reverse orientation.
In a preferred aspect of this embodiment, the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence. Large numbers of suitable vectors and promoters are known to those of skill in the art, and are commercially available. The following vectors are provided by way of example; Bacterial: pQE70, pQE60, pQE-9 (Qiagen), pBS, pDlO, phagescript, psiXl74, pbluescript SK, pbsks, pNHBA, pNHl6a, pNHl8A, pNH46A (Stratagene); ptrc99a, pKK223-3, pKK233-3, pDR540, pRITS
(Phannacia); Eukaryotic: pWLNEO, pSV2CAT, pOG44, pXTl, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharmacia). However, any other plasmid or vector may be used as long as they are replicable and viable in the host.
The present invention further includes novel expression vectors comprising operator and promoter elements operatively linked to nucleotide sequences encoding a protein of interest. One example of such a vector is pHE4-5 which is described in detail below.
As shown in FIGs. 8 and 9, components of the pHE4-5 vector (SEQ ID
N0:147) include: 1 ) a neomycinphosphotransferase gene as a selection marker, 2) an E. coli origin of replication, 3) a TS phage promoter sequence, 4) two lac operator sequences, S} a Shine-Delgarno sequence, 6) the lactose operon repressor gene (lacIq). The origin of replication (oriC) is derived from pUCl9 (LTI, Gaithersburg, MD). The promoter sequence and operator sequences were made synthetically. Synthetic production of nucleic acid sequences is well known in the art. CLONTECH 95/96 Catalog, pages 215-216, CLONTECH, 1020 East Meadow Circle, Palo Alto, CA 94303. A nucleotide sequence encoding a KGF-2 polypeptide is operatively linked to the promoter and operator by inserting the nucleotide sequence between the NdeI and Asp718 sites of the pHE4-5 vector.
As noted above, the pHE4-5 vector contains a lacIq gene. LacIq is an allele of the lacI gene which confers tight regulation of the lac operator.
Amann, E. et al., Gene 69: 301-3I5 (1988); Stark, M., Gene 51:255-267 (1987). The lacIq gene encodes a repressor protein which binds to lac operator sequences and blocks transcription of down-stream (i. e., 3') sequences. However, the IacIq gene product dissociates from the lac operator in the presence of either lactose or certain lactose analogs, e.g., isopropyl B-D-thiogalactopyranoside (IPTG). KGF-thus is not produced in appreciable quantities in uninduced host cells containing the pHE4-5 vector. Induction of these host cells by the addition of an agent such as IPTG, however, results in the expression of the KGF-2 coding sequence.
The promoter/operator sequences of the pHE4-5 vector (SEQ ID N0:148) comprise a TS phage promoter and two lac operator sequences. One operator is located 5' to the transcriptional start site and the other is located 3' to the same site. These operators, when present in combination with the IacIq gene product, confer tight repression of down-stream sequences in the absence of a lac operon inducer, e.g., IPTG. Expression of operatively linked sequences located down-stream from the lac operators may be induced by the addition of a lac operon inducer, such as IPTG. Binding of a lac inducer to the IacIq proteins results in their release from the lac operator sequences and the initiation of transcription of operatively linked sequences. Lac operon regulation of gene expression iS reviewed iri Devlin, T., TEXTBOOK OF BIOCHEMISTRY WITH
CLINICAL CORRELATIONS, 4th Edition (1997), pages 802-807.
The pHE4 series of vectors contain all of the components of the pHE4-5 vector except for the KGF-2 coding sequence. Features of the pHE4 vectors include optimized synthetic TS phage promoter, lac operator, and Shine-Delagarno sequences. Further, these sequences are also optimally spaced so that expression of an inserted gene may be tightly regulated and high level of expression occurs upon induction.
Among known bacterial promoters suitable for use in the production of proteins of the present invention include the E. coli IacI and IacZ promoters, the T3 and T7 promoters, the gpt promoter, the lambda PR and PL promoters and the 1 ~' trp promoter. Suitable eukaryotic promoters include the CMV immediate early promoter, the HSV thymidine kinase promoter, the early and late SV40 promoters, the promoters of retroviral LTRs, such as those of the Rous Sarcoma Virus (RSV), and metallothionein promoters, such as the mouse metallothionein-I
promoter.
The pHE4-5 vector also contains a Shine-Delgarno sequence 5' to the AUG initiation codon. Shine-Delgarno sequences are short sequences generally located about 10 nucleotides up-stream (i. e. , 5') from the AUG initiation codon.
These sequences essentially direct prokaryotic ribosomes to the AUG initiation codon.
Thus, the present invention is also directed to expression vector useful for the production of the proteins of the present invention. This aspect of the invention is exemplified by the pHE4-5 vector (SEQ ID N0:147).
A cDNA encoding KGF-2 X33 inserted into the pHE4-5 expression vector was deposited at the ATCC on January 9, 1998 as ATCC No. 209575.
Promoter regions can be selected from any desired gene using CAT

(chloramphenicol transferase) vectors or other vectors with selectable markers.
Two appropriate vectors are pKK232-8 and pCM7. Particular named bacterial promoters include lacI, lacZ, T3, T7, gpt, lambda P~, PL and trp. Eukaryotic promoters include CMV immediate early, HSV thymidine kinase, early and late SV40, LTRs from retrovirus, and mouse metallothionein-I. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art.
In a further embodiment, the present invention relates to host cells containing the above-described constructs. The host cell can be a higher eukaryotic cell, such as a mammalian cell, or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell.
Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-Dextran mediated transfection, or electroporation (Davis, L. et al., Basic Methods in Molecular Biology (1986)).
1 S The constructs in host cells can be used in a conventional manner to produce the gene pmduct encoded by the recombinant sequence. Alternatively, the polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.
Mature proteins can be expressed in mammalian cells, yeast, bacteria, or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook et al., Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor, N.Y. ( 1989), the disclosure of which is hereby incorporated by reference.
Transcription of the DNA encoding the polypeptides of the present invention by higher eukaryotes is increased by inserting an enhancer sequence into the vector. Enhancers are cis-acting elements of DNA, usually about from 10 to 300 by that act on a promoter to increase its transcription. Examples including the SV40 enhancer on the late side of the replication origin by 100 to 270, a cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
For secretion of the translated protein into the lumen of the endoplasmic reticulum, into the periplasmic space or into the extracellular environment, appropriate secretion signals may be incorporated into the expressed polypeptide.
The signals may be endogenous to the polypeptide or they may be heterologous signals.
The polypeptide may be expressed in a modified form, such as a fusion protein, and may include not only secretion signals, but also additional heterologous functional regions. For instance, a region of additional amino acids, particularly charged amino acids, may be added to the N-terminus of the polypeptide to improve stability and persistence in the host cell, during purification, or during subsequent handling and storage. Also, peptide moieties may be added to the polypeptide to facilitate purification. Such regions may be removed prior to final preparation of the polypeptide. The addition of peph'de moieties to polypeptides to engender secretion or excretion, to improve stability and to facilitate purification, among others, are familiar and routine techniques in the art. A preferred fusion protein comprises a heterologous region from immunoglobulin that is useful to solubilize receptors. For example, EP-A-O 464 533 (Canadian counterpart 2045869) discloses fusion proteins comprising various portions of constant region of immunoglobin molecules together with another human protein or part thereof. In many cases, the Fc part in fusion protein is thoroughly advantageous for use in therapy and diagnosis and thus results, for example, in improved pharmacokinetic properties (EP-A 0232 262). On the other hand, for some uses it would be desirable to be able to delete the Fc pat after the fusion protein has been expressed, detected and purified in the advantageous manner described. This is the case when Fc portion proves to be a hindrance to use in therapy and diagnosis, for example when the fusion protein is to be used as antigen for immunizations. In drug discovery, for example, human proteins, such as, shILS-receptor has been fused with Fc portions for the purpose of high-throughput screening assays to identify antagonists of hIL-5. See, D. Bennett et al., Journal of Molecular Recognition, Vol. 8 52-58 (1995) and K. Johanson et al., The Journal ofBiological Chemistry, Vol. 270, No.16, pp 9459-9471 ( 1995).
Generally, recombinant expression vectors will include origins of replication and selectable markers permitting transformation of the host cell, e.g., the ampicillin resistance gene of E. coli and S. cerevisiae TRP1 gene, and a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence. Such promoters can be derived from operons encoding glycolytic enzymes such as 3-phosphoglycerate kinase (PGK), a-factor, acid phosphatase, or heat shock proteins, among others. The heterologous structural sequence is assembled in appropriate phase with translation initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein into the periplasmic space or extracellular medium.
Optionally, the heterologous sequence can encode a fusion protein including an N-terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product.
Useful expression vectors for bacterial use are constructed by inserting a structural DNA sequence encoding a desired protein together with suitable translation initiation and termination signals in operable reading phase with a functional promoter. The vector will comprise one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to, if desirable, pmvide amplification within the host. Suitable prokaryotic hosts for transformation include E. coli, Bacillus subtilis, Salmonella typhimurium and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus, although others may also be employed as a matter of choice.
As a representative but nonlimiting example, useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 37017). Such commercial vectors include, for example, pKK223-3 (Phatmacia Fine Chemicals, Uppsala, Sweden) and GEM1 (Promega Biotec, Madison, WI, USA) These pBR322 "backbone"
sections are combined with an appropriate promoter and the structural sequence to be expressed.
Following transformation of a suitable host strain and growth of the host strain to an appropriate cell density, the selected promoter is induced by appropriate means (e.g., temperature shift or chemical induction) and cells are cultured for an additional period.
Cells are typically harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification.
Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonicadon, mechanical disruption, or use of cell lysing agents, such methods are well known to those skilled in the art.
Various mannmalian cell culture systems can also be employed to express recombinant protein. Examples of mammalian expression systems include the COS-7 lines of monkey kidney fibroblasts, described by Gluzman, Cell 23:175 (1981), and other cell lines capable of expressing a compatible vector, for example, the C 127, 3T3, CHO, HeLa and BHK cell lines. Mammalian expression vectors will comprise an origin of replication, a suitable promoter and enhancer, and also any necessary ribosome binding sites, polyadenylation site, splice donor and acceptor sites, transcriptional termination sequences, and 5' flanking nontranscribed sequences. DNA sequences derived from the SV40 splice, and polyadenylation sites may be used to provide the required nontranscribed genetic elements.
The KGF-2 polypeptide can be recovered and purified from recombinant cell cultures by methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography.
Protein refolding steps can be used, as necessary, in completing configuration of the mature pmtein. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps.
The polypeptides of the present invention may be a naturally purified product, or a product of chemical synthetic procedures, or produced by recombinant techniques from a prokaryotic or eukaryotic host (for example, by bacterial, yeast, higher plant, insect and mammalian cells in culture).
Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated.
Polypeptides of the invention may also include an initial methionine amino acid residue.
Therapeutic Applications of ~GF 2 As used in the section below, "KGF-2" is intended to refer to the full-length and mature forms of KGF-2 described herein and to the KGF-2 analogs, derivatives and mutants described herein.
Increase levels of Platelets In one aspect, the invention is directed to increasing platelet level or number in blood for the purpose of alleviating thrombocytopenia. Determining the true cause of thrombocytopenia is a difficult and challenging clinical problem.
Thrombocytopenia results from various causes, but ultimately occurs when platelets are destroyed, sequestered in the body, or not produced. The differential diagnosis of thrombocytopenia is extensive and complex, and there is a significant overlap among disorders (Doyle B. and Porter D.L., A.A.C.N. Clin.
Issues 1997 Aug; 8(3): 469-480). Thrombocytopenia may be caused by a variety of mechanisms including, but not limited to, drug induced hypersensitivity, idiopathic thrombocytopenia pupura (ITP), posttransfusion purpura, neonatal thrombocytopenia, bone marrow deficiencies identified with metastatic tumors to the bone, aplastic anemia, myelofibrosis, acute and monocytic leukemia, microangiopathic hemolytic anemia which includes disseminated intravascular coagulation (DIC), thrombotic thrombocytopenic purpura (TTP), hemolytic-uremic syndrome, prosthetic valve hemolytic syndrome, cancer chemotherapy, Zieve's syndrome, sepsis, HELLP preeclamptic syndrome, megaloblastic anemia due to B21 and folic acid deficiency, infections such as peritonitis (without septicemia), congenital rubella syndrome, HIV-1 virus infections, Epstein-Barr infectious mononucleosis, rheumatoid-collagen diseases such as systemic lupus, hypertension of pregnancy associated with preeclampsia, thyrotoxicosis and uremia (Clinical guide to laboratory tests. (3rd ed). Philadelphia, W.B.
Saunders Company, 1995; Clinical Laboratory Medicine. Clinical application of laboratory data. (6th ed.) St. Louis, Mosby, 1995).
Accordingly, the present invention provides a process for administering KGF-2 polypeptides to an individual for alleviating thrombocytopenia. Suitable doses, formulations, and administration routes are described below.
Increase levels of Fibrinogen, Album~tn and Serum Proteins Fibrinogen is a glycoprotein that is synthesized in the liver. Fibrinogen can give rise to fibrin split products (FDPs) in the following manner.
Normally, thrombin catalyzes conversion of fibrinogen to fibrin by splitting two fibrinopeptide molecules, known as fibrinopeptide A and B, from the central portion of fibrinogen. This action exposes polymerization sites on the remaining portion of the fibrinogen molecule, which is now called fibrin monomer. Fibrin monomers spontaneously aggregate together in side-to-side and end-to-end configurations to form a fibrin gel. Thrombin also activates factor XIII, which helps introduce cmss-linking isopeptide bonds between the fibrin monomers to form stabilized insoluble fibrin polymer. Fibrin polymer forms the scaffolding for blood clots. Fibrinolysins are naturally occurring or acquired enzymes that may attack either fibrinogen ar fibrin or both, splitting off FDPs, which, in turn, are broken into smaller pieces. These split products may form a complex with fibrin monomers and interfere with polymerization. Fibrinogen degradation products can be produced by action of primary fibrinolysin on fibrinogen and also by action of plasmin on fibrinogen and fibrin monomers or fibrin clots formed in a variety of conditions, normal and abnormal. Plasmin attacks intravascular blood clots formed as part of normal hemostasis (e.g., trauma or surgery) as well as blood clots that produce disease (e.g., thrombosis or embolization).
Decreased plasma fibrinogen levels may occur from decreased liver production resulting from acute hepatitis or cirrhosis, from the action of fibrinolysins which are enzymes that destroy fibrin and may also attack fibrinogen, and from conversion of fibrinogen to fibrin that is too extensive to permit adequate replacement of the fibrinogen. Decreased levels of plasma fibrinogen may be found in conditions of abnormal hepatic synthesis such as that associated with acute hepatitis or cirrhosis. The major etiology of hypofibrinogenemia other than severe liver disease is fibrinogen depletion caused by disseminated intravascular coagulation (DIC)(Clinical guide to laboratory tests. (3rd ed). Philadelphia, W.B. Saunders Company,1995; Clinical Laboratory Medicine. Clinical application of laboratory data. (6th ed.) St. Louis, Mosby, 1995}.
Accordingly, in a further aspect, there is provided a process for utilizing KGF-2 polypeptides to enhance or increase the fibrinogen level or number in plasma. Preferably, a KGF-2 polypeptide is administered to an individual for the purpose of alleviating hypofibrinogenemia. Suitable doses, formulations, and administration routes are described below.
Albumin is a serum protein produced by the liver which is most active in maintaining the serum oncotic pressure. Serum albumin also acts as a transport protein for some drugs and a few other substances. Hypoalbuminemia can be caused by a variety of mechanisms including, but not limited to, hemorrhages, burns, exudates, rheumatic diseases, granulomatous processes, most bacterial infections, viral infections accompanied by tissue destruction, tissue necrosis, vasculitis, ulcerative bowel disease, serositis, subacute bacterial endocarditis, parasitic infestations, acute and chronic liver disease, amyloidosis, malnutrition, malignancy, congestive heart failure, constrictive pericarditis, cardiac valvular disease, nephrotic syndrome, trauma and crush injuries, gastrointestinal and lymphatic fistulae, and protein-losing gastroenteropathies (Clinical guide to laboratory tests. (3rd ed). Philadelphia, W.B. Saunders Company,1995; Clinical LaboratoryMedicine. Clinicalapplicationoflaboratorydata. (6thed.)St.Louis, Mosby, 1995).
Accordingly, in a fiuther aspect, there is provided a process for utilizing KGF-2 polypeptides to enhance or increase the level or number of serum albumin. Preferably, a KGF-2 polypeptide is administered to an individual for the purpose of alleviating hypoalbuminemia. Suitable doses, formulations, and administration routes are described below.
Serum globulin refers to a heterologous group of proteins such as glycoproteins, lipoproteins, and immunoglobulins. The globulins form the main transport system for various substances as well as constituting the antibody system, the clotting proteins, complement, and certain special duty substances such as the "acute reaction" proteins. Hypoglobulinemia may result from a number of conditions or afflictions including, but not limited to, alpha-1 antityrpsin deficiencies, severe liver disease, estrogen therapy, megaloblastic anemia, hypogammaglobulinemia and aggammaglobuinemia (Clinical guide to laboratory tests. (3rd ed). Philadelphia, W.B. Saunders Company, l 995;
Clinical LaboratoryMedicine. Clinical applicationoflaboratorydata. (tithed.) St. Louis, Mosby, 1995).
Accordingly, in a further aspect, there is provided a process for utilizing KGF-2 polypeptides to enhance or increase the level or number of serum globulin. Preferably, a KGF-2 polypeptide is administered to an individual for the purpose of alleviating hypoglobulinemia. Suitable doses, formulations, and administration routes are described below.
KGF-2 can also be used to increase the levels of total serum protein in individuals with protein loss and/or decreased protein synthesis. Human serum contains many proteins including albumin, globulins, prothrombin, fibrinogen, WO 99/41282 PCT/t3S99/03018 and other proteins synthesized exclusively by hepatocytes. Extensive liver injury may lead to decreased levels of these proteins (Clinical guide to laboratory tests.
(3rd ed). Philadelphia, W.B. Saunders Company, 1995; Clinical Laboratory Medicine. Clinical application of laboratory data. (6th ed.) St. Louis, Mosby, 1995). Serum albumin levels are influenced by a variety of nonhepatic factors, most notably nutritional status, hormonal factors, and plasma oncotic pressure.
Nephrotic syndrome or protein-losing enteropathy may lead to depressed serum albumin levels. Low serum albumin levels are also caused by familial idiopathic dysproteinemia, a rare genetic condition in which the albumin Level is greatly decreased while all the globulin fractions are elevated (Clinical guide to laboratory tests. (3rd ed). Philadelphia, W.B. Saunders Company,1995; Clinical LaboratoryMedicine. Clinical application oflaboratorydata. (6th ed.) St.
Louis, Mosby, 1995). Serum globulins include alpha and beta globulins as well as serum immunoglobulins. Alpha-1 globulins include alpha-1 antitrypsin, alpha-1 acid glycoprotein, alpha-1-fetoprotein, and certain carrier proteins such as cortisol-binding protein (transcortin) and thyroxine-binding globulin. Alpha-1 globulin is absent or nearly so in alpha-1 antitrypsin deficiency (Clinical guide to laboratory tests. (3rd ed). Philadelphia, W.B. Saunders Company, 1995;
Clinical LaboratoryMedicine. Clinical application oflaboratory data. (6th ed.) St. Louis, Mosby, 1995). Alpha-2 globulins include haptoglobulin, alpha-2 macroglobulin, and ceruloplasmin. Haptoglobulin levels are decreased in severe liver disease, in patients on estrogen therapy, in megaloblastic anemia, and also in conditions in which free hemoglobin appears in the blood (RBC hemolysis) (Clinical guide to laboratory tests. (3rd ed). Philadelphia, W.B. Saunders Company, 1995; Clinical Laboratory Medicine. Clinical application of laboratory data. (6th ed.) St. Louis, Mosby, 1995). Ceruloplasmin levels are decreased in Wilson's disease, malnutrition, nephrotic syndrome, and protein losing enteropathy. Beta globulins include transferrin, beta-lipoprotein, and several components of complement. Transferrin is frequently decreased in protein malnutrition. Gamma globulins include immunoglobulins IgG, IgA, IgM, IgD, and IgE. The gammaglobulins are decreased in hyporganunaglobulinemia and agammaglobulinemia (Clinical guide to laboratory tests. (3rd ed).
Philadelphia, W.B. Saunders Company, 1995; Clinical Laboratory Medicine.
Clinical application of laboratory data. (6th ed.) St. Louis, Mosby, 1995).
Finally, serum contains a number of enzymes which have been used to distinguish and assess hepatocellular injury and biliary tract dysfunction or obstruction.
For example, aspartate aminotransferase (AST) and alanine aminotransferase (ALT) catalyze the transfer of the 'y-amino groups of aspartate and alanine, respectively, to the y-keto groups of ketoglutarate, leading to the formation of oxaloacetic acid and pyruvic acid. Uremia may lead to spuriously low aminotransferase levels (Harrison's Principles of Internal Medicine 11 th edition Eugene Braunwald et.
al., Editors (1987)).
Thus, total protein loss can be caused by a variety of mechanisms including, but not limited to, protein-losing gastroenteropathies, acute burns, and nephrotic syndrome. Decreased protein synthesis can be caused by a variety of mechanisms including, but not limited to, chronic liver disease, malabsorption syndrome, malnutrition, and agammaglobulinemia. Accordingly, in a further aspect, there is provided a process for utilizing KGF-2 polypeptides to enhance or increase the level or number of total serum protein. Preferably, a KGF-2 polypeptide is administered to an individual for the purpose of alleviating diseases or conditions associated with protein loss. Suitable doses, formulations, and administration routes are described below.
Treatment ojCystitis Hemorrhagic cystitis is a syndrome associated with certain disease states as well as exposure to drugs, viruses, and toxins. It manifests as diffuse bleeding of the endothelial lining of the bladder. Known treatments include intravesical, systemic, and nonphannacologic therapies (West, N.J., Phannacotherapy 17:696-706 (1997). Some cytotoxic agents used clinically have side effects resulting in the inhibition of the proliferation of the normal epithelial in the bladder, leading to potentially life-threatening ulceration and breakdown in the epithelial lining. For example, cyclophosphamide is a cytotoxic agent which is biotransformed principally in the liver to active alkylating metabolites by a mixed function microsomal oxidase system. These metabolites interfere with the growth of susceptible rapidly proliferating malignant cells. The mechanism of action is believed to involve cross-linking of tumor cell DNA (Physicians' Desk reference, 1997).
Cyclophosphamide is one example of a cytotoxic agent which causes hemorrhagic cystitis in some patients, a complication which can be severe and in some cases fatal. Fibrosis of the urinary bladder may also develop with or without cystitis. This injury is thought to be caused by cyclophosphamide metabolites excreted in the urine. Hematuria caused by cyclophosphamide usually is present for several days, but may persist. In severe cases medical or surgical treatment is required. Instances of severe hemorrhagic cystitis result in discontinued cyclophosphamide therapy. In addition, urinary bladder malignancies generally occur within two years of cyclophosphamide treatment and occurs in patients who previously had hemorrhagic cystitis (CYTOXAN
(cyclophosphamide) package insert). Cyclophosphamide has toxic effects on the prostate and male reproductive systems. Cyclophosphamide treatment can result in the development of sterility, and result in some degree of testicular atrophy.
As shown in Figures 23 and 24, systemic administration of KGF-2 to an individual stimulates proliferation of bladder and prostatic epithelial cells.
Thus, in one aspect, the present invention provides a method of stimulating proliferation of bladder epithelium and prostatic epithelial cells by administering to an individual an effective amount of a KGF-2 polypeptide. More importantly, as Figures 25 and 26 demonstrate, KGF-2 can be used to reduce damage caused by cytotoxic agents having side effects resulting in the inhibition of bladder and prostate epithelial cell proliferation. To reduce such damage, KGF-2 can be administered either before, after, or during treatment with or exposure to the cytotoxic agent. Accordingly, in a further aspect, there is provided a method of reducing damage caused by an inhibition of the normal proliferation of epithelial cells of the bladder or prostate by administering to an individual an effective amount of KGF-2. As indicated, inhibitors of normal proliferation of bladder or prostate epithelium include radiation therapy (causing acute or chronic radiation damage) and cytoxic agents such as chemotherapeutic or antineoplastic drugs including, but not limited to, cyclophosphamide, busulfan, and ifosfamide. In a further aspect, KGF-2 is administered to reduce or prevent fibrosis and ulceration of the urinary bladder. Preferably, KGF-2 is administered to reduce or prevent hemorrhagic cystitis. Suitable doses, formulations, and administration routes are described below.
Stimulate the Proliferation of Nasal Mucosa Sinus infections usually occur in the setting of upper respiratory tract infections, allergies, or anatomic defects (within the sinuses or nasal septum) and may lead to symptoms of headache, facial pain, fever, and purulent rhinorrhea.
(Evens KL. Drugs 56(1): 59-71 (1998)). The symptoms of nasal allergy and chronic ethmoid sinusitis overlap and treatment failure in allergic patients may suggest possible chronic sinusitis. Chronic ethmoidal sinusitis may be the leading cause of rhinorrhea and nasal obstruction in patients with perennial allergies.
(Bertrand et al., Acta Otorhinolaryngol Belg 51 (4):227-237 (1997)). Chronic sinusitis is identified when symptoms of nasal obstruction, rhinorrhea, post nasal drip, intermittent facial pain (Evens KL. Drugs 56( 1 ): 59-71 ( 1998)) and anosmia (Jones et al., Int. J. Pediatr. Otorhinolaryngol. 28(1):25-32 (1993)) persist for 3 months or more. In addition to poor mucociliary transport and obstructed ostia, chronic sinusitis is also associated with mucosal edema, mucous hypersecretion, and persistent or recurrent infection.
Acute sinusitis tends to occur in patients with a history of rhinitis, which may be allergic or non-allergic in origin. Patients with anatomic variants (Evens KL. Drugs 56(1): 59-71 (1998)) and cystic fibrosis (Brihaye et al., Acta Otorhinolaryngol Belg 51(4):323-337 {1997); Ramsey et al., J. Allergy Clin.

Immunol. 90(3Pt2):547-552 (1992); Davidson et al., Laryngoscope 105(4Pt1):354-358 (1995); 3ones et al., Int. J. Pediatr. Otorhinolaryngol.
28(1):25-32 (1993)) are also at high risk for developing sinusitis. Acute inflammation and infection are associated with reduced mucociliary transport of secretions and obstruction of the ostia, which drain the sinuses. Initial medical treatment for acute sinusitis begins with analgesia to control pain; topical and systemic treatment of rhinitis, which may include antihistamines, mast cell stabilizers, and steroids; measures to clear secretions including steam inhalation and saline nasal douches; and when infection can be documented, antibiotics.
(Evans KL. Drugs 56( 1 ): 59-71 ( 1998)).
A general understanding of the pathophysiology of sinusitis is now available. In particular, it is known that occlusion of the sinus ostia starts the cycle of events, which lead to and sustain sinusitis. (Reilly J.S.
Otolaryngology Head and Neck Surgery 103(5):856-862( 1990)). The goals of treatment for both acute and chronic sinusitis are to control the rhinitis, improve ventilation to the sinuses, and to improve the function of the sinuses for clearance of secretions.
(Evans KL. Drugs 56(1): 59-71 (1998)). Nasal sinus surgery is performed only after there is evidence of persistent chronic sinusitis uncontrolled by medical treatment.
Surgical treatment is only considered when medical measures fail to control the sinusitis. The goals of treatment are identical: to improve ventilation and to facilitate or restore sinus drainage. An additional goal is that surgical treatment may sometimes improve the penetration of topical drugs into the sinus.
Up to 250,000 procedures are performed each year in the United States. The majority of the procedures performed use endoscopic methods (Stammberger H.
Otolaryngology Head and NeckSurgery 94(2):143-147 (1986); Stammberger H.
Otolaryngology Head and NeckSurgery 94(2):147-156 (1986)) with CT or x-ray tomographic assessment to identify specific lesions. The extent of surgery is kept to a minimum and is intended to remove only the primary anatomic cause that perpetuates the chronic sinusitis.

In tire course of perfonairlg sinus surgery, the mucosal surface is stritpped away or damaged. In cases where the surgery has been extensive, the underlying bone within the sinus may be exposed for periods of up to 6 months before the mucosa is fully reconstituted and recovery of ciliary density may require up to 2 S years to achieve. Although not proven, delayed re-epithelialization of the nasal sinuses following surgery is believed to be associated with increased risk of infection, scarnng (subepithelial fibrosis) leading to recurrent disease, and cyst formation. After 2 years of follow up approximately 2-5% of patients will have recurrent disease and up to 15% may require surgical revision. (Evans KL.
Drugs 56( 1 ): 59-71 ( 1998)) Other complications, which might be favorably impacted by accelerated re-epithelialization, include infectious complications (Evans KL.
Drugs 56( 1 ): 59-7I (1998)), failure to regain olfaction (Klossek et al., Otolaryngology Head and Neck Surgery 117(4):355-361 (1997)), recurrence of polyps (Klossek et al., Otolaryngology Head and Neck Surgery 117(4):355-361 (1997)) and requirement for additional post operative medical treatment (Sml'th et al., Otolaryngology Head and Neck Surgery 108(6):688-696 (1993)). Minor complications have been reported to occur in up to 8% of cases and approximately 40% of patients continued to require medical treatment for nasal congestion and discomfiture even following surgery. (Smith et al., Otolaryngology Head and Neck Surgery 108(6):688-696 (1993)).
Following endoscopic nasal surgery patients return at weekly intervals for repeat endoscopy and debridement. At each visit local anesthesia is applied, and the sinus cavity is cleaned, including removal of adhesions (which occur in 10-15% of cases) and mucus crusts. The duration of post op care is proportional to the extent of the surgery and the loss of the sinus epithelium. For simple cases 6 to 8 weeks of care may be needed. For complex or more extensive cases,12 to 18 weeks may be required. Accelerated healing of the sinus epithelium would reduce the duration of postoperative discomfiture to the patient and reduce the cost of care.
Administration of KGF-2 to an individual stimulates proliferation of the sinus epithelia and nasal mucosa. KGF-2 administration also results in goblet cell hyperplasia in the respiratory epithelium of the nasal air passage way. Thus, in one aspect, the present invention provides a method of stimulating the healing of mucosa of the sinus following damage by surgery or other pathological conditions by adminstration of KGF-2. Further, the present invention, KGF-2 is administered to stimulate the healing wounds located in the nasal and sinus mucosa. The present invention also provides for KGF-2 administration to improve function of the sinuses by increasing and/or protecting ciliary density and mucosal integrity. Another aspect of the invention is to provide a method decreasing sinus infection, scarring, polyps, cyst formation and recurrence as well as increasing olfaction by KGF-2 adminstration. Suitable doses, formulations, and administration routes are described below.
Increase Proliferation of Salivary and Lacrimal Glands Saliva is secreted from three major salivary glands: the parotid, sublingual and submandibular (or submaxillary} salivary glands. Tears are secreted from lacrimal glands. Loss of the ability to produce adequate amounts of saliva and tears is a major clinical problem affecting millions of people and there are few therapeutic options for these sufferers. Patients with xerostomia, or dry mouth, do not produce adequate amounts of saliva, have difficulty swallowing, have painful cracks in their mouths, and experience a decrease in their ability to taste.
This condition may be caused by Sjogren's syndrome, as a secondary event to radiation used with patients with head and neck tumors, and to drugs. Patients with Sjogren's syndrome sometimes have keratoconjunctivitis sicca or dry eye.
This condition may be caused by damage to the lacrimal gland due to Sjogren's syndrome, sarcoidosis, aging, HIV infection, burns, etc. (temp, M.A, Adv. Exp.
Med Biol. 438:791-803 ( 1998)}. Patients experience irritation, blurring of vision, burning, pain, and increased risk of infections. Millions of patients suffer with Sjogren's syndrome and therapy is sub-optimal. Stress is placed on reducing the risk of dental caries and the use of moistening agents. If there is remaining salivary tissue patients are treated with pilocarpine, which induces saliva production (Guchelaar, H.J. and Meerwaldt, V.A., Support Care Cancer 5:281-288 (1997); Garg, A.C. andMalo, M.,J. Am. Dent. Assoc.128:1128-1133 (1997);
Wiseman, L.R. and Faulds, D., Drugs 49:143-155 (1995); and LeVeque, F.G., et al., J. Clin. Oncol. 11:1124-1131 (1993)). A recent study has shown that treatment with IFN-a can reduce Iymphocytic infiltration in the salivary glands of patients with Sjogren's syndrome and improve function (Shiozawa, S., et al., J. Interferon Cytokine Res. 18:255-262 (1998)). However, such therapy, if approved, would impact only those patients whose salivary gland dysfunction is due to this autoimmune disease.
As is the case with xerostomia, patients with keratoconjunctivitis sicca have few therapeutic options. At this time there are approximately 10 million patients in the US which require artificial tear preparations (Letup, M.A, Adv.
Exp. Med Biol. 438:791-803 (1998)). There are no treatments available at this time to stimulate replacement of the cells in the salivary and lacrimal gland cells.
Two classifications of patients with "dry eye" are commonly used (Pflugfelder, S.C.,Adv. Dent. Res. 10 (1): 9-12 (1996)). One class being aqueous adequate dry eye. This clinical disorder is associated with meibomian gland dysfunction resulting in a deficiency in the superficial lipid layer of the tear film.
Drop out of meibomian glands can be visualized by transillumination biomicroscopy of the eyelids. The second class being aqueous deficient dry eye.
This clinical disorder may be immunologic (Sjogren's syndrome) or non-immunologic (non-Sjogren's syndrome}. Aqueous tear deficiencies lead to ocular surface disease, termed keratoconjunctivitis sicca, which results from either pathology in the Iacrimal gland or in mucin-producing epithelial cells.
Lower goblet cell densities have been reported in the conjunctiva) epithelium in patients with dry eye syndrome (Ralph, RA., Investigative Ophthalmology 14 (4): 299-302, (1975)).
Although dry eye syndromes have not been associated with deficiencies in specific growth factors, the lacrimal gland produces epidermal growth factor, which may play a role in ocular surface wound healing (Wilson, S.E., American Journal of Ophthalmology Ill (6): 763-765 (1991)). The presence of endogenous growth factors in lacrimal secretions provides the rationale for the use of topically applied keratinocyte growth factor to support the growth and differentiation of the conjunctival epithelium. In addition, atrophy or Iosses of secretory acini in the Iacrimal gland contribute to defective tear production.
KGF-2 effects on the proliferative rate of cells in the salivary and Iacrimal glands in normal rats were investigated in Example 24. Three major salivary glands exist in mammals: the parotid, sublingual and submandibular or submaxillary salivary glands. One intravenous administration of KGF-2 induced an almost forty-fold increase in the proliferation of serous cells in the parotid gland, an eighteen-fold elevation in the proliferation of serous and mucous cells in the submandibular gland, and a ten-fold increase in the proliferation of serous cells in the lacrimal gland. The enhanced proliferation in the salivary and lacrimal gland cells induced by KGF-2 was reversible.
KGF-2 produces a dramatic increase in the proliferation of normal serous and mucous secretory cells in salivary and Iacrimal glands ofnormai rats but does not appear to elicit a proliferative response of ductal cells. (See, Example 24).
This effect on the salivary and lacrimal glands reverses upon removal of treatment suggesting KGF-2 would prove safe and effective in promoting regeneration of the cells of these glands.
In addition to stimulating proliferation of the salivary glands, daily i.v.
injections of KGF-233 resulted in grossly visible thickening of the oral and esophageal mucosa in cynomolgus monkeys. (See, Example 36). The grossly visible thickening of the oral and esophageal mucosa correlated with oral and esophageal mucosal hyperkeratosis.
Since KGF-2033 has shown proliferative effects on secretory tissues such as salivary glands and the pancreas, the effect of systemic administration of KGF-2~33 on the lacrimal glands was also investigated in Example 25. The results demonstrates that the Iacrimal gland proliferates after 1, 2, and 3 daily i.v.

treatrnents with KGF-233. However, the gland fails to demonstrate elevated proliferation after 7 daily administrations ofthis gmwth factor, a situation that has been observed in many organs and tissues. (See, Example 25). These results indicate that topical or systemic administration of KGF-2A33 would be expected stimulate a therapeutic increase in the secretory capacity of the glands by its proliferative effect on lacrimal epithelial cells.
Accordingly, the invention provides for a method of stimulating the growth of the parotid, sublingual and submandibular (or submaxillary) salivary gland cells by administration of KGF-2 to a patient in need thereof. The invention also provides a method of stimulating the growth of lacrimal gland cells by administration of KGF-2 to a patient in need thereof. In particular, KGF-2 can be administered to treat or prevent keratoconjunctivitis sicca, xerostomia or other pathologies and injuries to the lacrimal gland, salivary gland or mucosa caused by radiation therapy, autoimmune disease, sarcoidosis, aging, HN
infection, burns, etc. Suitable doses, formulations, and administration routes are described below.
Stimulating Proliferation of Goblet Cells As indicated above, administration of KGF-2 to an individual also stimulates proliferation of Goblet cells in the respiratory epithelium of the nasal air passage way (Example 37). As demonstrated in Example 29, KGF-2 administration also results in proliferation of Goblet cells in the conjunctiva. The present inventors have further demonstrated that KGF-2 administration stimulates proliferation of Goblet cells in the large and small intestines. Thus, the present invention further provides a method for stimulating proliferation of Goblet cells by administration of KGF-2 to an individual in need thereof. Stimulating Goblet cell proliferation can be used for a number of purposes, including treating or preventing dry eye and radiation induced damage (such as during cancer therapies). Suitable doses, fonmuladons, and administration routes are described below.
Lung Epithelial Cell Prol feration KGF-2 stimulates proliferation of lung epithelial cells. Thus, KGF-2 can be administered prophylactically to reduce or prevent damage to the lungs caused by various pathological states. KGF-2 can also be administered during or after a damaging event occurs to promote healing. For example, KGF-2 can stimulate proliferation and differentiation and promote the repair of alveoli and bronchiolar epithelium to prevent, attenuate, or treat acute or chronic lung damage.
Emphysema, which results in the progressive loss of alveoli, and inhalation injuries, i. e. , resulting from smoke inhalation and burns, that cause necrosis ofthe bronchiolar epithelium and alveoli could be effectively treated using KGF-2 as could damage attributable to chemotherapy, radiation treatment, lung cancer, asthma, black lung and other lung damaging conditions. Also, KGF-2 could be used to stimulate the proliferation of and differentiation of type II
pneumocytes, which may help treat or prevent disease such as hyaline membrane diseases, such as infant respiratory distress syndrome and bronchopulmonary dysplasia, in premature infants.
KGF-2 stimulates proliferation of lung epithelial cells by direct intratracheal administration. Further, administration of nebulized KGF-2 also stimulates cell proliferation as does KGF-2 administered intraveneously.
Moreover, as demonstrated in Example 32, KGF-2 is useful prophylactically for lung fibrosis.
As used herein, by "individual" is intended an animal, preferably a mammal (such as apes, cows, horses, pigs, boars, sheep, rodents, goats, dogs, cats, chickens, monkeys, rabbits, ferrets, whales, and dolphins), and more preferably a human.

Pharmaceutical Compositions The KGF-2 polypeptides of the present invention may be employed in combination with a suitable pharmaceutical carrier to comprise a pharmaceutical composition. Such compositions comprise a therapeutically effective amount of the polypeptide, agonist or antagonist and a pharmaceutically acceptable carrier or excipient. Such a carrier includes but is not limited to saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. The formulation should suit the mode of administration.
The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Associated with such containers can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration. In addition, the polypeptides, agonists and antagonists of the present invention may be employed in conjunction with other therapeutic compounds.
The polypeptide having KGF-2 activity may be administered in pharmaceutical compositions in combination with one or more pharmaceutically acceptable excipients. It will be understood that, when administered to a human patient, the total daily usage of the pharmaceutical compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the type and degree of the response to be achieved; the specific composition an other agent, if any, employed; the age, body weight, general health, sex and diet of the patient;
the time of administration, route of administration, and rate of excretion of the composition; the duration of the treatment; drugs (such as a chemotherapeutic agent) used in combination or coincidental with the specific composition; and like factors well known in the medical arts. Suitable formulations, known in the art, can be found in Remington's Pharmaceutical Sciences (latest edition), Mack Publishing Company, Easton, PA.
The KGF-2 composition to be used in the therapy will be formulated and dosed in a fashion consistent with good medical practice, taking into account the clinical condition of the individual patient (especially the side effects of treatment with KGF-2 alone), the site of delivery of the KGF-2 composition, tl~e method of administration, the scheduling of administration, and other factors known to practitioners. The "effective amount" of KGF-2 for purposes herein is thus determined by such considerations.
The pharmaceutical compositions may be administered in a convenient manner such as by the oral, topical, intravenous, intraperitoneal, intramuscular, intraarticular, subcutaneous, intranasal, intratracheal, intraocular, inhalation, or intradermal routes. The pharmaceutical compositions are administered in an I5 amount which is effective for treating and/or prophylaxis of the specific indication. In most cases, the KGF-2 dosage is from about 1 ~,glkg to about 30 mg/kg body weight daily, taking into account the routes of administration, symptoms, etc. However, the dosage can be as low as 0.001 p.g/kg. For example, in the specific case of topical administration dosages are preferably administered from about 0.01 ~g to 9 mg per cm2. In the case of intranasal and intraocular administration, dosages are preferably administered from about 0.001 pg/ml to about 10 mg/ml, and more preferably from about 0.05 mg/ml to about 4 mg/ml.
As a general proposition, the total pharmaceutically effective amount of the KGF-2 administered parenterally per more preferably dose will be in the range of about 1 ~.g/kg/day to I00 mg/kg/day ofpatient body weight, although, as noted above, this will be subject to therapeutic discretion. If given continuously, the KGF-2 is typically administered at a dose rate of about 1 ~g/kg/hour to about ~,g/kg/hour, either by 1-4 injections per day or by continuous subcutaneous infusions, for example, using a mini-pump. An intravenous bag solution or bottle solution may also be employed.
More preferably, KGF-2 is administered intravenously at dose range of about 0.5 mg/kg to about 30 mg/kg to treat cystitis, hypoalbuminemia, hypofibrinogenimia, hypogammaglobulinemia, hemorrhagic cystitis, stimulating salivary gland epithelia, lung damage or injuries, stimulating sinus and nasal mucosa. KGF-2 is preferably nebulized and administered by inhalation at a dose range of about 6 mg to about 20 mg to treat pulmonary diseases and conditions as well as for nasal and sinus epithelium stimulation.
A course of KGF-2 treatment to affect the fibrinolytic system appears to be optimal if continued longer than a certain minimum number of days, 7 days in the case of the mice. The length of treatment needed to observe changes and the interval following treatment for responses to occur appears to vary depending on the desired effect. Such treatment lengths are indicated in the Examples below.
The KGF-2 polypeptide is also suitably administered by sustained-release systems. Suitable examples of sustained-release compositions include semi-permeable polymer matrices in the form of shaped articles, e.g., films, or mirocapsules. Sustained-release matrices include polylactides (U.S. Pat. No.
3,773,919, EP 58,481), copolymers of L-glutamic acid and gamma-ethyl-I,-glutamate (U. Sidman et al., Biopolymers 22:547-556 (1983)), poly (2-hydroxyethyl methacrylate) (R. Langer et al., J. Biomed Mater. Res. 15:167-277 (1981), and R. Langer, Chem. Tech.12:98-105 (1982)), ethylene vinyl acetate (R.
Langer et al., Id ) or poly-D- (-)-3-hydroxybutyric acid (EP 133,988).
Sustained-release KGF-2 compositions also include liposomally entrapped KGF-2.
Liposomes containing KGF-2 are prepared by methods known per se: DE
3,218,121; Epstein, et al., Proc. Natl. Acad. Sci. USA 82:3688-3692 (1985);
Hwang et a1, Proc. Natl. Acad. Sci. USA 77:4030-4034 (1980); EP 52,322; EP
36,676; EP 88,046; EP 143,949; EP 142,641; Japanese Pat. Appl. 83-118008;
U.S. Pat. Nos. 4,485,045 and 4,544,545; and EP 102,324. Ordinarily, the Iiposomes are of the small (about 200-800 Angstroms) unilamellar type in which the lipid content is greater than about 30 rnol. percent cholesterol, the selected proportion being adjusted for the optimal KGF-2 therapy.
For parenteral administration, in one embodiment, the KGF-2 is formulated generally by mixing it at the desired degree of purity, in a unit dosage injectable form (solution, suspension, or emulsion), with a pharmaceutically acceptable carrier, i.e., one that is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation. For example, the formulation preferably does not include oxidizing agents and other compounds that are known to be deleterious to polypeptides.
KGF-2 may also be administered to the eye to treat lacrimal gland injuries, disorders and pathologies in animals and humans as a drop, or within ointments, gels, liposomes, or biocompatible polymer discs, pellets or carried within contact lenses. The intraocular composition may also contain a physiologically compatible ophthalmic vehicle as those skilled in the art can select using conventional criteria. The vehicles may be selected from the known ophthalmic vehicles which include but are not limited to water, polyethers such s polyethylene glycol 400, polyvinyls such as polyvinyl alcohol, povidone, cellulose derivatives such as carboxymethylcellulose, methylcellulose and hydroxypropyl methylcellulose, petroleumn derivatives such as mineral oil and white petrolatum, animal fats such as lanolin, vegetable fats such as peanut oil, polymers of acrylic acid such as carboxylpolyrnethylene gel, polysaccharides such as dextrans and glycosaminoglycans such as sodium chloride and potassium, chloride, zinc chloride and buffer such as sodium bicarbonate or sodium lactate.
High molecular weight molecules can also be used. Physiologically compatible preservatives which do not inactivate the KGF-2 present in the composition include alcohols such as chlorobutanol, benzalknonium chloride and EDTA, or any other appropriate preservative known to those skilled in the art.
KGF-2 can also be intranasally administered to the nasal mucosa to treat disorders, injuries and pathologies of the nasal mucosa and sinus epithelia in animals and humans as drops or in a spray form. Various types of nasal delivery devices are well known in the art. In general, peptides are administered in an aqueous solution are often administered by means of metered-dose spray pumps.
For intranasal administration, suspensions will generally be aqueous solutions which contain a physiologically compatible vehicle as those skilled in the art can select using conventional criteria. For example, the suspension may be prepared from water alone (e.g., sterile or pyrogen-free water), water and a physioIogcially acceptable non aqueous vehicle (e.g., ethanol, propylene glycol, polyethylene glycols such as PEG 400, etc.). Such suspensions may additionally contain other excipients,, for ~ example, preservatives (e.g., benzalkonium chloride, phenylethylalchohol, and other known quaternary amines, etc.), wetting agents/surfactants (e.g., polysorbates such as Tween 80; sorbitan esters such as Span 80), buffering agents (e.g., acetic acid/sodium acetate, citric acid/disodium hydrogen phosphate, sodium dihydrogen phosphate/disodium hydrogen phosphate, and citric acid/sodium citrate, etc.), osmotic pressure controlling agents (e.g., sodium chloride), muscosal absorption enhancers (e.g., bile salts, monolauryl esters of macrogols, phospholipids and fusidate derivatives), and viscosity enhancers (e.g., acacia, bentonite, carboxymethylcellulose, gelatin, hydroxymethylcellulose, methylcellulose and the like).
KGF-2 intranasal administration may also be achieved by means of an aerosol formulation in which the peptide composition is provided in a pressurized pack with a suitable propellant such as a chlorofluorocarbon (CFC), a hydrofluorocarbon (HFC), carbon dioxide or other suitable gas. The aerosol may also contain a surfactant such as lecithin. The dose of the drug may be controlled by a metered valve device well known to those in the art.
Generally, the formulations are prepared by contacting the KGF-2 uniformly and intimately with liquid carriers or finely divided solid carriers or both. Then, if necessary, the product is shaped into the desired formulation.
Preferably the carrier is a parenteral carrier, more preferably a solution that is isotonic with the blood of the recipient. Examples of such carrier vehicles include water, saline, Ringer's solution, and dextrose solution. Non-aqueous vehicles such as fixed oils and ethyl oleate are also useful herein, as well as liposomes. Suitable formulations, known in the art, can be found in Remington's Pharmaceutical Sciences (latest edition), Mack Publishing Company, Easton, PA.
The carrier suitably contains minor amounts of additives such as substances that enhance isotonicity and chemical stability. Such materials are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, succinate, acetic acid, and other organic acids or their salts; antioxidants such as ascorbic acid; low molecular weight (less than about ten residues) polypeptides, e.g., polyarginine or tripeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids, such as glycine, glutamic acid, aspartic acid, or arginine; monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, glucose, mannose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; counterions such as sodium; andlor nonionic surfactants such as polysorbates, poloxamers, or PEG.
KGF-2 is typically formulated in such vehicles at a concentration of about 0.01 p,g/ml to 100 mg/ml, preferably 0.01 pg/ml to 10 mg/ml, at a pH of about to 8. It will be understood that the use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of KGF-2 salts.
KGF-2 to be used for therapeutic administration must be sterile. Sterility is readily accomplished by filtrationthrough sterile filtration membranes (e.g., 0.2 micron membranes). Therapeutic KGF-2 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.
KGF-2 ordinarily will be stored in unit or mufti-dose containers, for example, sealed ampules or vials, as an aqueous solution or as a lyophilized formulation for reconstitution. As an example of a lyophilized formulation, 10 ml vials are filled with 5 ml of sterile-filtered 1 % (w/v) aqueous KGF-2 solution, and the resulting mixture is lyophilized. The infusion solution is prepared by reconstituting the lyophilized KGF-2 using bacteriostatic Water-for-Injection.
Dosaging may also be arranged in a patient specific manner to provide a predetermined concentration of an KGF-2 activity in the blood, as determined by an RIA technique, for instance. Thus patient dosaging may be adjusted to achieve regular on-going trough blood levels, as measured by RIA, on the order of from 50 to 1000 ng/ml, preferably 150 to 500 ng/ml.
Pharmaceutical compositions of the invention may be administered orally, rectally, parenterally, intracisternally, intradermally, intravaginally, WO 99/41282 PCTNS99/0301$

intraperitoneally, topically (as by powders, ointments, gels, creams, drops or transdermal patch), bucally, or as an oral or nasal spray. By "pharmaceutically acceptable carrier" is meant a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. The term "parenteral" as used herein refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion.
Preferred KGF-2 formulations are described in U.S. Provisional Application No. 60/068,493, filed December 22, 1997, and the U.S. Non Provisional Application, filed on December 22, 1998 (Attorney Docket No.
1488.1030001), both of which are herein incorporated by reference.
Gene Therapy The KGF-2 polypeptide of the present invention may be employed by expression of the polypeptide in vivo, which is often referred to as "gene therapy-".
The gene therapy method relates to the introduction of nucleic acid (DNA, RNA
and antisense DNA or RNA) sequences encoding KGF-2 polypeptides into an animal to achieve expression of a KGF-2 polypeptide. Such gene therapy and delivery techniques are known in the art. See, for example, WO 90/ 11092, which is herein incorporated by reference.
As discussed more fully below, the KGF-2 polynucleotide sequences preferably have a therapeutic effect after being taken up by a cell. Examples of polynucleotides that are themselves therapeutic are anti-sense DNA and RNA;
DNA coding for an anti-sense RNA; or DNA coding for tRNA or rRNA to replace defective or deficient endogenous molecules. For example, a promoter may be operably linked to a DNA sequence encoding for an antisense RNA. The antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of an mRNA molecule into a polypeptide (Okano, J. Neurochem 56:560 ( 1991 )). The antisense RNA must be of sufl~cient length and complementarity to prevent translation of its target mRNA.

The heterologous polynucleotides can also code for therapeutic polypeptides. Therapeutic polypeptides include those that can compensate for defective or deficient polypeptide in an animal, or those that act through toxic effects to limit or remove harmful cells from the body. In the present invention, S the therapeutic polypeptide comprise KGF-2 polypeptides.
Thus, for example, cells from a patient may be engineered with a polynucleotide (DNA or RNA) comprising KGF-2 polynucleotide operably linked to a promoter ex vivo, with the engineered cells then being provided to a patient to be treated with the KGF-2 polypeptide. Such methods are well-known in the art. For example, see Belldegrun, A., et al., J. Natl. Cancer Inst. 85: 207-(1993); Ferrantini, M. etal., CancerResearch 53: 1107-I 112 (1993};
Fetrantini, M. et al., J. Immunology 153: 4604-4615 (1994); Kaido, T., et al., Int. J.
Cancer 60: 221-229 (1995); Ogura, H., et al., Cancer Research S0: 5102-5106 (1990);
Santodonato, L., et al., Human Gene Therapy 7:1-10 (1996); Santodonato, L., et al., Gene Therapy 4:1246-1255 (1997}; and Zhang, J.-F. et al., Cancer Gene Therapy 3: 31-38 (1996)). The cells to be engineered may be any cell type where KGF-2 may have a therapeutic effect. Such cells include, but are not limited to, muscle cells, epithelial cells, bladder, prostate, testes, lacrimal gland, salivary gland, sinus epithelia, conjunctiva, bone marrow stem and progenitor cells, lung, liver, pancreas, esophagus, etc. . . These engineered cells may be reintroduced into the patient through direct injection to the tissue of origin, the tissues surrounding the tissue of origin, veins or arteries; or through catheter injection.
Similarly, cells may be engineered in vivo for expression of a therapeutic polypeptide in vivo by, for example, procedures known in the art. The constructs can be delivered by any method that delivers materials to the cells of an animal, such as injection into the interstitial space of tissues. The conshvcts may be delivered in a pharmaceutically acceptable liquid or aqueous carrier well known to those skilled in the art.
In certain embodiments, the KGF-2 polynucleotide constructs may be delivered as naked polynucleotides. By "naked" polynucleotides is meant that the polynucleotides are free from any delivery vehicle that acts to assist, promote, or _87_ facilitate entry into the cell, including viral sequences, viral particles, liposome formulation, lipofectin, precipitating agents and the like. Such methods are well known in the art and described, for example, in U.S. Patent Nos. 5,593,972, 5,589,466, and 5,580,859.
The naked polynucleotides used in the invention can be those which do not integrate into the genome of the host cell. These may be non-replicating sequences, or specific replicating sequences genetically engineered to lack the genome-integration ability. Alternatively, the naked polynucleotides used in the invention may integrate into the genome of the host cell by, for example, homologous recombination, as discussed below. Preferably, the naked KGF-2 polynucleotide construct is contained in a plasmid. Suitable expression vectors for include, but are not limited to, vectors such as pRSVcat (ATCC 3 7152), pSVL
and MSG (Phanmacia, Uppsala, Sweden), pSV2dhfr (ATCC 37146) and pBC 12MI (ATCC 67109). Additional suitable plasmids are discussed in more detail above.
The naked polynucleotides can be administered to any tissue (such as muscle tissue) or organ, as described above. In another embodiment, the naked polynucleotides are adnunistered to the tissue surrounding the tissue of origin.
In another embodiment, the naked polynucleotides are administered systemically, through intravenous injection.
The naked polynucleotides are delivered by any method known in the art, including, but not limited to, direct needle injection at the delivery site, intravenous injection (especially portal vein injection), topical administration, catheter infusion, and so-called "gene guns". These delivery methods are known in the art and discussed in more detail below.
For naked polynucleotide injection, an effective dosage amount of polynucleotide will be in the range of from about 0.05 ~g/kg body weight to about 50 mg/kg body weight. Preferably, the dosage will be from about 0.005 mg/kg to about 20 mg/kg and more preferably from about 0.05 mg/kg to about 5 3 0 mg/kg. The appropriate and effective dosage of the polynucleotide construct can readily be determined by those of ordinary skill in the art and may depend on the condition being treated and the route of administration.
The constructs may also be delivered with delivery vehicles such as viral sequences, viral particles, liposome formulations, lipofectin, precipitating agents, etc. Such methods of delivery are known in the art. For example, the polynucleotide construct can be delivered specifically to hepatocytes through the method of Wu et al., J. Biol. Chem. 264:6985-16987 (1989).
In certain embodiments, the KGF-2 polynucleotide constructs are complexed in a liposome preparation. Liposomal preparations for use in the instant invention include cationic (positively charged), anionic (negatively charged) and neutral preparations. However, cationic liposomes are particularly preferred because a tight charge complex can be formed between the cationic liposome and the polyanionic nucleic acid. Cationic liposomes have been shown to mediate intracellular delivery of plasmid DNA (Felgner et al., Proc. Natl.
Acad. Sci. USA (1987) 84:7413-7416); mRNA (Malone et al., Proc. Natl. Acad.
Sci. USA ( 1989) 86:6077-6081 ); and purified transcription factors (Debs et al., J. Biod. Chem. (1990) 265:10189-10192), in functional form.
Cationic liposomes are readily available. For example, N[l-2,3-dioleyloxy)propylJ-N,N,N-triethylammonium (DOTMA) liposomes are particularly useful and are available under the trademark Lipofectin, from GIBCO
BRL, Grand Island, N.Y. (See, also, Felgner et al., Proc. Natl Acad. Sci. USA
(1987) 84:7413-7416). Other commercially available liposomes include transfectace (DDAB/DOPE) and DOTAP/DOPE (Boehringer).
Other cationic liposomes can be prepared from readily available materials using techniques well known in the art. See, e.g. PCT Publication No. WO
90/11092 for a description of the synthesis of DOTAP (1,2-bis(oleoyloxy)-3 (trimethylammonio)propane) liposomes. Preparation of DOTMA liposomes is explained in the literature, see, e.g., P. Felgner et al., Proc. Natl. Acad.
Sci. USA
84:7413-7417. Similar methods can be used to prepare liposomes from other cationic lipid materials.
Similarly, anionic and neutral liposomes are readily available, such as from Avanti Polar Lipids (Birmingham, Ala.), or can be easily prepared using readily available materials. Such materials include phosphatidyl, choline, cholesterol, phosphatidyl ethanolamine, dioleoylphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), dioleoylphoshatidyl ethanolamine (DOPE), among others. These materials can also be mixed with the DOTMA ~d DOTAP starting materials in appropriate ratios. Methods for making liposomes using these materials are well known in the art.
For example, commercially dioleoylphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), and dioleoylphosphatidyl ethanolamine (DOPE) can be used in various combinations to make conventional liposomes, with or without the addition of cholesterol. Thus, for example, DOPG/DOPC
vesicles can be prepared by drying 50 mg each of DOPG and DOPC under a stream of nitrogen gas into a sonication vial. The sample is placed under a vacuum pump overnight and is hydrated the following day with deionized water.
The sample is then sonicated for 2 hours in a capped vial, using a Heat Systems model 350 sonicator equipped with an inverted cup (bath type) probe at the maximum setting while the bath is circulated at 15 °C. Alternatively, negatively charged vesicles can be prepared without sonication to produce multilamellar vesicles or by extrusion through nucleopore membranes to produce unilamellar vesicles of discrete size. Other methods are known and available to those of skill in the art.
The liposomes can comprise multilamellar vesicles (MLVs), small unilamellar vesicles (SUVs), or large unilamellar vesicles (LUVs), with SUVs being preferred. The various liposome-nucleic acid complexes are prepared using methods well known in the art. See, e.g., Straubinger et al., Methods of Immunology (1983),101:512-527. For example, MLVs containing nucleic acid can be prepared by depositing a thin film of phospholipid on the walls of a glass tube and subsequently hydrating with a solution of the material to be encapsulated. SUVs are prepared by extended sonication of MLVs to produce a homogeneous population of unilamellar liposomes. The material to be entrapped is added to a suspension of preformed MLVs and then sonicated. When using liposomes containing cationic lipids, the dried lipid film is resuspended in an appropriate solution such as sterile water or an isotonic buffer solution such as 10 mM Tris/NaCI, sonicated, and then the preformed liposomes are mixed directly with the DNA. The liposome and DNA form a very stable complex due to binding of the positively charged liposomes to the cationic DNA. SUVs find use with small nucleic acid fragments. LUVs are prepared by a number of methods, well known in the art. Commonly used methods include Caz+-EDTA chelation (Papahadjopoulos et al., Biochim. Biophys. Acta (1975) 394:483; Wilson et al., Cell (1979) 17:77); ether injection (Deamer, D. and Bangham, A., Biochim.
Biophys. Acta (1976) 443:629; Ostro et al., Biochem. Biophys. Res. Commun.
(1977) 76:836; Fraley et al., Proc. Natl. Acad. Sci. USA (1979) 76:3348);
detergent dialysis (Epoch, H. and Strittmatter, P., Proc. Natl. Acad. Sci. USA
(1979) 76:145); and reverse-phase evaporation (REV) (Fraley et al., J. Biol.
Chem. (1980) 255:10431; Szoka, F. and Papahadjopoulos, D., Proc. Natl. Acad.
Sci. USA (1978) 75:145; Schaefer-Ridder et al., Science (1982) 215:166).
Additional examples of useful cationic lipids include dipalmitoyl-phophatidylethanolamine 5-carboxyspen-nylamide (DPPES); 5-carboxyspermylglycine dioctadecylamide (DOGS); dimethyldioctdecyl-ammonium bromide (DDAB); and (f)-N,N-dimethyl-N-[2-(sperminecarboxamido)ethyl]-2,3-bis(dioleyloxy)-1-propaniminium pentahydrochloride (DOSPA). Non-diether cationic lipids, such as DL-1,2-dioleoyl-3-dimethylaminopropyl-[3-hydroxyethylammonium (DORI diester),1,2-O-dioleyl-3-dimethylaminopropyl-(3-hydroxyethylammonium (DORIE diether), 1-O-oleyl-2-oleoyl-3-dimethylaminopropyl-[i-hydroxyethylammonium (DORI
ester/ether), and their salts promote in vivo gene delivery. Cationic cholesterol derivativessuchas, {3~i[N N',N'-dimethylamino~thane]-carbomoyl}-cholesterol . (DC-Chol), are also useful.
Preferred cationic lipids include: (t)-N-(2-hydmxyethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propaniminium bromide; 3,5-(N,N-di-lysyl)diamino-benzoylglycyl-3-(DL-1,2-dioleoyl-dimethylaminopropyl-~3-hydroxyethylamine) (DLYS-DABA-GLY-DORI diester); 3,5-(NN-dilysyl)-diaminobenzoyl-3-(DL-1,2-dioleoyl-dimethylaminopropyl-~3-hydroxyethylamine) (DLYS-DABA-DORI

WO 99/41282 PC'T/US99/03018 diester); and 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine. Also preferred is the combinations of the following lipids: (t)-N-(2-hydroxyethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1 propaniminium bromide and 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine; and (f)-N-(2-hydroxyethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propaniminium bromide, and 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine in a 1:1 ratio.
The lipid formulations may have a cationic lipid alone, or also include a neutral lipid such as cardiolipin, phosphatidylcholine, phosphatidylethanolamine, dioleoylphosphatylcholine, dioleoylphosphatidyl-ethanolamine,1,2-dioleoyl-sn-glycero-3-phosphatidylethanolamine (DOPE), sphingomyelin, and mono-, di- or tri-acylglycerol).
Lipid formulations may also have cationic lipid together with a lysophosphatide. The lysophosphatide may have a neutral or a negative head group. Useful lysophosphatides include lysophosphatidylcholine, lysophosphatidyl-ethanolamine, and 1-oleoyl lysophosphatidylcholine.
Lysophosphatide lipids are present Other additives, such as cholesterol, fatty acid, ganglioside, glycolipid, neobee, niosome, prostaglandin, sphingolipid, and any other natural or synthetic amphiphiles, can be used. A preferred molar ratio of cationic lipid to neutral lipid in these lipid formulations is from about 9:1 to about 1:9; an equimolar ratio is more preferred in the lipid-containing formulation in a 1:2 ratio of lysolipid to cationic lipid.
Generally, the ratio of DNA to liposomes will be from about 10:1 to about 1:10. Preferably, the ratio will be from about 5:1 to about 1:5. More preferably, the ratio will be about 3:1 to about 1:3. Still more preferably, the ratio will be about 1:1.
U.S. Patent No. 5,676,954 reports on the injection of genetic material, complexed with cationic liposomes carriers, into mice. U.S. Patent Nos.
4,897,355, 4,946,787, 5,049,386, 5,459,127, 5,589,466, 5,693,622, 5,580,859, 5,703,055, and international publication no. WO 94/9469 provide cationic lipids for use in transfecting DNA into cells and mammals. U.S. Patent Nos.
5,589,466, 5,693,622, 5,580,859, 5,703,055, and international publication no. WO 94/9469 provide methods for delivering DNA-cationic lipid complexes to mammals.
In certain embodiments, cells are be engineered, ex vivo or in vivo, using a retroviral particle containing RNA which comprises a polynucleotide encoding KGF-2 operably linked to a promoter. Retmviruses from which the retroviral plasmid vectors may be derived include, but are not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus, Rous sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, Myeloproliferative Sarcoma Virus, and mammary tumor virus.
The retroviral plasmid vector is employed to transduce packaging cell lines to form producer cell lines. Examples of packaging cells which may be transfected include, but are not limited to, the PE501, PA317, tar-2, ~r-AM, PA 12, T19-14X, VT-19-17-H2, ~rCRE, ~rCRIP, GP+E-g6, GP+envAml2, and DAN cell lines as described in Miller, Human Gene Therapy 1:5-14 (1990), which is incorporated herein by reference in its entirety. The vector may transduce the packaging cells through any means known in the art. Such means include, but are not limited to, electroporation, the use of liposomes, and CaP04 precipitation. In one alternative, the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid, and then administered to a host.
The producer cell line generates infectious retroviral vector particles which include a KGF-2 polynucleotide operably linked to a promoter. Such retroviral vector particles then may be employed, to transduce eukaryotic cells, either in vitro or in vivo. The transduced eukaryotic cells will express the desired polypeptide.
In certain other embodiments, cells are engineered, ex vivo or in vivo, with the KGF-2 polynucleotide operably linked to a promoter contained in an adenovirus vector. Adenovirus can be manipulated such that it encodes and expresses the desired gene pmduct, and at the same time is inactivated in terms of its ability to replicate in a normal lytic viral life cycle. Adenovirus expression is achieved without integration of the viral DNA into the host cell chromosome, thereby alleviating concerns about insertional mutagenesis. Furthermore, adenoviruses have been used as live enteric vaccines for many years with an excellent safety profile (Schwartz, A. R. et al. (1974) Am. Rev. Respir.
Dis.109:233-238). Finally, adenovirus mediated gene transfer has been demonstrated in a number of instances including transfer of alpha-1-antitrypsin and CFTR to the lungs of cotton rats (Rosenfeld, M. A. et al. ( 1991 ) Science 252:431-434; Rosenfeld et al., (1992) Cell 68:143-155). Furthermore, extensive studies to attempt to establish adenovirus as a causative agent in human cancer were uniformly negative (Green, M. et al. (1979} Proc. Natl. Acad. Sci. USA
76:6606).
Suitable adenoviral vectors useful in the present invention are described, for example, in Kozarsky and Wilson, Curr. Opin. Genet. Devel. 3:499-503 (1993); Rosenfeld et al., Cell 68:143-155 (1992); Engelhardt et al., Human Genet. Ther. 4:759-769 (1993); Yang et al., Nature Genet. 7:362-369 (1994);
Wilsonetal.,Nature 365:691-692 (1993); and U.S. PatentNo. 5,652,224, which are herein incorporated by reference. For example, the adenovirus vector Ad2 is useful and can be grown in human 293 cells. These cells contain the E 1 region of adenovirus and constitutively express EIa and Elb, which complement tTie defective adenoviruses by providing the products of the genes deleted from the vector. In addition to Ad2, other varieties of adenovirus (e.g., Ad3, AdS, and Ad7) are also useful in the present invention.
Preferably, the adenoviruses used in the present invention are replication deficient. Replication deficient adenoviruses require the aid of a helper virus andlor packaging cell line to form infectious particles. The resulting virus is capable of infecting cells and can express a polynucleotide of interest which is operably linked to a promoter, for example, the KGF-2 polynucleotide of the present invention, but cannot replicate in most cells. Replication deficient adenoviruses may be deleted in one or more of all or a portion of the following genes: E 1 a, E 1 b, E3, E4, E2a, or L 1 through L5.
In certain other embodiments, the cells are engineered, ex vivo or in vivo, using an adeno-associated virus (AAV). AAVs are naturally occurring defective viruses that require helper viruses to produce infectious particles (Muzyczka, N., Curr. Topics in Microbiol. Immunol. 158:97 (1992)). It is also one of the few viruses that may integrate its DNA into non-dividing cells. Vectors containing as little as 300 base pairs of AAV can be packaged and can integrate, but space for exogenous DNA is limited to about 4.5 kb. Methods for producing and using such AAVs are known in the art. See, for example, U.S. Patent Nos. 5,139,941, 5,173,414, 5,354,678, 5,436,146, 5,474,935, 5,478,745, and 5,589,377.
For example, an appropriate AAV vector for use in the present invention will include all the sequences necessary for DNA replication, encapsidation, and host-cell integration.. The KGF-2 polynucleotide construct is inserted into the AAV vector using standard cloning methods, such as those found in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press (1989). The recombinant AAV vector is then transfected into packaging cells which are infected with a helper virus, using any standard technique, including lipofection, electroporation, calcium phosphate precipitation, etc.
Appropriate helper viruses include adenoviruses, cytomegaloviruses, vaccinia viruses, or herpes viruses. Once the packaging cells are transfected and infected, they will produce infectious AAV viral particles which contain the KGF-2 polynucleotide construct. These viral particles are then used to transduce eukaryotic cells, either ex vivo or in vivo. The tnuisduced cells will contain the KGF-2 polynucleotide construct integrated into its genome, and will express the molecule of interest.
Another method of gene therapy involves operably associating heterologous control regions (e.g., a promoter of interest) and endogenous polynucleotide sequences (e.g., KGF-2) via homologous recombination (see, e.g., U.S. Patent No. 5,641,670, issued June 24, 1997; International Publication No.
WO 96/29411, published September 26,1996; International Publication No. WO
94/12650, published August 4, 1994; Koller et al., Proc. Natl. Acad. Sci. USA
86:8932-8935 (1989); and Zijlstra et al., Nature 342:435-438 (1989). This method involves the activation of a gene which is present in the target cells, but which is not normally expressed in the cells, or is expressed at a lower level than desired.
Polynucleotide constructs are made, using standard techniques known in the art, which contain the promoter of interest with targeting sequences flanking the promoter of interest. The targeting sequence is sufficiently complementary to an endogenous sequence to permit homologous recombination of the promoter-targeting sequence with the endogenous sequence. The targeting sequence will be sufficiently near the 5' end of the desired endogenous polynucleotide sequence so the promoter will be operably linked to the endogenous sequence upon homologous recombination. The promoter and the targeting sequences can be amplified using PCR. Preferably, the amplified promoter contains distinct restriction enzyme sites on the 5' and 3' ends. Preferably, the 3' end of the first targeting sequence contains the same restriction enzyme site as the 5' end of the amplified promoter and the 5' end of the second targeting sequence contains the same restriction site as the 3' end of the amplified promoter. The amplified promoter and targeting sequences are digested and ligated together.
The promoter-targeting sequence construct is delivered to the cells, either as naked polynucleotide, or in conjunction with transfection-facilitating agents, such as liposomes, viral sequences, viral particles, whole viruses, lipofection, precipitating agents, etc., described in more detail above. The promoter-targeting sequence can be delivered by any method, included direct needle injection, intravenous injection, topical administration, catheter infusion, particle accelerators, etc. The methods are described in more detail below.
The promoter-targeting sequence construct is taken up by cells.
Homologous recombination between the construct and the endogenous sequence takes place, such that an endogenous sequence (e.g., KGF-2) is placed under the control of the promoter. The promoter then drives the expression of the endogenous sequence (e.g., KGF-2).
KGF-2 polynucleotides of the present invention may be used in gene therapy to treat metabolic, infectious and other diseases described herein.
Preferably, a KGF-2 polynucleotide of the present invention is operably linked a promoter so as to alleviate the symptoms of, or cures the disease to be treated as described in detail herein.
Any mode of administration of any of the above-described polynucleotides constructs can be used so long as the mode results in the expression of one or more molecules in an amount sufficient to provide a therapeutic effect. This includes direct needle injection, systemic injection, ;.catheter infusion, biolistic injectors, particle accelerators (i.e., "gene guns"), gelfoam sponge depots, other commercially available depot materials, osmotic PCPs (e~g~~ ~ ~P~ps), oral or suppositorial solid (tablet or pill) pharmaceutical formulations, and decanting or topical applications during surgery. For example, direct injection of naked calcium phosphate-precipitated plasmid into rat liver and rat spleen or a protein-coated plasmid into the portal vein has resulted in gene expression of the foreign gene in the rat livers (Kaneda et al. , Science 243: 3 75 ( 1989)).
A preferred method of local administration is by direct injection.
Preferably, a recombinant molecule of the present invention complexed with a delivery vehicle is administered by direct injection into or locally within the area of the liver. Administration of a composition locally within the area of the liver refers to injecting the composition centimeters and preferably, millimeters within the liver.
Another method of local administration is to contact a KGF-2 polynucleotide-promoter construct of the present invention in or amend a surgical wound. For example, a patient can undergo surgery and the polynucleotide construct can be coated on the surface of tissue inside the wound or the construct can be injected into areas of tissue inside the wound.
Therapeutic compositions useful in systemic administration, include recombinant molecules of the present invention complexed to a targeted delivery vehicle of the present invention. Suitable delivery vehicles for use with systemic administration comprise liposomes comprising ligands for targeting the vehicle to a particular site, for example, ligands for targeting the vehicle to a tissue of interest. Targeting vehicles for other tissues and organs are well known to skilled artisans.
Preferred methods of systemic administration, include. intravenous injection, aerosol, oral and percutaneous (topical) delivery. Intravenous injections can be performed using methods standard in the art. Aerosol delivery can also be performed using methods standard in the art (see, for example, Stribling et al., Proc. Natl. Acad. Sci. USA 189:11277-11281,1992, which is incorporated herein by reference). Oral delivery can be performed by complexing a polynucleotide construct ofthe present invention to a carrier capable of withstanding degradation by digestive enzymes in the gut of an animal. Examples of such carriers, include plastic capsules or tablets, such as those known in the art. Topical delivery can be performed by mixing a polynucleotide construct of the present invention with a lipophilic reagent (e.g., DMSO) that is capable of passing into the skin.
Detennining an effective amount of substance to be delivered can depend upon a number of factors including, for example, the chemical structure and biological activity of the substance, the age and weight of the animal, the precise condition requiring treatment and its severity, and the route of administration.
The frequency of treatments depends upon a number of factors, such as the amount of polynucleotide constructs administered per dose, as well as the health and history of the subject. The precise amount, number of doses, and timing of doses will be determined by the attending physician or veterinarian Therapeutic compositions of the present invention can be administered to any animal, preferably to mammals and birds. Preferred mammals include humans, dogs, cats, mice, rats, rabbits sheep, cattle, horses and pigs, with humans being particularly preferred.
Example 1 Bacterial Expression and Purification of KGF 2 The DNA sequence encoding KGF-2, ATCC # 75977, is initially amplified using PCR oligonucleotide primers corresponding to the 5' and 3' end sequences of the processed KGF-2 cDNA (including the signal peptide sequence).
The 5' oligonucleotide primer has the sequence 5' CCCCACATGTGGAAATGGATACTGACACATTGTGCC 3' (SEQ ID
No. 3) contains an Afl III restriction enzyme site including and followed by nucleotides of KGF-2 coding sequence starting from the presumed initiation colon. The 3' sequence 5' CCCAAGCTTCCACAAACGTTGCCTTCCTCTATGAG 3' (SEQ ID No. 4) contains complementary sequences to Hind III site and is followed by 26 nucleotides of KGF-2. The restriction enzyme sites are compatible with the restriction enzyme sites on the bacterial expression vector pQE-60 (Qiagen, Inc.
Chatsworth, CA). pQE-60 encodes antibiotic resistance (Amps , a bacterial origin of replication (ori), an IPTG-regulatable promoter operator (P/0), a ribosome binding site (RBS) , a 6-His tag and restriction enzyme sites. pQE-b0 is then digested with NcoI and HindIII. The amplified sequences are ligated into pQE-60 and are inserted in fiame. The ligation mixture is then used to transform E. coli strain M1 S/rep 4 (Qiagen, Inc.) by the procedure described in Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, Cold Spring Laboratory Press, (1989). M15/rep4 contains multiple copies ofthe plasmid plZEP4, which expresses the lacI repressor and also confers kanamycin resistance (Kan~ .
Transformants are identified by their ability to grow on LB plates and ampicillin/kanamycin resistant colonies are selected. Plasmid DNA is isolated and confirmed by restriction analysis. Clones containing the desired constructs are grown overnight (O/N) in liquid culture in LB media supplemented with both Amp (100 ug/ml) and Kan (25 ug/ml) . The O/N culture is used to inoculate a large culture at a ratio of 1:100 to 1:250. The cells are grown to an optical density 600 (O.D.~ of between 0.4 and 0.6. IPTG ("Isopropyl-B-D-thiogalacto pyranoside") is then added to a final concentration of 1 mM. IPTG interacts with the lacI repressor to cause it to dissociate from the operator, forcing the promoter to direct transcription. Cells are grown an extra 3 to 4 hours. Cells are then harvested by centrifugation. The cell pellet is solubilized in the chaotropic agent 6 Molar Guanidnine HCI. After clarification, solubilized KGF-2 is purified from this solution by chromatography on a Heparin affinity column under conditions that allow for tight binding of the proteins (Hochuli, E., et aL, J.
Chromatography 411:177-184 (1984)). KGF-2 (75% pure) is eluted from the column by high salt buffer.

Example 2 Bacterial Expression and Purification of a Truncated Version of KGF 2 The DNA sequence encoding KGF-2, ATCC # 75977, is initially amplified using PCR oligonucleotide primers corresponding to the 5' and 3' sequences of the truncated version of the KGF-2 polypeptide. The truncated version comprises the polypeptide minus the 36 amino acid signal sequence, with a methionine and alanine residue being added just before the cysteine residue which comprises amino acid 37 of the full-length protein. The 5' oligonucleotide primer has the sequence 5' CATGCCATGGCGTGCCAAGCCCTTGGTCAGGACATG 3' (SEQ ID
No. 5) contains an NcoI restriction enzyme site including and followed by 24 nucleotides of KGF-2 coding sequence. The 3' sequence 5' CCCAAGCTTCCACAAACGTTGCCTTCCTC TATGAG 3' (SEQ ID No. 6) contains complementary sequences to Hind III site and is followed by 26 nucleotides of the KGF-2 gene. The restriction enzyme sites are compatible with the restriction enzyme sites on the bacterial expression vector pQE-60 (Qiagen, Inc. Chatsworth, CA). pQE-60 encodes antibiotic resistance (Amps , a bacterial origin of replication (ori), an IPTG-regulatable promoter operator (P/O) , a ribosome binding site (RBS) , a 6-His tag and restriction enzyme sites. pQE-60 is then digested with NcoI and HindIII. The amplified sequences are ligated into pQE-60 and are inserted in frame. The ligation mixture is then used to transform E. coli strain M15/rep 4 (Qiagen, Inc.) by the procedure described in Sambrook, J., et al., Molecular Cloning: A Laboratory Manual, Cold Spring Laboratory Press, (1989). Ml5/rep4 contains multiple copies of the plasmid pREP4, which expresses the lacI repressor and also confers kanamycin resistance (Kan~.
Transformants are identified by their ability to gmw on LB plates and ampicillin/kanamycin resistant colonies are selected. Plasmid DNA is isolated and confirmed by restriction analysis. Clones containing the desired constructs are grown overnight (O/N) in liquid culture in LB media supplemented with both Amp (100 ug/ml) and Kan (25 ug/ml). The O/N culture is used to inoculate a large culture at a ratio of 1:100 to 1:250. The cells are grown to an optical density 600 (O.D.6°°) of between 0.4 and 0.6. IPTG ("Isopropyl-B-D-thiogalacto pyranoside") is then added to a final concentration of 1 mM. IPTG induces by inactivating the laci repressor, clearing the P/O leading to increased gene expression. Cells are grown an extra 3 to 4 hours. Cells are then harvested by centrifugation. The cell pellet is solubilized in the chaotropic agent 6 Molar Guanidine HCI. After clarification, solubilized KGF-2 is purified from this solution by chromatography on a Heparin unity column under conditions that IO allow for tight binding the proteins (Hochuli, E. et al., J. Chromatography 411:177-184 (1984)) . KGF-2 protein is eluted from the column by high salt buffer.
Example 3 Cloning and Expression of KGF 2 Using the Baculovirus Expression System The DNA sequence encoding the full length KGF-2 protein, ATCC #
75977, is amplified using PCR oligonucleotide primers corresponding to the 5' and 3' sequences of the gene:
The 5' primer has the sequence 5' GCGGGATCCGCCATCA TGTGGAAATGGATACTCAC 3' (SEQ ID No.
7) and contains a BamHI restriction enzyme site (in bold) followed by 6 nucleotides resembling an efficient signal for the initiation of translation in eukaryotic cells (Kozak, M., J. Mol. Biol., 196:947-950 (1987)) and just behind the first 17 nucleotides of the KGF-2 gene (the initiation codon for translation "ATG" is underlined).
The 3' primer has the sequence 5' GCGCGGTACCACAAACGTTGCCTTCCT 3' (SEQ ID No. 8) and contains the cleavage site for the restriction endonuciease Asp718 and 19 nucleotides complementary to the 3' non-translated sequence of the KGF-2 gene. The amplified sequences are isolated from a 1 % agarose gel using a commercially available kit from Qiagen, Inc., Chatsworth, CA. The fragment is then digested with the endonucleases BamHI and Asp718 and then purified again on a 1 agarose gel. This fragment is designated F2.
The vector pA2 (modification ofpVL941 vector, discussed below) is used for the expression of the KGF-2 protein using the baculovirus expression system (for review see: Summers, M.D. & Smith, G.E., A manual of methods for baculovirus vectors and insect cell culture procedures, Texas Agricultural Experimental Station Bulletin No.1555 ( 1987)). This expression vector contains the strong polyhedrin promoter of the Autographa californica nuclear polyhidrosis virus (AcMNPV) followed by the recognition sites for the restriction endonucleases BamHI and Asp718. The polyadenylation site of the simian virus (SV) 40 is used for efficient polyadenylation. For an easy selection of recombinant vintses the beta-galactosidase gene from E. coli is inserted in the same orientation as the polyhedrin promoter followed by the polyadenylation signal ofthe polyhedrin gene. The polyhedrin sequences are flanked at both saes by viral sequences for the cell-mediated homologous recombination of co transfected wild-type viral DNA. Many other baculovirus vectors could be used such as pAc373, pVL941 and pAcIMI (Luckow, V.A. & Summers, M.D., Virology, 170:31-39).
The plasmid is digested with the restriction enzymes BamHI and Asp718.
The DNA is then isolated from a 1 % agarose gel using the commercially available kit (Qiagen, Inc., Chatsworth, CA). This vector DNA is designated V2.
Fragment F2 and the plasmid V2 are ligated with T4 DNA ligase. E. coli I-iB 1 O1 cells are then transformed and bacteria identified that contained the plasmid (pBacKGF-2) with the KGF-2 gene using PCR with both cloning oligonucleotides. The sequence of the cloned fragment is confirmed by DNA
sequencing.
5 pg of the plasmid pBacKGF-2 is co-transfected with 1.0 pg of a commercially available linearized baculovirus ("BaculoGoldTM baculovirus DNA", Phanningen, San Diego, CA) using the lipofection method (Felgner, et al., Proc. Natl. Acad. Sci. USA, 84:7413-7417 (1987)).
1 ~g of BaculoGoldTM virus DNA and 5 pg of the plasmid pBacKGF-2 are mixed in a sterile well of a microtiter plate containing 50 ~tl of serum free Grace's medium (Life Technologies Inc., Gaithersburg, MD). Afterwards 10 ~1 Lipofectin plus 90 pl Grace's medium are added, mixed and incubated for 15 minutes at room temperature. Then the transfection mixture is added drop-wise to the SP9 insect cells (ATCC CRL 1711 ) seeded in a 35 mm tissue culture plate with 1 ml Grace's medium without serum. The plate is rocked back and forth to mix the newly added solution. The plate is then incubated for 5 hours at 27°C.
After 5 hours the transfection solution is removed from the plate and I ml of Grace's insect medium supplemented with 10% fetal calf serum is added. The plate is put back into an incubator and cultivation continued at 27°C
for four days.
After four days the supernatant is collected and a plaque assay performed similar as described by Summers and Smith (supra). As a modification an agarose gel with "Blue Gal" (Life Technologies Inc., Gaithersburg) is used which allows an easy isolation of blue stained plaques. (A detailed description of a "plaque assay" can also be found in the user's guide for insect cell culture and baculovimlogy distributed by Life Technologies Inc., Gaithersburg, page 9-10).
Four days after the serial dilution, the viruses are added to the cells and blue stained plaques are picked with the tip of an Eppendorf pipette. The agar containing the recombinant viruses is then resuspended in an Eppendorf tube containing 200 pl of Grace's medium. The agar is removed by a brief centrifugation and the supernatant containing the recombinant baculovirus is used to infect Sf 9 cells seeded in 35 mm dishes. Four days later the supernatants of these culture dishes are harvested and then stored at 4 ° C.
S~ cells are grown in Grace's medium supplemented with 10% heat-inactivated FBS. The cells are infected with the recombinant baculovirus V-KGF-2 at a multiplicity of infection (MOI) of 2. Six hours later the medium is removed and replaced with SF900 II medium minus methionine and cysteine (Life Technologies Inc., Gaithersburg). 42 hours later S ~,Ci of 3sS
methionine and 5 pCi 3sS cysteine (Amersham) are added. The cells are further incubated for 16 hours before they are harvested by centrifugation and the labelled proteins visualized by SDS-PAGE and autoradiography.
Example 4 Most of the vectors used for the transient expression of the KGF-2 protein gene sequence in mammalian cells should carry the SV40 origin of replication.
This allows the replication of the vector to high copy numbers in cells (e.g., COS
cells) which express the T antigen required for the initiation of viral DNA
synthesis. Any other mammalian cell line can also be utilized for this purpose.
A typical mammalian expression vector contains the promoter element, which mediates the initiation of transcription of mRNA, the protein coding sequence, and signals required for the termination of transcription and polyadenylation ofthe transcript. Additional elements include enhancers, Kozak sequences and intervening sequences flanked by donor and acceptor sites for RNA splicing. Highly efficient transcription can be achieved with the early and late promoters from SV40, the long terminal repeats (LTRs) from Retroviruses, e.g., RSV, HTLVI, HIVI and the immediate early promoter of the cytomegalovirus (CMV). However, cellular signals can also be used (e.g., human actin promoter). Suitable expression vectors for use in practicing the present invention include, for example, vectors such as pSVL and pMSG (Pharmacia, Uppsala, Sweden), pRSVcat (ATCC 37152), pSV2dhfr (ATCC 37146) and pBC 12MI (ATCC 67109). Mammalian host cells that could be used include, human Hela, 283, H9 and 3urkart cells, mouse NIH3T3 and C127 cells, Cos 1, Cos 7 and CV1, African green monkey cells, quail QCI-3 cells, 293T cells, mouse L cells and Chinese hamster ovary cells.
Alternatively, the gene can be expressed in stable cell lines that contain the gene integrated into a chromosome. The co-transfection with a selectable marker such as dhfr, gpt, neomycin, hygromycin allows the identification and isolation of the transfected cells.

The transfected gene can also be amplified to express large amounts of the encoded protein. The DHFR (dihydrofolate reductase) is a useful marker to develop cell lines that carry several hundred or even several thousand copies of the gene of interest. Another useful selection marker is the enzyme glutamine synthase (GS) (Murphy et al., Biochem J. 22 7: 277-279 ( 1991 ); Bebbington et al.~, BiolTechnology l0:169-175 (1992)). Using these markers, the mammalian cells are grown in selective medium and the cells with the highest resistance are selected. These cell Iines contain the amplified genes) integrated into a chromosome. Chinese hamster ovary (CHO) cells are often used for the production of proteins.
The expression vectors pC 1 and pC4 contain the strong promoter (LTR) of the Rous Sarcoma Virus (Cullen et al., Molecular and Cellular Biology, 438-447 (March, 1985)) plus a fragment of the CMV-enhancer (Boshart et al., Ce1141:521-530 (1985)). Multiple cloning sites, e.g., with the restriction enzyme cleavage sites BamHI, XbaI and Asp718, facilitate the cloning of the gene of interest. The vectors contain in addition the 3' intron, the polyadenylation and termination signal of the rat preproinsulin gene.
A. Expression of Recombinant KGF-2 in COS Cells The expression of plasmid, KGF-2 HA was derived from a vector pcDNAI/Amp (Invitrogen) containing: I) SV40 origin of replication, 2) ampiciIlin resistance gene, 3) E. coli replication origin, 4) CMV promoter followed by a polylinker region, a SV40 intron and polyadenylation site. The HA
tag correspond to an epitope derived from the influenza hemagglutinin protein as previously described (Wilson, L, etal., Ce1137:767, (1984)). The infusion ofHA
tag to the target pmtein allows easy detection of the recombinant protein with an antibody that recognizes the HA epitope. A DNA fragment encoding the entire KGF-2 precursor HA tag fused in frame with the HA tag, therefore, the recombinant protein expression is directed under the CMV promoter.
The plasmid construction strategy is described as follows:

The DNA sequence encoding KGF-2, ATCC # 75977, is constructed by PCR using two primers: the 5' primer 5' TAACGAGGATCCGCCATCATGTGGAAATGGATACTGACAC 3' (SEQ
ID No. 9) contains a BamHI site followed by 22 nucleotides of KGF-2 coding sequence starting from the initiation codon; the 3' sequence 5' TAAGCACTCGAGTGAGTGTACCACCATTGGAAGAAATG 3' (SEQ ID
No. 10) contains complementary sequences to an XhoI site, HA tag and the last 26 nucleotides of the KGF-2 coding sequence (not including the stop codon).
Therefore, the PCR product contains a BamHI site, KGF-2 coding sequence followed by an XhoI site, an HA tag fused in frame, and a translation termination stop codon next to the HA tag. The PCR amplified DNA fragment and the vector, pcDNA-3'HA, are digested with BamHI and XhoI restriction enzyme and ligated resulting in pcDNA-3'HA-KGF-2. The ligation mixture is transformed into E. coli strain XLl Blue (Stratagene Cloning Systems, La Jolla, CA) the transformed culture is plated on ampicillin media plates and resistant colonies are selected. Plasmid DNA was isolated from transformants and examined by PAR
and restriction analysis for the presence of the correct fi~agment. For expression ofthe recombinant KGF-2, COS cells were transfected with the expression vector by DEAE-DEXTRAN method (Sambrook, J., et al., Molecular Cloning: A
Laboratory Manual, Cold Spring Laboratory Press, ( 1989)). The expression of the KGF-2 HA protein was detected by radiolabelling and immunoprecipitation method (Harlow, E. & Lane, D., Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, (1988)). Cells were labeled for 8 hours with 'SS-cysteine two days post transfection. Culture media were then collected and cells were lysed with detergent (RIPA buffer (150 mM NaCI, 1% NP-40, 0.1% SDS, 1% NP-40, 0.5% DOC, SOmM Tris, pH 7.5) (Wilson, L, et al., Id. 37:767 (1984)). Both cell lysate and culture media were precipitated with a HA
specific monoclonal antibody. Proteins precipitated were analyzed on 1 S% SDS-PAGE
gels.

B. F~nression and purification of human KGF-2 protein using the CHO Expression System The vector pC 1 is used for the expression of KFG-2 protein. Plasmid pC 1 is a derivative of the plasmid pSV2-dhfr [ATCC Accession No. 37146]. Both plasmids contain the mouse DHFR gene under control of the SV40 early promoter. Chinese hamster ovary- or other cells lacking dihydrofolate activity that are transfected with these plasmids can be selected by growing the cells in a selective medium (alpha minus MEM, Life Technologies) supplemented with the chemotherapeutic agent methotrexate. The amplification of the DHFR genes in cells resistant to methotrexate (MTX) has been well documented (see, e.g., Alt, F.W., Kellems, R.M., Bertino, J.R., and Schimke, R.T., 1978, J. Biol. Chem.
253:1357-1370, Hamlin, J.L. and Ma, C. 1990, Biochem. et Biophys. Acta, 1097:107-143, Page, M.J. and Sydenham, M.A. 1991, Biotechnology Vol. 9:64-68). Cells grown in increasing concentrations of MTX develop resistance to the drug by overproducing the target enzyme, DHFR, as a result of amplification of the DHFR gene. If a second gene is linked to the DHFR gene it is usually co amplified and over-expressed. It is state of the art to develop cell lines carrying more than 1,000 copies of the genes. Subsequently, when the methotrexate is withdrawn, cell lines contain the amplified gene integrated into the chromosome(s).
Plasmid pC 1 contains for the expression of the gene of interest a strong promoter of the long terminal repeat (LTR) of the Rouse Sarcoma Virus (Cullen, et al., Molecular and Cellular Biology, March 1985:438-4470) plus a fragment isolated from the enhancer of the immediate early gene of human cytomegalovirus (CMV) (Boshart et al., Cell 41:521-530, 1985). Downstream of the promoter are the following single restriction enzyme cleavage sites that allow the integration of the genes: BamHI, Pvull, and Nrul. Behind these cloning sites the plasmid contains translational stop codons in all three reading frames followed by the 3' intron and the polyadenylation site of the rat preproinsulin gene. Other high efficient promoters can also be used for the expression, e.g., the human ~i-actin promoter, the SV40 early or late promoters or the long terminal repeats from other retroviruses, e.g., HIV and HTLVI. For the polyadenylation ofthe mRNA other signals, e.g., from the human growth hormone or globin genes can be used as well.
Stable cell lines carrying a gene of interest integrated into the chromosomes can also be selected upon co-transfection with a selectable marker such as gpt, 6418 or hygromycin. It is advantageous to use more than one selectable marker in the beginning, e.g., G418 plus methotrexate.
The plasmid pC 1 is digested with the restriction enzyme BamHI and then dephosphorylated using calf intestinal phosphates by procedures known in the art.
The vector is then isolated from a 1 % agarose gel.
The DNA sequence encoding KFG-2, ATCC No. 75977, is amplified using PCR oligonucleotide primers corresponding to the S' and 3 ' sequences of the gene:
The 5' primer has the sequence 5'TAACGAGGATCCGCCATCATGTGGAAATGGATACTGACAC 3' (SEQ
ID No. 9) containing the underlined BamH 1 restriction enzyme site followed by 21 bases of the sequence of KGF-2 of Figure 1 (SEQ ID NO:1 ). Inserted into an expression vector, as described below, the 5' end of the amplified fragment encoding human KGF-2 provides an efficient signal peptide. An efficient signal for initiation of translation in eukaryotic cells, as described by Kozak, M., J. Mol.
Biol. 196:947-950 (1987) is appropriately located in the vector portion of the construct.
The 3' primer has the sequence 5' TAAGCAGGATCCTGAGTGTACCACCATTGGAAGAAATG 3' (SEQ ID
NO. 10) containing the BamHl restriction followed by nucleotides complementary to the last 26 nucleotides of the KGF-2 coding sequence set out in Figure 1 (SEQ ID NO:1 ), not including the stop codon.
The amplified fragments are isolated from a 1 % agarose gel as described above and then digested with the endonuclease BamHI and then purified again on a 1 % agarose gel.
The isolated fragment and the dephosphorylated vector are then ligated with T4 DNA ligase. E. coli HB101 cells are then transformed and bacteria identified that contained the plasmid pC 1. The sequence and orientation of the inserted gene is confirmed by DNA sequencing.
Transfection of CH~DHFR-cells Chinese hamster ovary cells lacking an active DHFR enzyme are used for transfection. 5 ~g of the expression plasmid C 1 are cotransfected with 0.5 pg of the plasmid pSVneo using the lipofecting method (Felgner et al., supra). The plasmid pSV2-neo contains a dominant selectable marker, the gene neo from Tn5 encoding an enzyme that confers resistance to a group of antibiotics including 6418. The cells are seeded in alpha minus MEM supplemented with 1 mg/ml 6418. After 2 days, the cells are trypsinized and seeded in hybridoma cloning plates (Greiner, Germany) and cultivated for 10-I4 days. Afterthis period, single clones are trypsinized and then seeded in 6-well petri dishes using different concentrations of methotrexate (25 nM, 50 nM, 100 nM, 200 nM, 400 nM).
Clones growing at the highest concentrations of mettlotrexate are then transferred to new 6-well plates containing even higher concentrations of methotrexate (500 nM, 1 pM, 2 pM, 5 ~.M). The same procedure is repeated until clones grow at a concentration of 100 p,M.
The expression of the desired gene product is analyzed by Western blot analysis and SDS-PAGE.
Example S
Transcription and Translation of Recombinant KGF-2 in vitro A PCR product is derived f mm the cloned cDNA in the pA2 vector used for insect cell expression of KGF-2. The primers used for this PCR were:
5'ATTAACCCTCACTAAAGGGAGGCCATGTGGAAATGGATACTGACA
CATTGTGCC 3' (SEQ ID No. 11) and 5' CCCAAGCTTCCACAAACGTTGCCTTCCTCTATGAG 3' (SEQ ID No.12).
The first primer contains the sequence of a T3 promoter S' to the ATG
initiation colon. The second primer is complimentary to the 3' end of the KGF-open reading frame, and encodes the reverse complement of a stop colon.
The resulting PCR product is purified using a commercially available kit from Qiagen. 0.5 ~,g of this DNA is used as a template for an in vitro transcription-translation reaction. The reaction is performed with a kit commercially available from Promega under the name of TNT. The assay is performed as described in the instructions for the kit, using radioactively labeled methionine as a substrate, with the exception that only %2 of the indicated volumes of reagents are used and that the reaction is allowed to proceed at 33 °C for 1.5 hours.
Five ~,1 of the reaction is electrophoretically separated on a denaturing 10 to 15% polyacrylamide gel. The gel is fixed for 30 minutes in a mixture of water:Methanol:Acetic acid at 6:3:1 volumes respectively. The gel is then dried under heat and vacuum and subsequently exposed to an X-ray film for 16 hours.
The film is developed showing the presence of a radioactive protein band corresponding in size to the conceptually translated KGF-2, strongly suggesting that the cloned cDNA for KGF-2 contains an open reading frame that codes for a protein of the expected size.
Example 6 Expression via Gene Therapy Fibroblasts are obtained from a subject by skin biopsy. The resulting tissue is placed in tissue-culture medium and separated into small pieces.
Small chunks of the tissue are placed on a wet surface of a tissue culture flask, approximately ten pieces are placed in each flask. The flask is fumed upside down, closed tight and le$ at room temperature over night. After 24 hours at room temperature, the flask is inverted and the chunks of tissue remain fixed to the bottom of the flask and fresh media (e.g., Ham's F12 media, with 10% FBS, penicillin and streptomycin, is added.) This is then incubated at 37°C
for approximately one week. At this time, fresh media is added and subsequently changed every several days. After an additional two weeks in culture, a monolayer of fibroblasts emerge. The monolayer is trypsinized and scaled into larger flasks.
pMV-7 (Kirschmeier, P.T. et al, DNA, 7:219-25 (1988)) flanked by the long terminal repeats of the Moloney marine sarcoma virus, is digested with EcoRI and HindIII and subsequently treated with calf intestinal phosphatase.
The linear vector is fractionated on agarose gel and purified, using glass beads.
The cDNA encoding a polypeptide of the present invention is amplified using PCR primers which correspond to the 5' and 3' end sequences respectively.
The 5' primer containing an EcoRI site and the 3' primer further includes a HindIII
site. Equal quantities of the Moloney marine sarcoma virus linear backbone and the amplified EcoRI and HindIII fragment are added together, in the presence of T4 DNA ligase. The resulting mixture is maintained under conditions appropriate for ligation of the two fragments. The ligation mixture is used to transform bacteria HB 1 O 1, which are then plated onto agar-containing kanamycin for the purpose of confirming that the vector had the gene of interest properly inserted.
The amphotropic pA317 or GP+aml2 packaging cells are grown in tissue culture to confluent density in Dulbecco's Modified Eagles Medium (DMEM) with 10% calf serum (CS), penicillin and streptomycin. The MSV vector containing the gene is then added to the media and the packaging cells are transduced with the vector. The packaging cells now produce infectious viral particles containing the gene (the packaging cells are now referred to as producer cells).
Fresh media is added to the transduced producer cells, and subsequently, the media is harvested from a 10 cm plate of confluent producer cells. The spent media, containing the infectious viral particles, is filtered through a millipore filter to remove detached producer cells and this media is then used to infect fibroblast cells. Media is removed from a sub-confluent plate of fibroblasts and quickly replaced with the media from the producer cells. This media is removed and replaced with fresh media. If the titer of virus is high, then virtually all fibroblasts will be infected and no selection is required. If the titer is very low, then it is necessary to use a retroviral vector that has a selectable marker, such as neo or his.
The engineered fibroblasts are then injected into the host, either alone or after having been grown to confluence on cytodex 3 microcarrier beads. The fibroblasts now produce the protein product.
Example 7 to Tissue distribution of KGF 2 mRNA expression Northern blot analysis is carried out to examine the levels of expression of the gene encoding the KGF-2 protein in human tissues, using methods described by, among others, Sambrook et al. , cited above. A probe corresponding to the entire open reading frame of KGF-2 of the present invention (SEQ ID
NO: l ) was obtained by PCR and was labeled with'ZP using the rediprimeTM DNA
labeling system (Amersham Life Science), according to manufacturer's instructions. After labelling, the probe was purified using a CHROMA SPIN-1 OOTM column (Clontech Laboratories, Inc.), according to manufacturer's protocol number PT1200-1. The purified labelled probe was then used to examine various human tissues for the expression of the gene encoding KGF-2.
Multiple Tissue Northern (MTN) blots containing poly A RNA from various human tissues (H) or human immune system tissues (IM) were obtained from Clontech and were examined with labelled probe using ExpressHybTM
Hybridization Solution (Clontech) according to manufacturer's protocol number PT1190-1. Following hybridization and washing, the blots are mounted and exposed to film at -70°C overnight, and films developed according to standard procedures.
A major mRNA species of apporximately 4.2 kb was observed in most human tissues. The KGF-2 mRNA was relatively abundant in heart, pancreas, placenta and ovary. A minor mRNA species of about 5.2 kb was also observed ubiquitously. The identity of this 5.2 kb mRNA species was not clear. It is possible that the 5.2 kb transcript encodes an alternatively spliced form of or a third member of the KGF family. The KGF-2 cDNA was 4.1 kb, consistent with the size of the mRNA of 4.2 kb.
Example 8 Keratinocyte Proliferation Assays Dermal keratinocytes are cells in the epidermis of the skin. The growth and spreading of keratinocytes in the skin is an important process in wound healing. A proliferation assay of keratinocyte is therefore a valuable indicator of protein activities in stimulating keratinocyte growth and consequently, wound healing.
Keratinocytes are, however, difficult to grow in vitro. Few keratinocyte cell lines exist. These cell lines have different cellular and genetic defects. In order to avoid complications of this assay by cellular defects such as loss of key growth factor receptors or dependence of key growth factors for growth, primary dermal keratinocytes are chosen for this assay. These primary keratinocytes are obtained from Clonetics, Inc. (San Diego, CA).
K~ratxnocvte proliferation assay with alam_a_TBlue alamarBlue is a viable blue dye that is metabolized by the mitochondria when added to the culture media. The dye then turns red in tissue culture supernatants. The amounts of the red dye may be directly quantitated by reading difference in optical densities between 570 nln and 600 nm. This reading reflects cellular activities and cell number.
Normal primary dermal keratinocytes ( CC-025 S, NHEK-Neo pooled) are purchased from Clonetics, Inc. These cells are passage 2. Keratinocytes are grown in complete keratinocyte growth media (CC-3001, KGM; Clonetics, Inc.) until they reach 80% confluency. The cells are trypsinized according to the manufacturer's specification. Briefly, cells were washed twice with Hank's balanced salt solution. 2-3 ml of trypsin was added to cells for about 3-S min at room temperature. Trypsin neutralization solution was added and cells were collected. Cells are spun at 600 xg for S min at room temperature and plated into new flasks at 3,000 cells per square centimeter using pre-warmed media.
For the proliferation assay, plate 1,000-2,000 keratinocytes per well of the Corning flat bottom 96-well plates in complete media except for the outermost rows. Fill the outer wells with 200 ,ul of sterile water. This helps to keep temperature and moisture fluctuations of the wells to the minimum. Gmw cells overnight at 37 °C with 5% CO2. Wash cells twice with keratinocyte basal media (CC-3101, KBM, Clonetics, Inc.) and add 100 ~l of KBM into each well.
Incubate for 24 hours. Dilute growth factors in KBM in serial dilution and add 15 100 ~cl to each well. Use KGM as a positive control and KBM as a negative control. Six wells are used for each concentration point. Incubate for two to three days. At the end of incubation, wash cells once with KBM and add 100 ~cl of KBM with 10% v/v alamarBlue pre-mixed in the media. Incubate for 6 to 16 hours until media color starts to turn red in the KGM positive control.
Measure 20 O.D. 570 nm minus O.D. 600 nm by directly placing plates in the plate reader.
Results Stimulation of Keratinocyte proliferation by KGF 2 To demonstrate that KGF-2 (Cys37-Ser208 with a 6X (His) tag (SEQ ID
NOs: 29-30; Figure 5) and N-terminal deletion mutants KGF-2033 and KGF-25 228 were active in stimulating epidermal keratinocyte growth, normal primary human epidermal keratinocytes were incubated with the E. coli-expressed and purified KGF-2 protein (batch number E3), KGF-233 (batch number E1) and KGF-2028 (batch number E2). The KGF-2 protein stimulated the growth of epidermal keratinocytes with an EC50 of approximately 5 ng/ml, equivalent to 30 that of FGF7/KGF-1 (Figure 6A). In contrast, other FGF's such as FGF-1 and FGF-2 did not stimulate the growth of primary keratinocytes. The EC50 for KGF-233 was 0.2 ng/ml and that for KGF-2028 2ng/ml (See Figures 6B and C).
Thus, KGF-2 appeared to be as potent as FGF7/KGF in stimulating the proliferation of primary epidermal keratinocytes. However, KGF-2033 is more potent in stimulating keratinocyte proliferation than the "Cys (37)" KGF-2 described above and the KGF-228.
Scarring of wound tissues involves hyperproliferation of dermal fibroblasts. To determine whether the stimulatory effects of KGF-2 was specific for keratinocytes but not for fibroblasts, mouse Balb.c.3T3 fibroblasts and human lung fibroblasts were tested. Neither types of fibroblasts responded to KGF-2 in proliferation assays. Therefore, KGF-2 appeared to be a mitogen specific for epidermal keratinocytes but not mesenchymal cells such as fibroblasts. This suggested that the likelihood of KGF-2 causing scarring of the wound tissues was low.
i 5 Example 9 A. Mitogenic Effects of KGF 2 on Cells Transfected with Spec fc FGF
Receptors To determine which FGF receptor isoform(s) mediate the proliferative effects of KGF-2, the effects of KGF-2 on cells expressing specific FGF
receptor isoforms were tested according to the method described by Santos-Ocampo et al.
J. Biol. Chem. 271:1726-1731 (1996). FGF7/KGF was known to induce mitogenesis of epithelial cells by binding to and specifically activating the FGFR2iiib form (Mild et al. Science 251:72-75 ( 1991 )). Therefore, the proliferative effects of KGF-2 in mitogenesis assays were tested using cells expressing one of the following FGF receptor isoforms: FGFR1 iiib, FGFR2iiib, FGFR3iiib, and FGFR4.
Mitogenesis assay of cells exnressin FGF receptors Thymidine incorporation of BaF3 cells expressing specific FGF receptors were performed as described by Santos-Ocampo et al. J. Biol. Chem. 271:1726-1731 ( 1996). Briefly, BaF3 cells expressing specific FGF receptors were washed and resuspended in Dubeco's modified Eagle's medium, 10% neonatal bovine serum, L-glutanime. Approximately 22,500 cells were plated per well in a 96-well assay plate in media containing 2 ~.g/ml Heparin. Test reagents were added to each well for a total volume of 200 ~1 per well. The cells were incubated for 2 days at 37 °C. To each wll, l ~Ci of 3H-thymidine was then added in a volume of 50 ~,1. Cells were harvested after 4-5 hours by filtration through glass fiber paper. Incorporated 3H-thymidine was counted on a Wallac beta plate scintillation counter.
Results The results revealed that KGF-2 protein (Thr (36) - Ser (208) of Figure 1 (SEQ ID N0:2) with a N-terminal Met added thereto) strongly stimulated the proliferation of Baf3 cells expressing the KGF receptor, FGFR2iiib isoform, as indicated by 3H-thymidine incorporation (Figure 7A). Interestingly, a slight stimulatory effect of KGF-2 on the proliferation of Baf3 cells expressing the FGFRliiib isoform was observed. KGF-2 did not have any effects on cells expressing the FGFR3iiib or the FGFR4 forms of the receptor.
FGF7/KGF stimulated the proliferation of cells expressing the KGF
receptor, FGFR2iiib but not FGFRliiib isoform. The difference between KGF-2 and FGF7/KGF was intriguing. In the control experiments, aFGF stimulated its receptors, FGFR1 iiib and iiic and bFGF stimulated its receptor FGFR2iiic.
Thus, these results suggested that KGF-2 binds to FGFR2iiib isoform and stimulates mitogenesis. In contrast to FGF7/KGF, KGF-2 also binds FGFR 1 iiib isoform and stimulates mitogenesis.
B. Mitogenic e, ffects ofICGF-2d 33 on Cells Tran sfected with Specif:c FGF
Receptors As demonstrated above FGFs or KGF-1 and -2 both bind to and activate the FGF 2iiib receptor (FGFR 2iiib). The proliferative effects of KGF-233 in mitogenesis assays were tested using cells expressing one of the following FGF
receptor isoforms: FGFR2iiib or FGFR2iiic (the 2iiic receptor-transfected cells are included as a negative control).
The experiments were performed as above in part A of this example.
Briefly, BaF3 cells were grown in RPMI containing 10% bovine calf serum (BCS
- not fetal serum), 10% conditioned medium from cultures of WEHI3 cells (grown in RPMI containing 5%BCS), SOnM ~i-mercaptoethanol, L-Glu (2% of a 1 OOX stock) and pen/strep ( 1 % of a 100X stock).
For the assay, BaF3 cells were rinsed twice in RPMI medium containing I O% BCS and 1 gg/ml heparin. BaF3 cells (22,000/well) were plated in a 96-well plate in 150 ~1 of RPMI medium containing 10% BCS and I ~,g/ml heparin.
Acidic FGF, basic FGF, KGF-I {HG15400) or KGF-2 proteins (HG03400, 03401, 03410 or 03411) were added at concentrations from approximately 0 to 10 nM. The cells were incubated in a final volume of 2001 for 48 hours at 37°C.
All assays were done in triplicate. Tritiated thymidine {0.5 ~Ci) was added to each well for 4 hours at 37°C and the cells were then harvested by filtration through a glass fiber filter. The total amount of radioactivity incorporated was then determined by liquid scintillation counting. The following positive controls were used: basic FGF and acidic FGF for FGFR2iiic cells; acidic FGF and KGF-1 for FGFR2iiib cells. The following negative controls were used: Basal medium (RPMI medium containing 10% BCS and I ~,g/ml heparin).
Results The results revealed that KGF-2 (Thr (36) - Ser (208) with N-terminal Met added), KGF-2033 and KGF-228 proteins strongly stimulated the proliferation of BaF3 cells expressing the KGF receptor, FGFR2iiib isoform, as indicated by3H-thymidine incorporation (Figures 7A-C). The KGF-2 proteins did not have any effects on cells expressing the FGFR2iiic forms of the receptor.
These results suggested that KGF-2 proteins bind to FGFR2iiib isoform and stimulates mitogenesis. In addition, it appears that KGF-2033 was able to stimulate the proliferation of the BaF3 cells better than the KGF-2 (Thr (36) -Ser (208)).
Example 10 A. Construction ojE. coli Optimized Full Length KGF 2 In order to increase expression levels of full length KGF-2 in an E. toll expression system, the codons of the amino terminal portion of the gene were optimized to highly used E. toll codons. For the synthesis of the optimized region of KGF-2, a series of six oligonucleotides were synthesized: numbers 1-(sequences set forth below). These overlapping oligos were used in a PCR
reaction for seven rounds at the following conditions:
Denaturation 95 degrees 20 seconds Annealing 58 degrees 20 seconds Extension 72 degrees 60 seconds A second PCR reaction was set up using 1 ~,1 of the first PCR reaction with KFG-2 sythetic primer 6 as the 3' primer and KGF-2 synthetic 5' BamHI as the 5' primer using the same conditions as described above for 25 cycles. The product produced by this final reaction was restricted with AvaII and BamHI.
The KGF-2 construct of Example 1 was restricted with AvaII and HindIII and the fragment was isolated. These two fragments were cloned into pQE-9 restricted with BamHI and HindIII in a three fragment ligation.
Primers used for constructing the optimized synthetic KGF-2 1 /208 KGF-2 Synthetic Primer 1:
ATGTGGAAATGGATACTGACCCACTGCGCTTCTGCTTTCCCGCACC
TGCCGGGTTGCTGCTGCTGCTGCTTCCTGCTGCTGTTC (SEQ ID NO: 31 ) KGF-2 Synthetic Primer 2:
CCGGAGAAACCATGTCCTGACCCAGAGCCTGGCAGGTAACCGGAA
CAGAAGAAACCAGGAACAGCAGCAGGAAGCAGCAGCA (SEQ
ID NO: 32) KGF-2 Synthetic Primer 3:
GGGTCAGGACATGGTTTCTCCGGAAGCTACCAACTCTTCTTCTTCTT
CTTTCTCTTCTCCGTCTTCTGCTGGTCGTCACG (SEQ ID NO: 33) KGF-2 Synthetic Primer 4:
GGTGAAAGAGAACAGTTTACGCCAACGAACGTCACCCTGCAGGTG
GTTGTAAGAACGAACGTGACGACCAGCAGAAGACGG (SEQ ID
NO:34) KGF-2 Synthetic Primer 5:
CGTTGGCGTAAACTGTTCTCTTTCACCAAATACTTCCTGAAAATCGA
AAAAAACGGTAAAGTTTCTGGGACCAAA (SEQ ID N0:35) KGF-2 Synthetic Primer 6:
TTTGGTCCCAGAAACTTTACCGTTTTTTTCGATTTTCAG (SEQ ID
N0:36) KGF-2 Synthetic 5' BamHI
AAAGGATCCATGTGGAAATGGATACTGACCCACTGC (SEQ ID N0:37) The resulting clone is shown in Figure 10 (SEQ ID NOS: 38 and 39).
B. Construction of E. cold Optimized Mature KGF 2 In order to further increase expression levels of the mature form of KGF-2 in an E. coli expression system, the codons of the amino terminal portion of the gene were optimized to highly used E. coli codons. To correspond with the mature form of KGF-1, a truncated form of KGF-2 was constructed starting at threonine 36. E. coli synthetic KGF-2 from Example 10 A was used as a template in a PCR reaction using BspHI 5' KGF-2 as the 5' primer (sequence given below) and HindIII 3' KGF-2 as the 3' primer (sequence given below). Amplification was performed using standard conditions as given above in Example 10 A for 25 cycles. The resulting product was restricted with BspHI and HindII and cloned into the E. coli expression vector pQE60 digested with NcoI and HindIII.
BspHI 5' KGF-2 Primer:

TTTCATGACTTGTCAAGCTCTGGGTCAAGATATGGTTC (SEQ ID N0:40) HindIII 3' KGF-2 Primer:
GCCCAAGCTTCCACAAACGTTGCCTTCC (SEQ ID N0:41 ) The resulting clone is shown in Figure 11A (SEQ ID N0:42 and 43).
C. Construction of an Alternate E. coli Optimized Mature KGF-2 In order to further increase expression levels of the mature form of KGF-2 in an E. coli expression system, the codons of 53 amino acids at the amino terminal portion of the E. coli optimized gene were changed to alternate highly used E. coli codons. For the synthesis of the optimized region of KGF-2, a series ofsix oligonucleotides were synthesized: numbers 18062,18061,18058,18064, 18059, and 18063 (sequences set forth below). These overlapping oligos were used in a PCR reaction for seven rounds at the following conditions:
Denaturation 95 degrees 20 seconds Annealing 58 degrees 20 seconds Extension 72 degrees 60 seconds Following the seven rounds of synthesis, a S' primer to this region,18169 and a 3' primer to this entire region, 18060, were added to a PCR reaction, containing 1 microliter from the initial reaction of the six oligonucleotides.
This product was amplified for 30 rounds using the following conditions:
Denaturation 95 degrees 20 seconds Annealing SS degrees 20 seconds Extension 72 degrees 60 seconds A second PCR reaction was set up to amplify the 3' region of the gene using primers 18066 and 18065 under the same conditions as described above for 25 rounds. The resulting products were separated on an agarose gel. GeI slices containing the product were diluted in 1 OmM Tris, 1 mM -EDTA, pH 7.5 One microliter each from each of diluted gel slices were used in an additional PCR

reaction using primer 18169 as the 5' primer, and primer 18065 as the 3' primer.
The product was amplified for 25 cycles using the same conditions as above.
The product produced by this final reaction was and restricted with Eco R 1 and HindIII, and cloned into pQE60, which was also cut with Eco Rl and HindIII
(pQE6 now).
Sequences of the 5' Synthetic Primers:
18169 KGF2 5' EcoRI/I~
TCAGTGAATTCATTAAAGAGGAGAAATTAATCATGACTTGCCAGG
jSEQ ID N0:44]
18062 KGF2 synth new R1 sense TCATGACTTGCCAGGCACTGGGTCAAGACATGGTTTCCCCGGAAGCTA
[SEQ ID N0:45]
18061 KGF2 synth R2 sense' GCTTCAGCAGCCCATCTAGCGCAGGTCGTCACGTTCGCTCTTACAACC
[SEQ ID N0:46]
18058 KGF2 Synth R3 sense GTTCGTTGGCGCAAACTGTTCAGCTTTACCAAGTACTTCCTGAAAATC
[SEQ ID N0:47) 18066 KGF 2 20 by rya II sense TCGA,AAAAAACGGTAAAGTTTCTGGGAC (SEQ ID N0:48]
18064 KGF2 synth F 1 antisente' GATGGGCTGCTGAAGCTAGAGCTGGAGCTGTTGGTAGCTTCCGGGG
AA [SEQ ID N0:49]
_18059 KGF2 Srnth F2 antisence~
AACAGTTTGCGCCAACGAACATCACCCTGTAAGTGGTTGTAAGAG
[SEQ ID NO:SO]
18063 KGF2 Sy~t_h F'~ antisense~
TTCTTGGTCCCAGAAACTTTACCG'ITTTTTZ'CGATTTTCAGGAAGTA
[SEQ ID NO:S 1 18060 KGF 2 Ava II andsense~

TTCTTGGTCCCAGAAACTTTACCG [SEQ ID N0:52]
18065 KGF2 HindIII 3' Ston AGATCAGGCTTCTATTATTATGAGTGTACCACCATTGGAAGAAAG
[SEQ ID N0:53]
The sequence of the synthetic KGF-2 gene and it corresponding amino acid is shown in Figure 11B (SEQ ID NO: 54 and 55) Example ll Construction of KGF 2 Deletion Mutants Deletion mutants were constructed from the 5' terminus and 3' terminus of KGF-2 gene using the optimized KGF-2 construct from Example 10 A as a template. The deletions were selected based on regions of the gene that might negatively affect expression in E. coli. For the 5' deletion the primers listed below were used as the 5' primer. These primers contain the indicated restriction site and an ATG to code for the initiator methionine. The KGF-2 (FGF-12) 208 amino acid 3' HindIII primer was used for the 3' primer. PCR amplification for rounds was performed using standard conditions as set forth in Example 10.
The products for the KGF-2 36aa/208aa deletion mutant were restricted BspHI
for the 5' site and HindIII for the 3' site and cloned into the pQE60 which has been digested with BspHI and HindIII. All other products were restricted with NcoI
20 for the 5' restriction enzyme and HindIII for the 3' site, and cloned into the pQE60 which had been digested with NcoI and HindIII. For KGF-2 (FGF-12), 36aa/153aa and 128aa 3' HindIII was used as the 3' primer with FGF-12 36aa/208aa as the 5' primer. For FGF-12 62aa/153aa, 128aa 3' HindIII was used as the 3' primer with FGF-12 62aa/208aa as the 5' primer. The nomenclature of 25 the resulting clones indicates the first and last amino acid of the polypeptide that results from the deletion. For example, KGF-2 36aa/153aa indicates that the first amino acid of the deletion mutant is amino acid 36 and the last amino acid is amino acid 153 of KGF-2. Further, as indicated in Figures 12-20, each mutant has N-terminal Met added thereto.
Sequences of the Deletion Primers:
FGF 12 36aa/208aa~
5' BsphI GGACCCTCATGACCTGCCAGGCTCTGGGTCAGGAC [SEQ ID
N0:56]
FGF12 63aa/208aa-5' NcoI GGACAGCCATGGCTGGTCGTCACGTTCG [SEQ ID N0:57]
FGF12 77aa~208aa~
5' NcoI GGACAGCCATGGTTCGTTGGCGTAAACTG [SEQ ID N0:58]
FGF12 93aa/208aa~
5' NcoI GGACAGCCATGG~~~AAAApCGGTAAAGTTTC [SEQ ID N0:59]
FGF 12 104aa/208aa~
5' NcoI GGACCCCCATGGAGAACTGCCCGTAGAGC [SEQ ID N0:60]
FGF 12 123aa/208aa~
5' NcoI GGACCCCCATGGTCAAAGCCATTAACAGCAAC [SEQ ID N0:61 FGF 12 138aa/208aa~
5' NcoI GGACCCCCATGGGGAApCTCTATGGCTCAAA.AG [SEQ ID
N0:62]
FGF 12 3' HindIII ~ (Used for all above deletion clones) CTGCCCAAGCTTATTATGAGTGTACCACCATTGGAAG (SEQ ID N0:63]
FGF12 36aa/153aa~
5' BsphI (as above) 3'HindIII CTGCCCAAGCTTATTACTTCAGCTTACAGTCATTGT [SEQ ID
N0:64]
FGF12 63aa/153aa 5'NcoI and 3'HindIII, as above The sequences for the resulting deletion mutations are set forth in Figures 12-20. [SEQ ID NOS:65-82]

Example 12 Construction oJCysteine Mutants oJKGF 2 Construction of C-37 mutation primers 5457 5' BsphI and 5258 173aa 3' HindIII were used to amplify the KGF-2 (FGF- I2) template from Example 10 A.
Primer 5457 5' BsphI changes cysteine 37 to a serine. Amplification was done using the standard conditions outlined above in Example 10 A for 25 cycles.
The resulting product was restricted with BspHI and HindIII and cloned into E.
coli expression vector pQE60, digested with BspHI and HindIII. (Figure 21 ) [SEQ ID
N0:83]
For mutation of Cysteine 106 to serine, two PCR reactions were set up for oligonucleotide site directed mutagenesis of this cysteine. In one reaction, BsphI was used as the 5' primer, and 5455 was used as the 3' primer in the reaction. In a second reaction, 5456 was used as the 5' primer, and 5258 HindIII
was used as the 3' primer. The reactions were amplified for 25 rounds under standard conditions as set forth in Example 10. One microliter from each of these PCR reactions was used as template in a subsequent reaction using, as a 5' primer, 5453 BspHI, and as a 3' primer, 5258 HindIII. Amplification for 25 rounds was performed using standard conditions as set forth in Example 10. The resulting product was restricted with BspHI and HindIII and cloned into the E. coli expression vector pQE60, which was restricted with NcoI and HindIII.
Two PCR reactions were required to make the C-37/C-106 mutant.
Primers 5457 Bsphl and 5455 were used to create the 5' region of the mutant containing cysteine 37 to serine substitution, and primer 5456 and 5258 HindIII
were used to create the 3' region of the mutant containing cysteine 106 to serine substitution. In the second reaction, the 5457 BsphI primer was used as the 5' primer and the 5258 HindIII primer was used as the 3' primer to create the C-37/C-106 mutant using 1 pl from each of the initial reactions together as the template. This PCR product was restricted with BsphI and HindIII, and cloned into pQE60 that had been restricted with NcoI and HindIII. The resulting clone is shown in Figure 22 (SEQ ID N0:84) Sequences of the Cysteine Mutant Primers:
5457 BspHI: GGACCCTCATGACCTCTCAGGCTCTGGGT (SEQ ID N0:85) 5456: AAGGAGAACTCTCCGTACAGC (SEQ ID NO: 86) 5455: GCTGTACGGTCTGTTCTCCTT (SEQ ID NO: 87) 54535453 BsnHI: GGACCCTCATGACCTGCCAGGCTCTGGGTCAGGAC (SEQ
ID NO: 88) 5258 HindIII: CTGCCCAAGCTTATTATGAGTGTACCACCATTGGAAG
(SEQ ID NO: 89) Example 13 Production and Purification oJBGF 2 (FGF 12) The DNA sequence encoding the optimized mature protein described in Example 10 B (i.e., amino acids T36 through 5208 of KGF-2) was cloned into plasmid pQE-9 (Qiagen). E. coli (MlS/rep4;Qiagen) were grown to stationary phase overnight at 37°C in LB containing 100 ~g/ml Ampicillin and 25 ~g/ml Kanamycin. This culture was used to innoculate fresh LB media containing containing 100 pg/ml Ampicillin and 25 pg/ml Kanamycin at a dilution of 1:50.
The cells were grown at 37°C to an O.D.s9s of 0.7, induced by the addition of isopropyl 1-thin-b-D-galactopyranoside (IPTG) to a final concentration of lmM.
After 3-4 hours, the cells were harvested by centrifugation, and resuspended in a buffer containing 60mM NaP04 and 360mM NaCI at a ratio of 5 volumes of buffer: 1 volume of cell paste. After disruption in a Mautin Gaulin, the extract was adjusted to pH to 8.0 by the addition of NaOH and clarified by centrifugation.
The clarified soluble extract was applied to a Poros HS-50 column (2.OX 10.0 cm; PerSeptive Biosystems, Inc.) and bound proteins step-eluted with SOmM NaP04 pH 8.0 containing O.SM,1.OM and 1.SM NaCI. The KGF-2 eluted in the 1.SM salt fraction which was then diluted five-fold with SOmM NaP04 pH

WO 99/41282 PCT/US99/030i8 6.5 to a final salt concentration of 300mM. This KGF-2 containing fraction was then passed sequentially over a Poros HQ-20 column (2.0X7.0 cm; PerSeptive Biosystems, Inc.) and then bound to a Poros CM-20 column (2.0X9.0 cm;
PerSeptive Biosystems, Inc.). KGF-2 (FGF-12)-containing fractions that eluted at about SOOmM to about 750 mM NaCI were pooled, diluted and re-applied to an CM-20 column to concentrate. Finally, the protein was seperated on a gel filtration column (S-75; Pharmacia) in 40mM NaOAC pH6.5; 150mM NaCI
(Batch E-5) Alternatively, the gel filtration column was run in Phosphate Buffered Saline (PBS, Batch E-4). KGF-2 containing fractions were pooled and protein concentration determined by Bio-R.ad Protein Assay. Proteins were judged to be >90% pure by SDS-PAGE. Finally, endotoxin levels determined by Limulus Amebocyte Lysate Assay (Cape Cod Associates) were found to be _<
lEu/mg. Proteins prepared in this way were able to bind heparin which is a hallmark of FGF family members.
Example 14 A. Construction of N terminal deletion mutant KGF 2433 To increase the level of expression of KGF2 in E.coli, and to enhance the solubilty and stability properties of E.coli expressed KGF2, a deletion variant KGF-233 (KGF-2 as 69-208) (SEQ ID N0:96) which removes the first 68 amino acids of the pre-processed KGF2 was generated. The rationale for creating this deletion variant was based on the following observations. Firstly, mature KGF2 (KGF-2 as 36-208) contains an uneven number (three) of cysteine residues which can lead to aggregation due to infra-molecular disulphide bridge formation.
The KGF X33 deletion variant contains only two cysteine residues, which reduces the potential for infra-molecular disulphide bridge formation and subsequent aggregation. A decrease in aggregation should lead to an increase in the yield of active KGF2 protein. Secondly, the KGF X33 deletion variant removes a poly-serine stretch which is not present in KGF-1 and does not appear to be important for activity, but may hinder expression of the protein in E. coli.
Thus, removal of the poly-serine stretch may increase expression levels of active KGF-2 protein. Thirdly, expression of KGF 033 in E.coli, results in natural cleavage of KGF-2 between residues 68 and 69. Thus, it is anticipated that 033 will be processed efficiently and will be stable in E.coli.
Construction of KGF-233 in pQE6 To permit Polymerase Chain Reaction directed amplification and sub-cloning of KGF2 X33 into the E.coli protein expression vector, pQE6, two oligonucleotide primers (5952 and 19138) complementary to the desired region of KGF2 were synthesized with the following base sequence.
Primer 5952: 5' GCGGCACATGTCTTACAACCACCTGCAGGGTG 3' (SEQ
ID N0:91 ) Primer 19138: S' GGGCCCAAGCTTATGAGTGTACCACCAT 3' (SEQ ID
N0:92) In the case of the N-terminal primer (5952), an AflIII restriction site was incorporated, while in the case of the C-terminal primer (19138) a HindIII
restriction site was incorporated. Primer 5952 also contains an ATG sequence adjacent and in frame with the KGF2 coding region to allow translation of the cloned fragment in E.coli, while primer 19138 contains two stop codons (preferentially utilized in E.coli) adjacent and in frame with the KGF2 coding region which ensures correct translational termination in E.coli.
The Polymerase Chain Reaction was performed using standard conditions well known to those skilled in the art and the nucleotide sequence for the mature KGF-2 {aa 36-208) (constructed in Example lOC) as template. The resulting amplicon was restriction digested with AflIII and HindIII and subcloned into Ncol/HindIII digested pQE6 protein expression vector.

Construction of KGF-233 in pHEl To permit Polymerise Chain Reaction directed amplification and subcloning of KGF2 X33 into the E.coli expression vector, pHEI, two oligonucleotide primers (6153 and 6150) corresponding to the desired region of KGF2 were synthesized with the following base sequence.
Primer 6153: S' CCGGCGGATCCCATATGTCTTACAACCACCTGCAGG
3' (SEQ ID N0:93) Primer 61 S0: 5' CCGGCGGTACCTTATTATGAGTGTACCACCATTGG 3' (SEQ ID N0:94) In the case of the N-terminal primer (6153), an NdeI restriction site was incorporated, while in the case of the C-terminal primer (6150) an Asp718 restriction site was incorporated. Primer 6153 also contains an ATG sequence adjacent and in frame with the KGF2 coding region to allow translation of the cloned fragment in E.coli, while primer 6150 contains two stop codons (preferentially utilized in E.coli) adjacent and in frame with the KGF2 coding region which ensures correct translational termination in E.coli.
The Polymerise Chain Reaction was performed using standard conditions well known to those skilled in the art and the nucleotide sequence for the mature KGF-2 (aa 36-208) (constructed in Example lOC) as template. The resulting amplicon was restriction digested with NdeI and Asp718 and subcloned into NdeI/Asp718 digested pHEl protein expression vector.
Nucleotide sequence of KGF-2 A33:
ATGTCTTACAACCACCTGCAGGGTGACGTTCGTTGGCGTAAACTGT
TCTCTTTCACCAAATACTTCCTGAAAATCGAAAA
AAACGGTAAAGTTTCTGGGACCAAGAAGGAGAACTGCCCGTACAG
CATCCTGGAGATAACATCAGTAGAAATCGGAGTTG
TTGCCGTCAAAGCCATTAACAGCAACTATTACTTAGCCATGAACAA
GAAGGGGAAACTCTATGGCTCAA.AAGAATTTAAC

AATGACTGTAAGCTGAAGGAGAGGATAGAGGAAAATGGATACAAT
ACCTATGCATCATTTAACTGGCAGCATAATGGGAG
GCAAATGTATGTGGCATTGAATGGAAAAGGAGCTCCAAGG
AGAGGACAGAA.AACACGAAGGAAAAACACCTCTGCTCACTTTCTT
CCAATGGTGGTACACTCATAA (SEQ ID N0:95) Amino Acid sequence of KGF-2 X33:
MSYNHLQGDVRWRKLFSFTKYFLKIEKNGKVSGTKKENCPYSILEITS
VEIGV VAVKAINSNYYLAMNKKGKLYGSKEFN
NDCKLKERIEENGYNTYASFNWQHNGRQMYVALNGKGAPRRGQKTR
RKNTSAHFLPMVVHS (SEQ ID N0:96) B. Construction of an Optimized KGF 2433 In order to increase the expression levels of KGF2 X33 in E.coli, the codons of the complete gene were optimized to match those most highly used in E.coli. As the template utilised to generate the KGF2 033 was codon optimized within the N-terminal region, the C-terminal amino acids (84-208) required optimization.
Firstly, amino acids 172-208 were codon optimized to generate KGF2~33(s172-208). This was achieved by an overlapping PCR strategy.
Oligonucleotides PM07 and PMO8 (corresponding to amino acids 172-208) were combined and annealed together by heating them to 70 °C and allowing them to cool to 37°C. The annealed oligonucleotides were then utilized as template for a standard PCR reaction which was directed by primers PM09 and PM10. In a separate PCR reaction following standard conditions well known to those skilled in the art and using KGF2~33 as template, oligonucleotides PMOS (which overlaps with the Pstl site within coding region of KGF2) and PM11 were used to amplify the region of KGF2 corresponding to amino acids 84-172. In a third PCR reaction, the product of the first PCR reaction (corresponding to codon optimized amino acids 172-208) and the product of the second PCR reaction (corresponding to codon non-optimized amino acids 84-172) were combined and used as template for a standard PCR reaction directed by oligonucleotides PMOS
and PM 10. The resulting amplicon was digested with Pst 1/HindIII and sub-cloned into PstI/HindIII digested pQE6KGF2~33, effectively substituting the corresponding non colon optimized region, and creating pQE6KGF2~33(s172-208).
To complete the colon optimization of KGF2, a synthetic gene colon optimized for the region of KGF2 corresponding to amino acids 84-172 was generated utilising overlapping oligonucleotides. Firstly, four oligonucleotides (PM31, PM32, PM33 and PM 34) were combined and seven cycles of the following PCR was performed: 94 °C, 30 secs; 46.5 ° C, 30 secs;
and 72 °C, 30 secs.
A second PCR reaction directed by primers PM35 and PM36 was then performed following standard procedures, utilizing 1 p.l of the first PCR
reaction as template. The resulting colon optimized gene fragment was then digested with 1 S Pst l/Sal l and subcloned into Pst 1 /Sal l digested pQE6KGF2t133(s 172-208) to create a fully optimized KGF2 encoding gene, pQE6KGF2~33s. ' To create an alternative E.coli protein expression vector, KGF2A33s was PCR amplified utilising primers PM102 and PM130 on pQE6KGF2~33s. The resulting amplicon was digested with NdeI and EcoRV and subcloned into the pHEl expression vector which had been digested with NdeI and Asp718 (blunt ended) to create pHE 1 ~3 3 s.
Oligonucleotide Sequences used in construction of colon optimized KGF2 ~33s:
PMOS: CAACCACCTGCAGGGTGACG (SEQ ID N0:97) PM07:AACGGTCGACAAATGTATGTGGCACTGAACGGTAAAGGTG
CTCCACGTCGTGGTCAGAAAACCCGTCGTAAAAACACC (SEQ ID
N0:98) PM08:
GGGCCCAAGCTTAAGAGTGTACCACCATTGGCAGAAAGTGAGCAG
AGGTGTTTT"TACGACGGGTTTTCTGACCACG (SEQ ID N0:99) PM09: GCCACATACATTTGTCGACCGTT (SEQ ID NO:100) PM10: GGGCCCAAGCTTAAGAGTG (SEQ ID NO:101) PM11:GCCACATACATTTGTCGACCGTT (SEQ ID N0:102) PM31:
CTGCAGGGTGACGTTCGTTGGCGTAAACTGTTCTCCTTCACCAAAT
ACTTCCTGAAAATCGA~~.AAAAACGGTAAAGTTTC
TGGTACCAAG (SEQ ID N0:103) PM32:
AGCTTTAACAGCAACAACACCGATTTCAACGGAGGTGATTTCCAGG
ATGGAGTACGGGCAGTTTTCTTTCTTGGTACCAG
AAACTTTACC (SEQ ID N0:104) PM33:
GGTGTTGTTGCTGTTAAAGCTATCAACTCCAACTACTACCTGGCTAT
GAACAAGAAAGGTAAACTGTACGGTTCCAAAGA
ATTTAACAAC (SEQ ID NO:105) PM34:
GTCGACCGTTGTGCTGCCAGTTGAAGGAAGCGTAGGTGTTGTAACC
GTTTTCTTCGATACGTTCTTTCAGTTTACAGTCG
TTGTTAAATTCTTTGGAACC (SEQ ID N0:106) PM35: GCGGCGTCGACCGTTGTGCTGCCAG (SEQ ID N0:107) PM36: GCGGCCTGCAGGGTGACGTTCGTTGG (SEQ ID N0:108) PM102: CCGGCGGATCCCATATGTCTTACAACCACCTGCAGG (SEQ ID
N0:109) PM130: CGCGCGATATCTTATTAAGAGTGTACCACCATTG (SEQ ID
NO:1 I 0) Nucleotide sequence of KGF-2 ~33(s172-208):
ATGTCTTACAACCACCTGCAGGGTGACGTTCGTTGGCGTAAACTGT
TCTCCTTCACCAAATACTTCCTGAAAATCGAAAA
AAACGGTAAAGTTTCTGGTACCAAGAAAGAAAACTGCCCGTACTCC
ATCCTGGAAATCACCTCCGTTGAAATCGGTGTTG
TTGCTGTTAAAGCTATCAACTCCAACTACTACCTGGCTATGAACAA

GAAAGGTAAACTGTACGGTTCCAAAGAATTTAAC
AACGACTGTAAACTGAAAGAACGTATCGAAGAAAACGGTTACAAC
ACCTACGCTTCCTTCAACTGGCAGCACAACGGTCG
ACAAATGTATGTGGCACTGAACGGTAAAGGTGCTCCACGTCGTGGT
CAGAAAACCCGTCGTAAAAACACCTCTGCTCACTTTCTGCC
AATGGTGGTACACTCTTAA (SEQ ID NO:111) Amino Acid Sequence of KGF-2 ~33(s172-208):
MSYNHLQGDVRWRKLFSFTKYFLKIEKNGKVSGTKKENCPYSILEITS
VEIGVVAVKAINSNYYLAMNKICGKLYGSKEFNNDCKLKERIEENGYN
TYASFNWQHNGRQMYVALNGKGAPRRGQKTRRKNTSAHFLPNNVHS
(SEQ ID N0:112) C. Construction of N terminal deletion mutant KGF 2d4 To increase the level of expression of KGF2 in E.coli and to enhance the stability and solubility properties of E.coli expressed KGF2, a deletion variant KGF204 (amino acids 39-208) which removes the first 38 amino acids of pre-processed KGF2 was constructed, including the cysteine at position 37. As the resulting KGF2 deletion molecule contains an even number of cysteines, problems due to aggregation caused by infra-molecular disulphide bridge formation should be reduced, resulting in an enhanced level of expresssion of active protein.
To permit Polymerase Chain Reaction directed amplification and sub-cloning of KGF2 t14 into the E.coli protein expression vector, pQE6, two oligonucleotide primers (PM61 and 19138) were synthesized with the following base sequence.
PM61: CGCGGCCATGGCTCTGGGTCAGGACATG (SEQ ID N0:113) 19138: GGGCCCAAGCTTATGAGTGTACCACCAT (SEQ ID N0:114) In the case of the N-terminal primer (PM61 ), an NcoI restriction site was incorporated, while in the case of the C-terminal primer (19138) a HindIII
restriction site was incorporated. PM61 also contains an ATG sequence adjacent and in frame with the KGF2 coding region to allow translation of the cloned fragment in E.coli, while 19138 contains a stop codon (preferentially utilized in E.coli) adjacent to and in frame with the KGF2 coding region which ensures correct translational termination in E.coli.
The Polymerase Chain Reaction was performed using standard conditions well known to those skilled in the art and the full length KGF2 (aa 36-208) as template (constructed in Example l OC). The resulting amplicon was restriction digested with NcoI and HindIII and subcloned into NcoI/HindIII digested pQE6 protein expression vector.
Nucleotide Sequence of KGF-2 04:
ATGGCTCTGGGTCAAGATATGGTTTCTCCGGAAGCTACCAACTCTT
CCTCTTCCTCTTTCTCTTCCCCGTCTTCCGCTGG
TCGTCACGTTCGTTCTTACAACCACCTGCAGGGTGACGTTCGTTGGC
GTAAACTGTTCTCTTTCACCAAATACTTCCTGA
AAATCGAAAAAAACGGTAAAGTTTCTGGGACCAAGAAGGAGAACT
GCCCGTACAGCATCCTGGAGATAACATCAGTAGAA
ATCGGAGTTGTTGCCGTCAAAGCCATTAACAGCAACTATTACTTAG
CCATGAACAAGAAGGGGAAACTCTATGGCTCAAA
AGAATTTAACAATGACTGTAAGCTGAAGGAGAGGATAGAGGAAAA
TGGATACAATACCTATGCATCATTTAACTGGCAGC
ATAATGGGAGGCAAATGTATGTGGCATTGAATGGAAAAGGAGCTC
CAAGGAGAGGACAGAAAACACGAAGGAAAAACACC
TCTGCTCACTTTCTTCCAATGGTGGTACACTCATAA (SEQ ID NO:11 S) Amino Acid Sequence of KGF-2~4:
MALGQDMVSPEATNSSSSSFSSPSSAGRHVRSYNHLQGDVRVVRKLFSF
TKYFLKIEKNGKVSGTKKENCPYSILEITSVEIGVVAVKAINSNYYLAM

NKKGKLYGSKEFNNDCKLKERIEENGYNTYASFNWQHNGRQMYVAL
NGKGAPRRGQKTRRKNTSAHFLPMVVHS (SEQ ID NO:I 16) Example 1 S
Construction of Carboxy Terminal Mutations in KGF 2 The carboxyl terminus of KGF-2 is highly charged. The density of these charged residues may affect the stability and consequently the solubility of the protein. To produce muteins that might stabilize the protein in solution a series of mutations were created in this region of the gene.
To create point mutants 194 R/E, 194 R/Q, 191 K/E, 191 K/Q, 188R/E, 188R/Q, the 5952 KGF033 5' Afl III 5' primer was used with the indicated 3' primers, which contain the appropriate point mutations for KGF-2, in PCR
reactions using standard conditions well known to those skilled in the art with KGF-2033 as template. The resulting products were restricted with AflIII and Hind III and cloned into the E. coli expression vector, pQE60 restricted with NcoI
and Hind III.
KGF-2A33,194 It/E Construction:
The following primers were used:
5952 KGF D 33 5' Afl III:
5' GCGGCACATGTCTTACAACCACCTGCAGGGTG 3' (SEQ ID NO:I 17) KGF2 3'HindIII 194aa R to E:
5'CTGCCCAAGCTTTTATGAGTGTACCACCATTGGAAGAAAGTGAGC
AGAGGTGTTTI"TTTCTCGTGTTTTCTGTCC 3' (SEQ ID N0:118) KGF-2033,194 R/E Nucleotide sequence:
ATGTCTTACAACCACCTGCAGGGTGACGTTCGTTGGCGTAAACTGT
TCTCTTTCACCAAATACTTCCTGAAAATCGAAAA
AAACGGTAAAGTTTCTGGGACCAAGAAGGAGAACTGCCCGTACAG

CATCCTGGAGATAACATCAGTAGAAATCGGAGTTG
TTGCCGTCAAAGCCATTAACAGCAACTATTACTTAGCCATGAACAA
GAAGGGGAAACTCTATGGCTCAAAAGAATTTAAC
AATGACTGTAAGCTGAAGGAGAGGATAGAGGAAAATGGATACAAT
ACCTATGCATCATTTAACTGGCAGCATAATGGGAG
GCAAATGTATGTGGCATTGAATGGAAAAGGAGCTCCAAGGAGAGG
ACAGAAAACACGAGAAAAAAACACCTCTGCTCACT
TTCTTCCAATGGTGGTACACTCATAG (SEQ ID N0:119) KGF-2A33,194 R/E Amino acid sequence:
MSYNHLQGDVRWRKLFSFTKYFLKIEKNGKVSGTKKENCPYSILEITS
VEIGVVAVKAINSNYYLAMNKKGKLYGSKEFNNDCKLKERIEENGYN
TYASFNWQHNGRQMYVALNGKGAPRRGQKTREKNTSAHFLPMWHS
(SEQ ID N0:120) KGF-2 X33,194 R/Q Construction:
The following primers were used:
5952 KGF 033 5' Afl III:
5' GCGGCACATGTCTTACAACCACCTGCAGGGTG 3' (SEQ ID N0:121 ) KGF2 3' HindIII 194 as R to Q:
5'CTGCCCAAGCTTTTATGAGTGTACCACCATTGGAAGAAAGTGAGC
AGAGGTGTTTT'fC~GTCGTGTTTTCTGTCC 3' (SEQ ID N0:122) KGF-21133,194 RIQ Nucleotide Sequence:
ATGTCTTACAACCACCTGCAGGGTGACGTTCGTTGGCGTAAACTGT
TCTCTTTCACCAAATACTTCCTGAAAATCGAAAA
AAACGGTAAAGTTTCTGGGACCAAGAAGGAGAACTGCCCGTACAG
CATCCTGGAGATAACATCAGTAGAAATCGGAGTTG
TTGCCGTCAAAGCCATTAACAGCAACTATTACTTAGCCATGAACAA
GAAGGGGAAACTCTATGGCTCAAAAGAATTTAAC
AATGACTGTAAGCTGAAGGAGAGGATAGAGGAAAATGGATACAAT
ACCTATGCATCATTTAACTGGCAGCATAATGGGAG

GCAAATGTATGTGGCATTGAATGGAAAAGGAGCTCCAAGGAGAGG
ACAGAAAACACGAC~GAAAAACACCTCTGCTCACT
TTCTTCCAATGGTGGTACACTCATAG (SEQ ID N0:123) KGF-2 A33,194 R/Q Amino Acid Sequence:
MSYNHLQGDVRWRKLFSFTKYFLKIEKNGKVSGTKKENCPYSILEITS
VEIGV VAVKAINSNYYLAMNKKGKLYGSKEFNNDCKLKERIEENGYN
TYASFNWQHNGRQMYVALNGKGAPRRGQKTRQKNTSAHFLPMWHS
{SEQ ID N0:124) KGF-2433,191 K/E Construction:
The following primers were used:
5952 KGF 0 33 5' Afl III:
5' GCGGCACATGTCTTACAACCACCTGCAGGGTG 3' (SEQ ID N0:125) KGF2 3' HindIII 191 as K to E
5'CTGCCCAAGCTTTTATGAGTGTACCACCATTGGAAGAAAGTGAGC
AGAGGTGTI"I"I"TCCTTCGTGTTTCCTGTCCTCTCCTTGG 3' (SEQ ID
N0:126) KGF-233,191 K/E Nucleotide Sequence:
ATGTCTTACAACCACCTGCAGGGTGACGTTCGTTGGCGTAAACTGT
TCTCTTTCACCAAATACTTCCTGAAAATCGAAAA
AAACGGTAAAGTTTCTGGGACCAAGAAGGAGAACTGCCCGTACAG
CATCCTGGAGATAACATCAGTAGAAATCGGAGTTG
TTGCCGTCAAAGCCATTAACAGCAACTATTACTTAGCCATGAACAA
GAAGGGGAAACTCTATGGCTCAAAAGAATTTAAC
AATGACTGTAAGCTGAAGGAGAGGATAGAGGAAAATGGATACAAT
ACCTATGCATCATTTAACTGGCAGCATAATGGGAG
GCAAATGTATGTGGCATTGAATGGAAAAGGAGCTCCAAGGAGAGG
ACAGG~AACACGAAGGAA.AAACACCTCTGCTCACT
TTCTTCCAATGGTGGTACACTCATAG (SEQ ID N0:127) KGF-233,191 K/E Amino Acid Sequence:

WO 99l4I28Z PCTIUS99/03018 MSYNHLQGDVRWRKLFSFTKYFLKIEKNGKVSGTKKENCPYSILEITS
VEIGV VAVKAINSNYYLAMNKKGKLYGSKEFNNDCKLKERIEENGYN
TYASFNWQHNGRQMYVALNGKGAPRRGQETRRKNTSAHFLPMVVHS
(SEQ ID N0:128) KGF-2 X33,191 K/Q Construction:
The following primers were used:
5952 KGF033 5' Afl III:
5' GCGGCACATGTCTTACAACCACCTGCAGGGTG 3' (SEQ ID N0:129) KGF2 3' HindIII 191aa K to Q
5'CTGCCCAAGCTTTTATGAGTGTACCACCATTGGAAGAAAGTGAGC
AGAGGTGT'I"ITTCCTTCGTGTCTGCTGTCCTCTCCTTGG 3' (SEQ ID
N0:130) KGF-2 A33,191 K/Q Nucleotide Sequence:
ATGTCTTACAACCACCTGCAGGGTGACGTTCGTTGGCGTAAACTGT
TCTCTTTCACCAAATACTTCCTGAAAATCGAAAA
AAACGGTAAAGTTTCTGGGACCAAGAAGGAGAACTGCCCGTACAG
CATCCTGGAGATAACATCAGTAGAAATCGGAGTTG
TTGCCGTCAAAGCCATTAACAGCAACTATTACTTAGCCATGAACAA
GAAGGGGAAACTCTATGGCTCAAAAGAATTTAAC
AATGACTGTAAGCTGAAGGAGAGGATAGAGGAAAATGGATACAAT
ACCTATGCATCATTTAACTGGCAGCATAATGGGAG
GCAAATGTATGTGGCATTGAATGGAAAAGGAGCTCCAAGGAGAGG
ACAGCAGACACGAAGGAAAAACACCTCTGCTCACT
TTCTTCCAATGGTGGTACACTCATAG (SEQ ID N0:131) KGF-2 A33,191 K/Q Amino Acid Sequence:
MSYNHLQGDVRWRKLFSFTKYFLKIEKNGKVSGTKKENCPYSILEITS
VEIGV VAVKAINSNYYLAMNKKGKLYGSKEFNNDCKLKERIEENGYN
TYASFNWQHNGRQMYVALNGKGAPRRGQQTRRKNTSAHFLPMVVHS
(SEQ ID N0:132) KGF-2A33,188Lt/E Construction:
The following primers were used:
5952 KGF~33 5' Afl III:
5' GCGGCACATGTCTTACAACCACCTGCAGGGTG 3' (SEQ ID N0:133) KGF2 3' HindIII 188aa R to E:
5'CTGCCCAAGCTTTTATGAGTGTACCACCATTGGAAGAAAGTGAGC
AGAGGTGTTTTTCCTTCGTGTTTTCTGTCC1'~C_CCTTGGAGCTCCTTT
3' (SEQ ID N0:134) KGF-2~33,188It/E Nucleotide Sequence:
ATGTCTTACAACCACCTGCAGGGTGACGTTCGTTGGCGTAAACTGT
TCTCTTTCACCAAATACTTCCTGAAAATCGAAAA
AAACGGTAAAGTTTCTGGGACCAAGAAGGAGAACTGCCCGTACAG
CATCCTGGAGATAACATCAGTAGAAATCGGAGTTG
TTGCCGTCAAAGCCATTAACAGCAACTATTACTTAGCCATGAACAA
GAAGGGGAAACTCTATGGCTCAAAAGAATTTAAC -AATGACTGTAAGCTGAAGGAGAGGATAGAGGAAA.ATGGATACAAT
ACCTATGCATCATTTAACTGGCAGCATAATGGGAG
GCAAATGTATGTGGCATTGAATGGAAAAGGAGCTCCAAGGG~A GG
ACAGAAAACACGAAGGAAAAACACCTCTGCTCACT
TTCTTCCAATGGTGGTACACTCATAG (SEQ ID N0:135) KGF-2L~33,188R/E Amino Acid Sequence:
MYNHLQGDVRWRKLFSFTKYFLKIEKNGKVSGTKKENCPYSILEITSV
EIGVVAVKAINSNYYLAMNKKGKLYGSKEFNNDCKLKERIEENGYNT
YASFNWQHNGRQMYVALNGKGAPR_EGQKTRRKNTSAHFLPMVVHS
(SEQ ID N0:136) KGF-233,188 lt/Q Construction:
The following primers were used:

5952 KGF 033 S' Afl III:
5' GCGGCACATGTCTTACAACCACCTGCAGGGTG 3' (SEQ ID N0:137) KGF2 3' HindIII 188aa R to Q:
5'CTGCCCAAGCTTTTATGAGTGTACCACCATTGGAAGAAAGTGAGC
AGAGGTGTTT"TTCCTTCGTGTTTTCTGTCCC~CCTTGGAGCTCCTTT
3' (SEQ ID N0:138) KGF-233,188 R/Q Nucleotide Sequence:
ATGTCTTACAACCACCTGCAGGGTGACGTTCGTTGGCGTAAACTGT
TCTCTTTCACCAAATACTTCCTGAAAATCGAAAA
AAACGGTAAAGTTTCTGGGACCAAGAAGGAGAACTGCCCGTACAG
CATCCTGGAGATAACATCAGTAGAAATCGGAGTTG
TTGCCGTCAAAGCCATTAACAGCAACTATTACTTAGCCATGAACAA
GAAGGGGAAACTCTATGGCTCAAAAGAATTTAAC
AATGACTGTAAGCTGAAGGAGAGGATAGAGGAAAATGGATACAAT
ACCTATGCATCATTTAACTGGCAGCATAATGGGAG
GCAAATGTATGTGGCATTGAATGGAAAAGGAGCTCCAAGGCAGGG
ACAGAAAACACGAAGGAAAAACACCTCTGCTCACT
TTCTTCCAATGGTGGTACACTCATAG (SEQ ID N0:139) KGF-233,188 R/Q Amino Acid Sequence:
MSYNHLQGDVRWRKLFSFTKYFLKIEKNGKVSGTKKENCPYSILEITS
VEIGVVAVKAINSNYYLAMNKKGKLYGSKEFNNDCKLKERIEENGYN
TYASFNWQHNGRQMYVALNGKGAPRQGQKTRRKNTSAHFLPMWHS
(SEQ ID N0:140) KGF-2 ~33,183K/E Construction:
For mutation 183K/E , two PCR reactions were set up for oligonucleotide site directed mutagenesis of this lysine. In one reaction, 5952 KGFA 33 5' AflIII
was used as the 5' primer, and KGF2 183aa K to E antisense was used as the 3' primer in the reaction. In a second reaction, KGF2 5' 183aa K to E sense was used as the 5' primer, and KGF2 3' HindIII TAA stop was used as the 3' primer.
KGF-2 033 was used as template for these reactions. The reactions were amplified under standard conditions well known to those skilled in the art.
One microliter from each of these PCR reactions was used as template in a subsequent reaction using, as a 5' primer, 5453 BsphI, and as a 3' primer, 5258 HindIII.
Amplification was performed using standard conditions well known to those skilled in the art. The resulting product was restricted with Afl III and HindIII
and cloned into the E. coli expression vector pQE60, which was restricted with NcoI and HindIII.
The following primers were used:
5952 KGF X33 5' Afl III:
5' GCGGCACATGTCTTACAACCACCTGCAGGGTG 3' (SEQ ID N0:141 ) KGF2 5' 183aa K to E sense:
5' TTGAATGGAGAAGGAGCTCCA 3' (SEQ ID N0:142) KGF2 183aa K to E antisense:
5' TGGAGCTCC~TCCATTCAA 3' (SEQ ID N0:143) KGF2 3' HindIII TAA stop:
5' CTGCCCAAGCTTTTATGAGTGTACCACCATTGG 3' (SEQ ID N0:144) KGF-2 033,183K/E Nucleotide Sequence:
ATGTCTTACAACCACCTGCAGGGTGACGTTCGTTGGCGTAAACTGT
TCTCTTTCACCAAATACTTCCTGAAAATCGAAAA
AAACGGTAAAGTTTCTGGGACCAAGAAGGAGAACTGCCCGTACAG
CATCCTGGAGATAACATCAGTAGAAATCGGAGTTG
TTGCCGTCAAAGCCATTAACAGCAACTATTACTTAGCCATGAACAA
GAAGGGGAAACTCTATGGCTCAA.AAGAATTT,AAC
AATGACTGTAAGCTGAAGGAGAGGATAGAGGAAAATGGATACAAT
ACCTATGCATCATTTAACTGGCAGCATAATGGGAG
GCAAATGTATGTGGCATTGAATGGAG~GGAGCTCCAAGGAGAGG
ACAGAAAACACGAAGGAAAAACACCTCTGCTCACT

TTCTTCCAATGGTGGTACACTCATAG (SEQ ID N0:145) KGF-2 A33,183K/E Amino Acid Sequence:
MSYNHLQGDVRWRKLFSFTKYFLKIEKNGKVSGTKKENCPYSILEITS
VEIGVVAVKAINSNYYLAMNKKGKLYGSKEFNNDCKLKERIEENGYN
TYASFNWQHNGRQMYVALNGEGAPRRGQKTRRKNTSAHFLPMVVHS
(SEQ ID N0:146) Example 16 E, ffects of KGF 2 dad on Normal Urinary Bladder and Prostate and in Cyclophosphamide Induced Hemorrhagic Cystitis in Rats This example demonstrates that KGF-2 X33 is capable of stimulating urinary bladder proliferation in normal rats and that there is a therapeutic effect of KGF-2 033 in a rat model of cyclophosphamide-induced hemorrhagic cystitis.
Some cytotoxic agents used clinically have side effects resulting in the inhibition of the proliferation of the normal epithelium in the bladder, leading to potentially life-threatening ulceration and breakdown in the epithelial lining ofthe bladder. For example, cyclophosphamide causes hemorrhagic cystitis in some patients, a complication which can be severe and in some cases fatal. Fibrosis of the urinary bladder may also develop with or without cystitis. This injury is thought to be caused by cyclophosphamide metabolites excreted in the urine.
Hematuria caused by cyclophosphamide usually is present for several days, but may persist. In severe cases medical or surgical treatment is required.
Instances of severe hemorrhagic cystitis result in discontinued cyclophosphamide therapy.
In addition, urinary bladder malignancies generally occur within two years of cyclophosphamide treatment and occurs in patients who previously had hemorrhagic cystitis (CYTOXAN (cyclophosphamide) package insert).
Cyclophosphamide has toxic effects on the prostate and male reproductive systems. Cyclophvsphamide treatment can result in the development of sterility, and result in some degree of testicular atrophy.

Effects oJKGF-2 d33 on Normal Bladder,Testes and Prostate F.xperinrental Design Male Sprague-Dawley rats (160-220 g), (n=~4 to 6/treatment group) were used in these studies. KGF-2 a33 was administered at a dose of S mg/kg/day.
S Daily ip or sc injections of recombinant KGF-2 033 or buffer {40 mM sodium acetate + 1 S 0 mM NaCI at pH 6.S) were administered for a period of I -7 days and the rats were sacrificed the following day. To examine the reversibility of effects induced with KGF-2 033, additional animals were injected ip daily for 7 days with KGF-2 X33 or buffer and sacrificed after a 7 day treatment-free period.
On the day of sacrifice, rats were injected ip with 100 mg/kg of BrdU.
Two hours later the rats were overdosed with ether and selected organs removed.
Samples of tissues were fixed in 10% neutral buffered formalin for 24 hours and paraffin embedded. To detect BrdU incorporation into replicating cells, five micron sections were subjected to immunohistochemical procedures using a 1 S mouse anti-BrdU monoclonal antibody and the ABC Elite detection system.
The sections were lightly counterstained with hematoxylin.
Sections were read by blinded observers. The number of proliferating cells was counted in 10 random fields per animal at a l Ox magnification for the prostate. To assess the effects of KGF-2 X33 in the bladder, cross-sections of these tissues were prepared and the number of proliferating and non-proliferating cells were counted in ten random fields at 20x magnification. The results are expressed as the percentage of labeled to unlabeled cells. Data are presented as mean + SEM. Statistical analyses (two-tailed unpaired t_test) were performed with the StatView Software Package and statistical significance is defined as p 2S < O.OS.
Results Bladder Intraperitoneal injection of KGF-21133 induced proliferation of bladder epithelial cells over the 7 day study period (solid squares, Figure 23) but this did not influence the weight of the organ. Subcutaneous administration elicited a small increase in proliferation but this failed to achieve statistical significance (solid circles, Figure 23).
Prostate and Testes Both sc and ip administration of KGF-2 033 induced significant proliferation of the prostate (Figure 24) but this normalized after two injections.
Prolonged ip treatment with KGF-2 033 did not increase the weight of the prostate or testes.
Effects of KGF 2 d33 on Cyclophosphamide Induced Hemorrhagic Cystitis Experimental Design Male Sprague Dawley rats (300-400g) (n=5/group) were injected i.v. via the tail vein with buffer placebo or KGF-2 A33 at concentrations of 1 or 5 mg/kg 24 hours prior to a 200 mg/kg i.p. injection of cyclophosphamide. On the final day, 48 hours after cyclophosphamide injection, rats were injected ip with 100 mg/kg of BrdU. Two hours later the rats were killed by COZ administration.
I S Fixation of the bladder was done by direct injection of 10% formalin into the lumen of the bladder and rinsing of the exterior of the bladder with formalin.
Ai~er 5 minutes, the bladder and prostate were removed. The urinary bladder and prostate gland were paraffin embedded, cross-sectioned and stained with H&E
and a mouse anti-BrdU monoclonal antibody. The extent of urothelial damage was assessed using the following scoring system: Bladders were graded by two independent observers to describe the extent of the loss of urothelium.
(Urothelial damage was scored as 0, 25%, 50%, 75% and 100% loss of the urothelium). In addition, the thickness of the bladder wall was measured at Z
O
random sites per section and expressed in pm.
Results Macroscopic Observations In rats treated with placebo and cyclophosphamide, bladders were thick and rigid. Upon injection of 10% formalin, very little expansion of the bladders was noted. However, in the groups pretreated with KGF-2 033, a greater elasticity of the bladder was noted upon direct injection with formalin suggesting a lesser degree of fibrosis.
Microscopic Observations Bladder Figure 25 shows the results of KGF-2 033 pretreatment on the extent of ulceration in the bladder. In normal rats treated with i.p. saline (saline control), the bladders appeared normal histologically and no ulceration of the urothelium was observed. Administration of 200 mg/kg i.p. of cyclophosphamide resulted in ulceration of the bladder epithelium that was between 25 and 50% of the total epithelial area (with a mean of 37%). Administration of KGF-2 X33 24 hours prior to cyclophosphamide resulted in a significant reduction in the extent of ulceration (I mg/kg 0.4% p~.0128, and 5 mg/kg 5%, p~.0338%) when compared to placebo treated animals receiving cyclophosphamide.
I 5 Figure 26 shows the effects of KGF-2 X33 on the thickness of the urinary bladder wall which includes epitheliwn, smooth muscle layers and the serosal surface. In groups treated with buffer alone, the thickness of the bladder wall is approximately 40 pm. Treatment with cyclophosphamide results in a 5 fold increase in bladder wall thickness to 210 pm. KGF-2 X33 pretreatment of cyclophosphamide treated animals resulted in a significant inhibition of cyclophosphamide enlargement of the bladder wall ( 1 mg/kg 98.6 p,m (p=0.007) and at 5 mg/kg 52.3 ~m (p<0.0001 )) when compared to the cyclophosphamide treatrnent alone.
Prostate Gland In rats receiving buffer and cyclophosphamide, marked atrophy of the prostatic glands (acini) was observed accompanied by enlargement of interstitial spaces with remarkable edema when compared to normals. In addition, epithelial cells lining the prostatic glands were observed to be much shorter and less dense than in corresponding normal prostatic tissue. KGF-2 X33 pretreatment at both _ 144-1 mglkg and S mg/kg displayed a normal histological appearance of the prostatic gland. No increase in the interstitial spaces or edema was observed, and the epithelial cells lining the prostatic glands were similar in size and density to normal prostatic tissue.
Conclusion The results demonstrate that KGF-2 specifically induces proliferation of bladder epithelial cells and the epithelial cells lining the prostatic glands.
The results also demostrate that KGF-2 specifically results in a significant reduction in the extent of ulceration in cyclophosphamide-induced hemorrhagic cystitis.
l0 Example 17 Effects of gGF 2 d33 on Platelet Levels in Rats This experiment demonstrates that KGF-233 increases platelet levels.
Experimental Design Adult Sprague-Dawley rats were treated daily with a sc injection of buffer 15 or doses of KGF-2033 (as shown in the Table below) for a period of 4 weeks.
One set of animals was sacrificed at this time. Another set was sacrificed after a 4 week recovery period. Blood samples were taken by orbital sinus puncture after an overnight fast and collected in tubes containing EDTA anticoagulant or 3.13% w/v aqueous trisodium citrate anticoagulant for hematology and 20 coagulation assays, respectively. Blood chemistry was performed on samples taken into lithium heparin anticoagulant.
Results As shown in the Table below, significant increases in platelet levels were demonstrated in both male and female rats at 0.3 mg/kg (p<0.05), 1 mglkg 25 (p<0.01 ), 3 mg/kg (p<0.01 ), and 10 mg/kg (p<0.001 ) KGF-233 treatrnent relative to buffer controls. In most instances these effects were reversible after a 4-week recovery period.
Effect of KGF-2033 on platelet levels in rats Treatment Sex After 4 weeks After 4 weeks of of treatment recovery Platelet 0' / Platelet 10' B~~' Male 1166 t 116 1083 t 69 0.3 m Male 1277 t 96* 1048 ~ 116 1 m Male 1364 f 166** 1124 ~ 101 3 m Male 1327 t 74** 1129 ~ 57 10 m Male 1465 t 152*** 124$ f 62*

Buffer Female 1125 f 182 1078 f 123 0.3 m Female 1243 t 145 * 1091 f 106 1 m Female 1328 t 135** 1084 ~ 98 3 m Female 1339 f 116** 1151 ~ 177 10 m Female 1533 ~ 157*** 1233 ~ 146 1$ * nCIInS **.,~nn~ ***_,.n.",.

a ____, 1. -,..,..~ 1, ~v.vm Example l8 E, f,~'ects of KGF 2 d 33 on Plasma Fibrinogen in Rats This experiment demonstrates that KGF-2033 increases plasma fibrinogen levels.

Fxperimentarl Design Adult Sprague-Dawley rats were treated daily with a sc injection of buffer or doses of KGF-233 (as shown in the Table below) for a period of 4 weeks.
One set of animals was sacrificed at this time. Another set was sacrificed after a 4 week recovery period. Blood samples were taken by orbital sinus puncture after an overnight fast and collected in tubes containing EDTA anticoagulant or 3.23% w/v aqueous trisodium citrate anticoagulant for hematology and coagulation assays, respectively. Blood chemistry was performed on samples taken into lithium heparin anticoagulant.
Results As shown in the Table below, significant increases in plasma fibrinogen levels were demonstrated in male rats at 3 mg/kg (p<0,0p1) and 10 mg/kg (p<0.001) KGF-233 treatment relative to buffer controls, and in female rats at 1 mg/kg (p<0,001 ), 3 mg/kg (p<p.001 ), and 10 mg/kg (p<0.001 ) KGF-233 treatment relative to buffer controls. In most instances these effects were reversible after a 4-week recovery period.
Effect of KGF-233 on Plasma Fibrinogen Levels in Rats Treatment Sex After 4 weeks After 4 weeks of of treatment recovery Fibrino en m Fibrino en m dl dl Buffer Male 248 t 24 214 t 14 0.3 m Male 252 f 23 202 f 14 I m Male 287 t 49* 219 t 10 3 m Male 315 t 24*** 220 f 10 10 m Male 378 t 46*** 222 t 21 Treatment Sex After 4 weeks After 4 weeks of of Buffer Female treatment recovery 194 t 24 171 t 25 0.3 m Female 215 f 29 195 ~ 24 1 m Female 248 t 19* * * 191 ~ 14 3 m Female 248 ~ 23 * * 175 ~ 15 *

10 m Female 303 ~ 44*** 182 f 19 *_ , ., "~
~,~_ _ "
., ".".,, Y , ...~~, _ _ _Y ~ ~.uut Example 19 Effects oJKGF-Z d 33 on Total Serum Protein Levels in Rats This experiment demonstrates that KGF-233 increases total serum protein levels.
Experimental Design Adult Sprague-Dawley rats were treated daily with a sc injection of buffer or doses of KGF-2033 (as shown in the Table below} for a period of 4 weeks.
One set of animals was sacrificed at this time. Another set was sacrificed after 1 S a 4 week recovery period. Blood samples were taken by orbital sinus puncture after an overnight fast and collected in tubes containing EDTA anticoagulant or 3.13% w/v aqueous trisodium citrate anticoagulant for hematology and coagulation assays, respectively. Blood chemistry was performed on samples taken into lithium heparin anticoagulant.
Results As shown in the Table below, significant increases in total serum protein levels were demonstrated in male and female rats at 1 mg/kg (p<0.001 ), 3 mg/kg (p<0.001 ), and 10 mg/kg (p<0,0p 1 ) KGF-2Q33 treatment relative to buffer controls. In some instances these effects were reversible after a 4-week recovery period.
Effect of KGF-233 on Total Serum Protein Levels in Rats Treatment Sex After 4 weeks After 4 weeks of of treatment recovery Total Protein Total Protein dl dl Buffer Male 5.9 t 0.3 6.4 f 0.5 0.3m Male 6.1f0.3 6.2f0.3 1 m /k Male 6.60.2*** 6.510.2 3m Male 6.70.4*** 6.310.2 lO m Male 6.910.4*** 6.1 10.0 Buffer Female 5.9 f 0.3 6.7 t 0.2 0.3 m Female 6.1 f 0.2 6.6 t 0.1 1 m Female 6.6 ~ 0.2* * 6.5 t 0.3 *

3m Female 6.90.3*** 6.Sf0.2 10 m Female 7.0 f 0.4*** 6.3 f 0.3*

* ..~.,.,~ ~~_ . ..
_~-"

r ~~~.~, y - v.va, Y ~ V.VV1 Example 20 Effects of ~CGF 2 d 33 on Serum Albumin Levels in Rats This experiment demonstrates that KGF-233 increases serum albumin levels.

Experimental Design Adult Sprague-Dawley rats were treated daily with a sc inj ection of buffer or doses of KGF-233 {as shown in the Table below) for a period of 4 weeks.
One set of animals was sacrificed at this time. Another set was sacrificed after a 4 week recovery period. Blood samples were taken by orbital sinus puncture after an overnight fast and collected in tubes containing EDTA anticoagulant or 3.13% w/v aqueous trisodium citrate anticoagulant for hematology and coagulation assays, respectively. Blood chemistry was performed on samples taken into lithium heparin anticoagulant.
Results As shown in the Table below, significant increases in serum albumin levels were demonstrated in male rats at 1 mg/kg (p<p.01 ), 3 mg/kg (p<0.001 ), and 10 mg/kg (p<0.001 ) KGF-2033 treatment relative to buffer controls, and in female rats at 1 mg/kg (p<0.001), 3 mg/kg (p<0,001), and 10 mg/kg (p<p,001) KGF-2A33 treatment relative to buffer controls. In most instances these elects were reversible after a 4-week recovery period.
Effect of KGF-2A33 on Serum Albumin Levels in Rats Treatment Sex After 4 weeks After 4 weeks of of treatment recovery Albumin dl Albumin dl B~~' M~e 3.4 ~ 0.2 3.4 f 0.2 0.3 m Male 3.S f 0.1 3.410.1 I m Male 3.6f0.1** 3.510.0 3 m Male 3.7 t 0.2* * 3.4 f 0.2 *

10m Male 3.710.2*** 3.3f0.1 Treatment Sex After 4 weeks After 4 weeks of of treatment recovery Buffer Female 3.4 f 0.2 3.8 f 0.1 0.3 m Female 3.4 f 0.2 3.7 ~ 0.1 l m Female 3 .7 t 0.1 * 3.6 t 0.1 * *

3 m /k Female 3.9 ~ 0.2*** 3.5 t 0.1**

10m Female 3.910.2*** 3.510.2**

*_ , ., "~
,~ ~, y -v.v.r, Fr ~v.vl, wy V,VV1 Example 21 Effects of KGF-2 d33 on Serum Globulin Levels in Rats This experiment demonstrates that KGF-2033 increases serum globulin levels.
Experimental Design Adult Sprague-Dawley rats were treated daily with a sc injection of buffer or doses of KGF-233 (as shown in the Table below) for a period of 4 weeks.
One set of animals was sacrificed at this time. Another set was sacrificed after a 4 week recovery period. Blood samples were taken by orbital sinus puncture after an overnight fast and collected in tubes containing EDTA anticoagulant or 3.13% w/v aqueous trisodium citrate anticoagulant for hematology and coagulation assays, respectively. BIood chemistry was performed on samples taken into lithium heparin anticoagulant.
Results As shown in the Table below, significant increases in serum globulin levels were demonstrated in male and female rats at 1 mg/kg (p<0,001 ), 3 mg/kg (p<0.001), and 10 mg/kg (p<0,001) KGF-233 treatment relative to buffer controls. In some instances these effects were reversible after a 4-week recovery period.
Effect of KGF-233 on Serum Globulin Levels in Rata Treatment Sea After 4 weeks After 4 weeks of of ' treatment recovery Globulin dl Globulin /dl B~~ M~e 2.5f0.2 3.0f0.3 0.3 m Male 2.6 t 0.2 2.8 t 0.2 1 m Male 2.910.1*** 3.1 10.2 3m Male 3.Of0.2*** 2.9f0.1 lO m Male 3.210.2*** 2.810.1 Buffer Female 2.5 f 0.1 2.9 t 0.1 0.3 m Female 2.7 t 0.1 2.9 f 0.1 1 m Female 2.9 t 0.1 * * 2.9 t 0.2 *

3 m Female 3.0 f 0.2*** 3.0 t 0.2 10 m Female 3.1 f 0.3 * * 2.8 t 0.2 *

* n<llnS **.,~nm~rrr~_ ..."
,~

- ~ r -~~-s y - v.vvr Example 22 Effect of KGF-2 on the Proliferation of Cells in Normal Rats Introduction KGF-2, a member of the FGF family, induces proliferation of normal human and rat keratinocytes. It has approximately 57% homology to KGF-1 (a member of the FGF family). KGF-1 has been reported to induce proliferation of epithelia of many organs (Housley et al., Keratinocyte growth factor induces proliferation ofhepatocytes and epithelial cells throughout the rat gastrointestinal tract. JClin Invest 94: 1764-1777 (1994); Ulich et al., Keratinocyte growth factor is a growth factor for type II pneumocytes in vivo. JClin Invest 93: 1298-1306 (1994);Ulich et al., Keratinocyte growth factor is a growth factor for mammary epithelium in vivo. The mammary epithelium of lactating rats is resistant to the proliferative action of keratinocyte growth factor. Am J Pathol 144:862-868 (1994); Nguyen et al., Expression of keratinocyte growth factor in embryonic Iiver of transgenic mice causes changes in epithelial growth and differentiation resulting in polycystic kidneys and other organ malformations. Oncogene 12:2109-2119 ( 1996); Yi et al., Keratinocyte growth factor induces pancreatic ductal epithelial proliferation. Am J Pathol 145:80-85 (1994); and Yi et aL, Keratinocyte growth factor causes proliferation of urothelium in vivo. J
Urology IS 154:1566-1570 (1995)). We performed similar experiments with KGF-2 to determine if it induces proliferation ofnonmal epithelia in rats when administered systenucally using sc and ip routes.
Methods Male Sprague-DawIey rats, weighing 160-220 g, were obtained from Harlan Sprague Dawley for these studies. KGF-2 X33 (HG0341I-E2) was administered at a dose of 5 mg/kg/day. Daily ip or sc injections of KGF-2 X33 or recombinant buffer (40 mM sodium acetate + 150 mM NaCI at pH 6.5) were administered for a period of 1-7 days and the rats were sacrificed the following day (see below). To examine the reversibility of effects induced with KGF-2 033, additional animals were injected ip daily for 7 days with KGF-2 X33 or buffer and sacrificed after a 7 day treatment-free period.
On the day of sacrifice, rats were injected ip with 100 mg/kg of BrdU.
Two hours later the rats were overdosed with ether and selected organs removed.
In some studies organ weights were recorded. Samples of tissues were fixed in 10% neutral buffered fonnalin for 24 hours and paraffin embedded. To detect BrdU incorporation into replicating cells, five micron sections were subjected to immunohistochemical procedures using a mouse anti-BrdU monoclonal antibody (Boehringer Mannheim) and the ABC Elite detection system (Vector Laboratories). The sections were lightly counterstained with hematoxylin.
Sections were read by blinded observers. The number of proliferating cells was counted in 10 random fields per animal at a l Ox magnification for the following tissues: liver, pancreas, prostate, and heart. Ten random fields were used also for the lung analysis except the proliferation was quantitated at 20x magnification. Since the kidney has many functionally discrete areas, the proliferation was assessed in a coronal cross-section taken through the center of one kidney per animal. To assess the effects of KGF-2 X33 in the esophagus and bladder, cross-sections of these tissues were prepared and the number of proliferating and non-proliferating cells were counted in ten random fields at a lOx and 20x magnification, respectively. The results are expressed as the 1 S percentage of labeled to unlabeled cells.
Data are presented as mean t SEM. Statistical analyses (two-tailed unpaired t-test} were performed with the StatView Software Package (Abacus Concepts, Inc., Berkeley, CA) and statistical significance is defined as p <
0.05.
Results Figure 27 shows an overview of the experimental protocol. Six animals were used per group. However, during the analysis by the blinded observers it became clear that occasionally the BrdU injection was unsuccessful. Before the results were encoded, the data from 8 rats out of 116 rats (or 7% of the animals) were excluded from the study and the resultant group sizes are shown in the Table below.

Group sizes used in these studies n=
T ent Time i~p _sc KGF-2 033 1 ~y buffer 1 ~y KGF-2 X33 2 days 6 4 buffer 2 days 6 6 KGF-2 033 3 days 5 5 buffer 3 days 5 5 KGF-2 X33 7 days 6 6 buffer 7 days 6 5 KGF-2 X33 7 days + 7 days treatment-free 6 ND
buffer 7 days + '7 days treatment-free Liver When administered ip, KGF-2 X33 induced a rapid proliferation of hepatocytes (solid squares) (Figure 28) after 1 injection and this augmented mitotic activity persisted for three days, returning to normal after 7 days of daily injections. In contrast to the dramatic effect ip administration of KGF-2 exerted on the liver, when given sc (solid circle, Figure 28) this growth factor demonstrated minor effects. Proliferation was elevated after one day of treatment but returned to normal values after two daily injections.
Pancreas In contrast to the quickly reversible effects of ip administered KGF-2 X33 on the liver, such injections induced proliferation of the pancreas which continued over the 14 day study period (solid squares, Figure 29). Surprisingly, subcutaneous administration of KGF-2 X33 (solid circles) failed to induce proliferation at any time point.

wo maiasz PCT/US99/03018 Kidney and Bladder KGF-2 Q33 induced proliferation of renal epithelia when given either by the sc or ip route but the former induced a greater effect. SC administration induced a rapid increase in proliferation (solid circles) that peaked after 2 days which then returned to normal after 7 daily treatments (Figure 30). When KGF-2 A33 was given ip (solid squares, Figure 30}, there was a modest, but significant increase .in proliferation seen at days 2 and 3 only. Intraperitoneal injection of KGF-2 L133 also induced proliferation of bladder epithelial cells over the 7 day study period (solid squares, Figure 23). Subcutaneous administration elicited a small increase in proliferation but this failed to achieve statistical significance (solid circles, Figure 23}.
Prostate Both sc and ip administration of KGF-2 A33 induced significant proliferation of the prostate (Figure 24) but this normalized after two injections.
Esophagus KGF-2 X33 given sc or ip elicited an early, short-lived increase in the proliferation of the esophageal cells (I and 2 days, respectively) that rapidly returned to normal (results not shown).
Outer organs Systemic administration of KGF-2 A33 by the ip and sc routes failed to elicit proliferation of the lung epithelia over a 7 day dosing period (results not shown).
Discussion When administered in a sc route, stimulation of normal epithelial proliferation in some organs (liver, kidney, esophagus, and prostate) were observed but these effects, for the most part, were short-lived and all were reversible. The proliferation in these organs reversed even during daily sc administration of KGF-2.
The route of administration had dramatic effects on the observed proliferation. While daily ip administration increased the rate of liver proliferation over a 3 day period, animals given KGF-2 sc daily exhibited elevated rates after one day of treatment only. Even more surprising was the response of the pancreas. When animals were given KGF-2 ip, the pancreas exhibited a significantly elevated level of proliferation over the 14 day study period. However, sc administration of KGF-2 induced no increased mitotic activity in the pancreas. Likewise, ip, but not sc, treatment with KGF-2 elicited proliferation of the bladder mucosa.
IP administration of KGF-2 elicited a short-lived, small burst of proliferation in the kidney that was centered in the region containing collecting ducts. Daily sc treatment induced a prolonged, exaggerated proliferation in this Example 23 Effects ofKGF-2 d33 on Lung CellularProliferation Followinglntratracheal Administration This example demonstrates thatKGF-2 A33 is capable ofstimulating lung proliferation in normal rats following intratracheal administration (administration of KGF-2 X33 directly to the lung).
Methods Male Lewis rats (220-270 g), (n=5/treatment group) were used in these studies. KGF-2 X33 or placebo (40 mM sodium acetate + 150 mM NaC 1 at pH
6.5) was administered intratracheally at doses of 1 and 5 mg/kg in a volume of 0.6 mls followed by 3 mls of air. Treatments were administered on day 1 and day 2 of the experimental protocol.
On day 3, the day of sacrifice, rats were injected ip with 100 mg/kg of BrdU. Two hours later the rats were killed by C02 asphyxiation. Lungs were inflated with 10% buffered formalin via intratracheal catheter, and saggital sections of lung were paraffin embedded. To detect BrdU incorporation into replicating cells, five micron sections were subjected to immunohistochemical procedures using a mouse and-BrdU monoclonal antibody and the ABC Elite detection system. The sections were lightly counterstained with hematoxylin.
Sections were read by two blinded observers. The number of proliferating cells was counted in 10 random fields per section at a 20 x magnification. The results are expressed as the number of BrdU positive cells per field. Data are presented as mean f SEM. Statistical analyses (unpaired t-test) were performed with the Instat v2Ø1 and statistical significance is defined as p < 0.05.
Results Intratracheal injection of KGF-2 X33 at 1 and 5 mg/kg resulted in an increase in proliferation of lung epithelial cells as shown in Figure 31. KGF-X33 treatment resulted in statistically significant increases in the number of BrdU
positive cells/field at 1 mg/kg 23.4 cells/field (p=0.0002) and at 5 mg/kg 1 ~3 cells/field (p--0.0003) relative to buffer controls of 1.58 cells per field.
Example 24 KGF 2 Induces Proliferation ofNormat Rat Salivary and Lacrimal Glands Recombinant human KGF-2, a new member of the fibroblast growth factor (FGF) family was produced and purified. KGF-2 (FGF-10) has been shown to induce proliferation of mouse keratinocytes and rat prostate epithelial cells in vitro (Igarashi, M., etal., J. Biol. Chem. 273:13230-13235 (1998);
Emoto, H., et al., J. Biol. Chem. 272:23191-23194 (1997); and Lu, W., et al., FASEB
Journal 12:A1463 (1998)). The effects of KGF-2 on the proliferative rate of salivary and lacrimal glands in normal rats were studied in this example. The lacrimal glands and two major salivary glands (parotid and submandibular) of normal rats that had been treated for 1-7 days with KGF-2 were examined and, as shown below, were found to have increased cellular proliferation.

WO 99/41282 PC'f/US99103018 Materials and Methods:
Recombinant KGF 2 A bacterial expression vector (pHE4) encoding recombinant human KGF-2 was used to produce and purify KGF-2 X33 from an E. coli expression system as previously described. The purified protein was >95 % pure as determined by SDS-PAGE and reverse-phase high performance liquid chromatography and had low endotoxin levels (<0.5 EU/mg and <5 EU/ml).
Experimental Design Male Sprague-Dawley rats were obtained from Harlan Sprague Dawley, Inc, Indianapolis, IN and weighed 166-243 g when used in these studies. The animals were maintained according to recommended standards (10) in microisolator cages with recycled paper bedding (Harlan Sprague Dawley, Inc) and provided with pelleted rodent diet (Harlan Sprague Dawley, Inc) and bottled drinking water on an ad libitum basis. The animal protocols used in this study were reviewed and approved by the Human Genome Sciences Institutional Animal Care and Use Committee.
Animals were used in groups of 6 and were injected with KGF-233 (formulated in 40 mM sodium acetate and 150 mM sodium chloride at pH 6.5) at a dose of 5 mg/kg or with buffer (40 mM sodium acetate and 150 mM sodium chloride at pH 6.5). Systemic treatments were administered by the intravenous route daily for a period of 1-7 days and rats were sacrificed 24 hours after the last treatment. To examine the reversibility of effects induced with KGF-233, additional animals were injected daily for 7 days with KGF-233 or buffer and sacrificed after a 7 day treatment-free period.
Histology Two hours before sacrifice the rats were injected ip with 100 mg/kg of BrdU (Boehringer Mannheim Corp, Indianapolis, Il~. The salivary glands were isolated, weighed, and fixed in 10% neutral buffered formalin for 24 hours.
The eyes were removed intact and fixed.. After fixation the tissues were paraffin embedded. To detect BrdU incorporation into replicating cells, five micron sections were subjected to immunohistochemical procedures using a mouse anti-BrdU monoclonal antibody (Boehringer Mannheim Corp) and the ABC Elite detection system (Vector Laboratories, Inc, Burlingame, CA). The sections were lightly counterstained with hematoxylin. Additional paraffin sections were stained with hematoxylin and eosin (H&E).
All sections used for quantitation were read by blinded observers. Three areas were randomly selected at a 1 Ox magnification and the percent of positive cells per region of interest (% ROI) was assessed with the IPLab Spectrum software {version 3.1.1) from Signal Analytics Corp., Vienna, VA.
Statistical analyses Data are presented as mean t SEM. Statistical analyses were performed with the StatView Software package (Abacus Concepts, Inc., Berkeley, CA). A
factorial ANOVA with post hoc comparisons of Scheffe was used.
Results After 5 to 6 daily intravenous injections with KGF-233 at a dose of 5 mg/kg, some of the rats had a wet muzzle. The effects of KGF-233 on the proliferation rate of the parotid and submandibular salivary glands were observed.
Because both the salivary and lacrimal glands contain similar cells, and damage to both glands occurs in clinical settings, the lacrimal gland was examined as well.
Parotid gland KGF-2A33 at a dose of 5 mg/kg significantly increased proliferation of the serous secretory acini in the parotid gland after one injection.
Quantitative analysis also showed significant proliferation. As can be seen in Figure 32, one WO 99/41282 PC'T/US99/03018 injection of KGF-233 induced a 40-fold increase in proliferation. Following seven daily injections the response had diminished somewhat to an 18-fold increase. Animals treated for seven days and then allowed to recover for seven days had no elevated proliferation demonstrating that this dramatic effect was reversible. This increased proliferation resulted in an increase in organ size.
KGF- 2433 induced a three-fold increase in the weight of the parotid gland after seven daily injections but its size had returned to normal after a seven day recovery period (Figure 33).
Submandibular gland KGF-233 treatment significantly increased proliferation of the submandibular gland. Both serous and mucous secretory cells are affected although the ducts appear to be resistant to the proliferation-enhancing action of KGF-2A33. The proliferation was quantitated and the results shown in Figure 34.
KGF-233 enhanced growth ofcells after 1-3 daily treatments. After seven days there remained a slight elevation in proliferation but this did not attain statistical significance. Unlike the parotid gland, this increase in cellular proliferation was rapidly reflected in an increase in gland weight that started to normalize after treatment was withdrawn for 7 days (Figure 35).
Discussion Inhibition of saliva and tear production is a major clinical problem and currently the treatment for keratoconjunctivitis sicca alone costs the US over $100 million (temp, M.A, Adv. Farp. Med. Biol. 438:791-803 (1998)). The parotid gland is one of the three major salivary glands and is composed of serous cells that secrete a watery substance that contains enzymes such as amylase and lysozyme. The submandibular gland contains both serous and mucous secretory cells and secretes a thicker liquid (Wheater's functional histology. A text and colour atlas, New York: Churchill Livingstone, Ed. 3 (1993)).

WO 99141282 PC'TIUS99l03018 KGF-2 induced a dramatic increase in the proliferation of the parotid and submandibular glands within 24 hours of the first intravenous injection. The augmented proliferation of the parotid gland remained over the seven day treatment period while the submandibular gland had normalized during seven days of daily administration of KGF-2. These proliferative changes were reflected in the organs by an increase in weight although there were spatial differences in the response.
Continuous infusions of epidermal growth factor (EGF) have been reported to increase proliferation ofthe parotid and submandibular salivary glands in rats and mice but its effect is more pronounced in the ductal epithelium.
Ohlsson et al. reported an increase in both the serous and ductal cells following EGF administration in the mouse (Ohlsson, B., et al., Pancreas 14:94-98 ( 1997)).
The proliferation occurred in the parotid and submandibular glands after three and seven days of continuous treatment, respectively. There was a higher index of labeling of the ductal rather than the serous cells in the parotid gland and the submandibular gland with 12 and 3 fold increases, respectively. The prnliferation of the serous cells in the parotid and submandibular gland reached maximums of 5 and 2 fold, respectively. There was no change in organ weights. Breider et al., working in the rat, found continuous infusions of EGF for four weeks induced proliferation of the salivary gland ductular epithelium but no change of the acinar cells (Breider, M.A., et al., Vet Pathol 33:184 ( 1996)). As compared to EGF, KGF-2 induced a far more pronounced proliferation of the serous epithelium in both salivary glands.
Another growth factor has been reported to induce some changes in the salivary gland. KGF-1, which shares some homology with KGF-2, that has been reported to induce excessive salivation in transgenic mice within five days of birth (Guo, L., et al., EMBOJ. 12:973-986 (1993)). The submandibular gland of these mice showed an undifferentiated morphology with few or no secretory granules. KGF-2, administered systemically in rats over a period of one week, did not induce such a change in these cells.

In su~nmaiy, KGF-2 induced dramatic elevations in the proliferation ofthe saliva and tear producing cells of the salivary and lacrimal glands. It appeared to have little effect on the ductal cells in these organs although careful analysis of both cell types was not performed. These results suggest that KGF-2 may have clinically useful effects on salivary glands damaged as a result of radiation therapy, autoimmune diseases, or other medical conditions.
Example 25 KGF 2d33 Induces Proliferation in the F.xtraorbital Lacrimal Gland Since KGF-21133 has shown proliferative effects on secretory tissues such as salivary glands and the pancreas, the effect of systemic administration of KGF-2433 on the lacrimal glands was also investigated.
Materials and Methods Normal male Sprague Dawley rats were injected intravenously daily with KGF-2033 (HG03411-E6) or buffer. Two hours before sacrifice they received a systemic injection of BrdU so that proliferating cells could be detected immunohistologically. The lacrimal glands were excised and then fixed in 10%
neutral buffered formalin for histological processing. Animals were used in groups of 6, and the proliferation of the tissue was assessed by a blinded observer using a computerized morphometry unit. The proliferation results are expressed as the percentage of a region of interest that has proliferating cells (%
ROI). The error bars reflect the SEM. Statistical analysis was performed using StatView.
A
factorial ANOVA was performed followed by a Scheffd post-hoc test and statistical significant was defined as p< 0.05.
Results As can be seen from Figure 36, the lacrimal gland proliferates after 1, 2, and 3 daily i.v. treatments with KGF-233. However, the gland failed to demonstrate elevated proliferation after 7 daily administrations of this growth WO 99/41282 PGT/US99l03018 factor, a situation that has been observed in many organs and tissues. After 7 daily treatments, and then 7 days of rest, the buffer group displays a higher level of proliferation than the KGF-2 treated animals, possibly due to a down-regulation of KGF-2 receptors.
These results suggest that topical or systemic administration of KGF-233 may stimulate a therapeutic increase in the secretory capacity of the glands by its proliferative effect on lacrimal epithelial cells. These results also suggest that KGF-2 may have clinically useful effects on lacrimal glands damaged as a result Of radiation therapy, autoimmune diseases, or other medical conditions.
Exa»~nde 26 The Effect of KGF 2 on the Cornea and Conjunctiva This experiment was performed to determine the effect of KGF-2 on the cornea and conjunctiva.
Experimental Design KGF-2033 was intravenously injected at 5 mg/kg on a daily basis in male SD rats. The animals were sacrificed at days 1, 2, 3 and 7. Another group of animals were injected for 7 days and allowed to rest for 7 before sacrifice.
Two hours before sacrifice, the animals were injected with 100 mg/kg of BrdU. The eyes and the attached conjunctiva were removed. The number of replicating epithelial cells, which had incorporated BrdU, in the cornea and conjunctiva.
The results express the replicating epithelia as number of cells per mm of tissue.
Results Cornea As can be seen from Figure 37, an increase in the number of proliferating cells following KGF-2033 treatment on days 1 and 2 was observed. However, only the results from day 2 attained statistical significance. A factorial-ANOVA
was performed on the results which revealed that there was no interaction between KGF-21133 treatment and time. Therefore, KGF-233 does not appear to cause proliferation of epithelial cells in the cornea (p=0.077). However, as can be seen from the p value, the results approach significance.
Conjunctiva As can be seen from Figure 38, an increase in the number of proliferating cells following KGF-2033 treatment on days 1, 2 and 3 was observed. However, the number of proliferating cells returned to normal values by the 7"' day of treatment. A factorial-ANOVA on the results revealed that KGF-233 causes proliferation of the epithelial cells in the conjunctiva (p=0.0024).
Example 27 The effect of KGF-2 on pilocarpine-induced salivation Clinical syndromes of xerostomia (dry mouth) result from either radiation therapy to treat head and neck tumors or Sjorgen's Syndrome. Radiation therapy causes severe reduction in the saliva that is produced by a patient and can lead to problems such as difficulty eating and swallowing, mouth sores and lesions, and speech difficulties. Current treatments are sub-optimal. The most common treatment is oral administration of pilocarpine tablets to induce salivation.
Since KGF-233 exerts a proliferative effect on the cells of the salivary glands, it was interest to determine if KGF-233 injections also induced a functional difference in treating rats.
Materials and Methods Normal male Sprague Dawley rats were injected intravenously daily with KGF-233 or buffer for 7 days. Animals were used in groups of fi. The results were analyzed using Student's t-test. Saliva was collected after 7 days of injections, by first anesthetizing the rats and then injecting them with pilocarpine HCl (2 mg/kg i.p.).
Results Saliva was collected for 30 minutes directly from the mouth starting at the time of the pilocarpine injection. Saliva volume was calculated from weight and was then assayed for amylase content.
As shown in Figure 39 (A), KGF-233 treatment induces a large increase in saliva production in the treated animals. Figure 39(B) shows that the concentration of amylase in saliva is decreased following the daily KGF-21133 injections. Due to the massive proliferation brought on by KGF-233 treatment to the secretory acini of the parotid and submandibular salivary glands, it follows that there was an increase in saliva production. Thus, KGF-2 may be useful to treat xerostomia. -Example 28 Effect of ~CGF-2 on Maxillary Sinus and Nasal Septum KGF-2'033 was administered to normal S.D. rats intravenously at a dose of Smg/kg. Animals were sacrificed 1, 2, 3 and 7 days following treatment.
Additionally, one gmup of rats were injected daily with KGF-2033 for 7 days and allowed to rest for 7 days (Day 14). Two hours prior to sacrifice, each animal was injected with 100mg/kg of BrdU.
At the time of sacrifice, animals were decapitated and their nasal cavities were immediately flushed and fixed in neutral phosphate- buffered 10%
formalin.
The total number of labeled (BrdU) respiratory epithelial cells of both the nasal septum and maxillary sinus were counted and expressed per mm length of tissue.

Results Maxillary Sinus:
As shown in Figure 40, an increase was observed in the total number of proliferating cells following KGF-233 treatment on days 1, 2, 3 and 7, all of which attained statistical significance (Factorial-ANOVA: p=.0024, <.0ppl, .0190 and.0374, respectively). The number of proliferating cells returned to normal by Day 14.
Nasal Septum:
As shown in Figure 41, the trend seen in the respiratory epithelium of the maxillary sinus was also mirrored in that of the nasal septum. There was an increase in the total number of proliferating cells following KGF-2033 treatment on Days 1, 2, 3 and 7. All days, except Day 14, attained statistical significance (Factorial-ANOVA: p=.0022, <.0001, <.pppl ~d .0171, respectively). By day 14, the number of proliferating cells returned to normal.
Conclusion At a dose of Smg/kg, KGF-2033 causes a significant increase in the total number of Brdu labeled epithelial cells of both the maxillary sinus and nasal septum. By Day 14, the proliferation returns to normal levels.
Example 29 E, ffect of ~CGF-2 on Goblet Cells in the Conjunctiva KGF-233 was administered to male SD rats intravenously at a dose of Smg/kg. The animals were sacrificed at days 1, 2, 3 and 7; additional rats were injected for 7 days and allowed to rest for 7 more days before sacrifice. The eyes and the attached conjunctiva were removed. The tissue was stained with PAS
stain, and the number of PAS-positive cells (Goblet cells) in the conjunctiva was counted. The results are expressed as number of goblet cells per millimeter of tissue.
Results During the treatment period (days 1, 2, 3 and 7), there were no differences between the two treatment groups, KGF-2A33 and buffer. Animals that were treated for 7 days with KGF-2L133, and allowed to rest for 7 more days (Day 14 group on graph), had a higher number of goblet cells than those treated with buffer. This difference approached statistical significance (p= 0.085, Scheffe's post-hoc test). The results are shown in Figure 42.
1 o Example 30 Effect of RGF 2 on Ih Yivo Lung Prol ferat~ton Methods Male Sprague Dawley rats (approximately 200g, n=5 per group) were anesthetized with isoflurane gas while in a supine position. Buffer or KGF-2 15 ( 1 or S mg/kg) was administered intravenously (i.v.) via the tail vein in a volume of 0.5 ml. A single i.v. dose of KGF-233 or placebo was administered on experimental day 1. Animals were euthaluzed at 6, 24 or 48 hours following i.v.
injection. On the day of euthanasia, the rats were injected i.p. with 100 mg/kg BrdU two hours prior to sacrifice. Euthanasia was can~ied out by anesthetizing 20 with ketamine/xylazine followed by cervical dislocation.
Lung Tissue Harvest A longitudinal skin incision was made from the sternai region to the chin.
The left superior vena cava was clamped and the thoracic cavity was opened through the sternal borders and separated from the ribs. The lungs were released from the diaphragm and the pulmonary system was flushed of blood by inj ecting saline (10-15 mL) into the right ventricle. The blood/saline exited the pulmonary system via the left atrium. The left lobe was clamped at the main bronchus with artery forceps, while the right lung was inflated with 10-15 mL of 10%
formalin via a slit hole in the trachea over the neck region, clamped and allowed to fix for about five minutes. The upper right lung and lower right was then removed, placed in a tissue cassette and immersed in 10% formalin for 24 hours. Lung tissue was placed in 70% alcohol and submitted for histopathological analysis.
Tissue was embedded in paraffin, cross-sectioned and stained with H&E and a mouse anti-BrdU monoclonal antibody.
Histological Analysis The number BrdU positive cells was counted under light microscopy at a magnification of 20X. 10 random fields were counted from each tissue sample.
Counting was done by two independent observers who were blind to the treatment groups.
Statistical Analyse All statistical analysis was accomplishes using Instat v2.01. Experimental data was analyzed using an unpaired t test. A p value of < 0.05 was considered significant. The data expressed as the mean t SEM.
Results The results of the BrdU proliferation studies are shown in Figure 43. In rats receiving buffer, there were approximately 36 BrdU positive cells per field under 20x magnification. At 24 hours after a single i.v. treatment of 1 mg/kg KGF-2033, 97 BrdU positive cells per field were observed; an increase that was significant (p=0.0118) relative to the KGF-2 buffer control group. Rats receiving 5 mglkg KGF-2033 exhibited a significant increase in BrdU positive cells per field ( 197 cell per field; p<0.0001 ) when compared to the buffer control (37 cells per field). At the 6 and 48 hour time points, there were no differences observed between the groups with regard to the number of BrdU positive cells.
Conclusion At 6 hours after i.v. injection of KGF-2 X33, no increase in proliferation was observed by BrdU immunostaining.
A single i.v. administration of KGF-2 X33 at 5 mg/kg resulted in a significant increase in BrdU immunostaining in the lung relative to buffer controls in both experiments.
In one oftwo experiments performed, an i.v. dose of 1 mg/kg KGF-2 X33 resulted in a significant increase in BrdU immunostaining relative to buffer control.
At 48 hours after i.v. administration of KGF-2 X33, there was no observed difference in proliferation of lung cells among the groups.
Example 31 Effect of Nebulized gGF 2 on In Vivo Lung Proliferation Methods Male Lewis or SD rats (250-3 50 g, n=5 per group) were anesthetized with intramuscular ketamine/xylazine to immobilize them during the exposure period.
A Pari-Proneb LC Jet+ nebulizer was used to nebulize buffer or KGF-2033 .This nebulizer delivers a range of particle sizes whereby 74% of the particles are <5 p,m in size and 24% of the particles are <2 p in size. Rats inhaled nebulized buffer of KGF-233 (6 or 12 mg/rat) over a period of 45 to 60 minutes. Twenty-four hours after the exposure period, (Day 2), the rats were injected i.p.
with 100mg/kg BrdU two hours prior to sacrifice. Euthanasia was carried out by COZ
asphyxiation for tissue harvesting.
Lung Tissue Harvest A longitudinal skin incision was made from the sternum region to the chin. The left superior versa cava was clamped and the thoracic cavity was opened through the sternal borders and separated from the ribs. The lungs were released from the diaphragm. The pulmonary system was flushed of blood by injecting saline (10-15 mL) into the right ventricle. The blood/saline exited the pulmonary system via left auricle. The left lobe was clamped at the main bronchus with artery forceps, removed and snap frozen in liquid nitrogen for collagen content assay. The right lung was insuflated with 10-15 mL of 10%
formalin via a slit hole in the trachea over the neck region, clamped and allowed to fix for about five minutes. The upper right lung and lower right was then removed, placed in a tissue cassette and immersed in 10% formalin for 24 hours.
Lung tissue was placed in 70% alcohol and submitted for hisopathological analysis. Tissue was embedded in paraffin, cross-sectioned and stained with H&E and a mouse anti-BrdU monoclonal antibody.
Histological Analyse The number BrdU positive cells was counted under light microscopy at a magnification of 20X. Ten random fields were counted from each tissue sample. Counting was done by two independent observers who were blind to the treatment groups.
Statistical Analysis All statistical analysis was accomplished using Instat v2.01. Experimental data was analyzed using an unpaired t test. A p value of < 0.05 was considered significant. The data was expressed as the mean f SEM.

Nebulizer Design Five animals treated simultaneously on one nebulizer unit.
Results Microscopic Observations Upon histological observation of the BrdU cell counts, rats treated with KGF-2033 buffer (n=S) had an average of 9 BrdU positive cells per field.
Treatment with KGF-2A33 at 6 mg/rat (n=5) produced a not quite significant increase to 20 BrdU positive cells per field (p=0.0857) when compared to the buffer control. Treatment KGF-2033 at 12 mg/rat (n=5) resulted in a significant increase in BrdU positive cells per field (32 cells per field; p~.001 ) when compared to the buffer control. The results are shown in Figure 44.
Example 32 Prophylactic KGF-2 d33 in Bleomycin Lung Damage The purpose of this example was to examine the effect of KGF-2033 in the bleomycin-induced rat model of lung injury. Bleomycin causes fibrosis of the lung. KGF-233 was tested for its prophylactic (protective) effect in the lungs.
Parameters that were evaluated include body weight; histology, lung collagen content.
Methods Male Lewis rats (200-250 g, n=5 to 8 per group) were anesthetized with isoflurane gas and while in a supine position. A pharyngo-laryngo fiberscope was used to locate the pharynx and a metal feeding tube (20 gauge) attached to a 5 ml syringe was inserted into the trachea. Buffer or KGF-2 033 (0.5, 1 or Smg/kg) was administered intratracheally (i.t.) into the lungs in a volume of 0.6 ml followed by 4 cc of air on days 1 and 2 of the experiment. The air was then used to drive the liquid into the lungs. The rats were quickly placed in a recumbent position and allowed to recover. Resuscitation was done if necessary. On day 3 of the experiment, bleomycin (Sunits/O.SmI) was administered intratracheally to S all the animals except for the no treatment group. Clinical observations were made daily (e.g. weight loss, death etc.) On the last day of the experiment (Day 14), the rats were injected i.p. with 100mg/kg BrdU two hours prior to sacrifice.
Euthanasia was carned out by COZ asphyxiation for tissue harvesting, histopathological analysis and lung collagen content assay.
Lung Tissue Harvest A longitudinal skin incision was made from the xiphisternal region to the neck region. The thoracic cavity was opened through the sternal borders and separated from the ribs. The lungs were released from the diaphragm and the left lobe was clamped at the main bronchus .with artery forceps, removed and snap frozen in liquid nitrogen for collagen content assay. The right lung was inflated with 10% formalin via a slit hole in the trachea over the neck region, clamped and allowed to fix for about five minutes. The right lung was then removed, placed in a tissue cassette and immersed in 10% formalin for 24 hours. Lung tissue was placed in 70% alcohol and submitted for histopathological analysis. Tissue was embedded in paraffin, cross-sectioned and stained with H&E and mouse anti-BrdU monoclonal antibody.
Histological Analysis The number of BrdU positive cells was counted under light microscopy at a magnification of 20X. Ten random fields were counted from each tissue sample. Counting was performed by two independent observers who were blinded to the treatment groups.

WO 99!41282 PCTNS99/03018 Statistical Analysis All statistical analysis was accomplished using Instat v2.01. Experimental data was analyzed using an unpaired t test. A p value of < 0.05 was considered significant. The data was expressed as the mean f SEM.
Results Body Weight Figure 45 shows the body weights of the animals during this experiment.
Animals in the "no treatment" group continued to increase in weight throughout the experiment. Animals receiving bleomycin and KGF-2 buffer lost weight during the experiment until day 7. Following day 7, these animals began to gain weight. In the treatment groups, KGF-2 a33 at a dose of Img/kg, was able to prevent the weight loss observed in the other groups receiving bleomycin. This difference in body weight was seen on days 5, 7 and 9 relative to the buffer placebo group. Animals receiving 0.5 or 5 mg/kg KGF-2 X33 had body weights that were similar to the KGF-2 buffer controls.
Microscopic Observations No architectural damage was observed in H&E stained sections of the lung parenchyma of the "no treatment" group animals. Fibrosis scoring was done using a scale ranging from 0-9, (0=normal,1-3=mild, 4-6-moderate, 7-9=severe).
Animals in the no treatment groups had a mean score of 0.56. Architectural damage was observed in all other treatment groups (groups receiving bleomycin).
As shown in Figure 46, rats treated with bleomycin and KGF-2 buffer had a mean fibrosis score of 4.24 (**p=<0.0001) a significant change relative to the no treatment group. Animals receiving KGF-2 d33 displayed significant reductions in fibrosis scores at O.Smg/kg KGF-2 033 (2.91, *p=0.0248), lmg/kg KGF-2 X33 (2.07, p=0.0004) and Smg/kg KGF-2 A33 ( 1.98, p=0.0005) when compared to the KGF-2 buffer placebo group.

Cell proliferation, shown in Figure 47, was assessed by counting the number of BrdU positive cells per field. In a normal rat the lungs have an average of 7 BrdU positive cells per field illustrating the low level of cell proliferation occurring under normal physiological conditions. When rats were treated with KGF-2 buffer and bleomycin, there was a significant increase in the number of cells per field, 40 (p=<0.0001 ) when compared to the no treatment group. Rats receiving KGF-233 displayed a significantly lower number of BrdU positive cells at doses of O.Smg/kg (26, p=0.0003) positive lmg/kg (11, p=<0.0001 ) and at Smg/kg (24, p=<0.0001 ) when compared to the KGF-2 buffer controls.
These results suggest that KGF-2 may have clinically useful effects on lung fibrosis.
Example 33 Effect of KGF 2 d33 on Bladder Capacity in a Cyclophosphamide Induced Cystitis Rat Model Experimental Design Male SD rats (200g) received KGF-233 in doses of 0.1 mg/kg, 0.3 mg/kg, 1.0 mg/kg or 3.0 mg/kg intravenously. On day 1, 200 mg/kg of cyclophosphamide (CP) was administered i.p. Animals were sacrificed on day 4. Six animals were used in each experimental group. Experimental groups included a saline control group, a CP only control (200 mg/kg) group, a buffer +
CP (200 mg/kg) group a KGF-233 (1.0 mg/kg) + Cp (200 mg/kg), and a KGF-2033 (3.0 mg/kg) + Cp (200 mglkg) group.
Two additional parameters were performed at the time of sacrifice. These measurements included bladder capacity and bladder wet weight. Bladder capacity was defined as the amount of formalin (in milliliter quantity) injected into the bladder until one drop of formalin is visible exiting from the urethra.

Prior to filling the bladder is emptied of all urine by gently squeezing the bladder and forcing the urine out through the urethra. The bladder is allowed to fix several minutes. Once fixed the bladder is removed, drained and weighed providing a measure of wet weight. (* compared to CP only control; ~ compared to buffer control).
Results Microscopic Observations The bladder capacity of the saline control group was 1.10 + 0.1 ml. The CP only group was significantly reduced to 0.62 + p.I 5 ml and the buffer contml group was 0.45 ~ 0.01 ml. The bladder capacity of the KGF-2033 groups treated with 0.3 mg/kg and 1.0 mg/kg (1.20 ~ 0.25 ml, p = 0.0206; 1.28 + 0.27, p =
0.0172, respectively) were significantly increased compared to the buffer control The bladder capacity of the KGF-233 at 3.0 mg/kg (1.72 + 0.28, p = 0.28 ml) was significantly increased compared to the CP only control (p = 0.0066) and the buffer-treated control {p = 0.0018). The results are shown in Figure 48.
The bladder wet weight of the saline control group was 0.11 mg. The wet weight of the CP only group was 0.22 ~ 0.p1 mg and that of buffer control was 0.19 ~ 0.01 mg. The KGF-2A33 group (0.3 mg/kg) wet weight (0.15 + 0.007 mg) was significantly reduced compared to the CP only control (p = 0.0015) and almost significantly reduced compared to the buffer-treated control (p =
0.0595) group. The KGF-2033 groups treated with 1.0 mg/kg (0.15 ~ 0.02 mg; p =
0.0314) and 3.0 mg/kg (0.149 ~ 0.01 mg, p = 0.0057) were significantly reduced in bladder wet weight compared to the buffer-treated control. These results are shown in Figure 49.
Example 34 E, jfect ojICGF 2 d33 and Mesna in Cyclophosphamide Induced Cystitis The effect of KGF-233 and mesna on bladder wall thickness, bladder ulceration and urothelium was examined in the cyclophosphamide-induced cystitis rat model. Mesna is currently used as a prophylactic agent for the prevention of hemorrhagic cystitis induced by cyclophosphamide or structural analogs.
Experimental Method and Design Male SD rats (300-400 g) received either buffer or 5.0 mglkg KGF-2033 intravenously via the tail vein on day 0. On day 1, 200 mg/kg of cyclophosphamide (CP) i.p and Mesna (20 ~.g/g or 40 g.g/g,) i.v. via the tail vein.
One group of animals was injected i.p. with saline as a CP control. Five animals were used in each experimental group. Experimental groups included saline control group, a CP only control group (200 mg/kg), a Mesna (20 mg/kg) + CP
(200 mg/kg) group, a Mesna (40 mg/kg) + Cp (200 mg/kg) group, a buffer + CP
(200 mg/kg) group, and a KGF-2A33 (5 mg/kg) + Cp (200 mg/kg) group.
Animals were sacrificed on day 3, 48 hours after CP injection the animals were injected with 100 mglkg of BrdU. Two hour later the animals were euthanized by COZ asphyxiation. The bladders were fixed for histological processing by direct injection of 10% formalin into the lumen of the bladder and rinsing of the exterior of the bladder with formalin. After 5 minutes, the bladders were harvested. The uninary bladders were parafinn embedded, cross-sectioned and stained with H&E and a mouse anti-BrdU monoclonal antibody.
The extent of urothelial damage was assessed using the following scoring system: Urothelial damage (ulceration) was scored as 0-100% (in units of 10) loss of the urothelium. The amount of normal urothelium was assessed and scored as 0-100% (in increments of 10%). Normal urothelium was defined as a dense stratified epithelium containing at least a thickness a single layer of cells appearing as tall columnar or cuboidal cells with round nuceli. In addition , the thickness of the bladder wall was measured at 10 random sites per section and expressed in Vim. This measurement included the distance from the urothelium through the smooth muscle layers to serosal surface. All histology measurements were performed by blinded observers. Statistical analysis was performed using the students unpaired t-test on Instat 2.01 where significance is found if p <
0.05.
Results Microscopic Observations In normal rats treated i.p. with saline (CP vehicle control), the bladder wall was not enlarged and no ulceration of the urothelium was observed.
Pretreatment with buffer placebo 24 hours prior to i.p. administration of CP
(200 mglkg) resulted in 43% + 19 ulceration of the total bladder epithelial area.
Administration of KGF-2 033 24 hours prior to CP resulted in a reduction in the extent of ulceration ( 1.4% + 0.24; p= 0.0641 ) when compared to placebo treated animals receiving CP. Administration of Mesna at 20 or 40 mg/kg resulted in a reduction of ulceration to 1.4% + 0.24 or 6.2% ~4.7, respectively however these values were not statistically significant relative to the control. These results are shown in Figure 50.
In some cases the urothelial lining may not exhibit ulceration but may still be abnormal in appearance. Thus, the "percent normal umthelial" was established as a measure ofthe amount of urothelium with columnar or cuboidal morphology, rounded nuclei, and possessing at least 1 cell layer in thickness. Groups treated with KGF-2033 24 hours prior to CP administration had 97% + 3 normal urothelium (p = 0.0574) and 93% + 3 (p = 0.0625) normal urothelium respectively. These values were considered to be not quite significant when compared to the CP only group. These results are shown in Figure 51.
The thickness of the urinary bladder wall was also evaluated as an additional measure of CP damage. Bladder wall thickness was measured from the epithelium through smooth muscle layers to the serosal surface. In groups treated with saline (CP vehicle control) alone, the thickness of the bladder wall is approximately 100 ~.m + 16. KGF-2 033 pretreatment (252 pm + 30; p = 0.0331 ~ 29). Mesna treatment also resulted in significant decreases in bladder wall thickness at 20 mg/kg (198 ~m ~ 34; p = 0.0361) and 40 mg/kg (172 ~m ~ 46;
p = 0.0215} when compared to the CP only group. These results are shown in Figure 52.
Example 35 Synergistic Effect of KGF 2 d33 and Mesna in Cyclophosphamide Induced cystitis The synergistic effect of KGF-233 and mesna on bladder capacity and bladder wet weight in cyclophosphamide-induced cystitis rat, model was examined.
Experimental Design Male SD rats (350-400 g) (n=7) received, on day 0, either buffer or 5.0 mg/kg KGF-233 intravenously and on day 1, Mesna (40 ~g/g, i.v.) or both treatments on the respective administration days. Cyclophosphamide (300 mglkg, i.p.) was administered on day 1 to all treatment groups with the exception of the saline control. One group was added as a CP control with no treatment. On day 3, animals received BrdU (100mg/kg, i.p.) 2 hours prior to euthanasia. After urine was removed from the bladder, it was filled with formalin until it leaked out of the urethra. The volume of formalin injected into the bladder was recorded as the bladder capacity. Bladders were fixed with 10% neutral-buffered formalin, weighed and placed in tissue cassettes for histological analysis.
The experimental groups consisted of six animals per group. The following experimental groups were used: a saline control group, a CP only control (300 mg/kg) group, a buffer + CP (300 mg/kg) group, a KGF-2 X33 (3.0 mg/kg} + a CP (300 mg/kg) group, a Mesna (40 mg/kg) + CP (300 mg/kg) group, and a KGF-2 X33 {3.0 mg/kg) + Cp (300 mglkg) + Mesna (40 mg/kg}.

Results The bladder capacity of the saline control group was 2.21 + 0.1 ml. The group receiving CP only had a significantly reduced bladder capacity of 0.80 +
0.27 ml while the buffer control was 0.40 + 0.15 ml. The bladder capacity of the KGF-2 033 treated group (1.66 ~ 0.31 ml, p = 0.0038) was significantly increased when compared to the buffer control. Likewise, the Mesna (40 mg/kg) group (1.67 ~ 0.44 ml, p = 0.0147) was significantly increased in comparison to the buffer control. The animals treated with both KGF-2 a33 and Mesna (1.96 ~ 0.19 ml) also displayed a significant increase in capacity when compared to CP
only control (p=0.0044) and the buffer control group (p<0.0001 ). These results are shown in Figure 53.
The bladder wet weight of the saline control grnup was 0.16 mg. The wet weight of the CP only control was 0.30 ~ 0.01 mg. The KGF-2 033 group (0.33 ~ 0.3 mg) and the Mesna group (0.28 ~ 0.03 mg) were no significantly different from the control groups. However, animals treated with both KGF-2 X33 and Mesna exhibited a significant reduction in bladder wet weight (0.19 + 0.08 mg) when compared to the CP only group (p--0.193) and the buffer control (p=0.0099). These results are shown in Figure 54.
Example 36 KGF 2 d33 Induced Changes in Buccal Mucosa and Tongue In cynomolgus monkeys, daily intravascular injections of KGF-233 at 300 pg/kg resulted in grossly visible thickening of the oral (bucca.l) and esophageal mucosa. Microscopically, hyperkeratosis of the buccal mucosa, tongue and esophagus were noted in monkeys treated both daily and every other day with KGF-233. The buccal and esophageal mucosa hyperkeratosis correlated with the gmssly visible thickening of the buccal and esophageal mucosa.

Example 37 KGF 2 d33lnduced Goblet Cell Changes In male Sprague Dawley rats, daily intravenous injections of KGF-233 at 5 mg/kg resulted in goblet cell hyperplasia in the respiratory epithelium of the nasal air passage way. This was characterized by a minimal to mild goblet cell hyperplasia after 7 consecutive days of KGF-2033 treatment followed by a 7 day treatment-free period.
It will be clear that the invention may be practiced otherwise than as particularly described in the foregoing description and examples.
Numerous modifications and variations of the present invention are possible in light of the above teachings and, therefore, within the scope of the appended claims, the invention may be practiced otherwise than as particularly described.
The entire disclosure of all publications (including patents, patent applications, journal articles, laboratory manuals, books, or other documents}
cited herein are hereby incorporated by reference.

l ..

SEQUENCE LISTING
<110> Human Genome Sciences, Inc.
Jimenez, Pablo Rampy, Mark A.
Mendrick, Donna Russell, Deborah Louie, Arthur <120> Therapeutic Uses of Keratinocyte Growth Factor-2 <130> 1488.106PC02 <140>
<141>
<150> US 60/114,387 <151> 30-DEC-1998 <150> US 60/074,585 <151> 13-FEB-1998 <160> 198 <170> PatentIn Ver. 2.0 <210> 1 <211> 627 <212> DNA
<213> Homo Sapiens <220>
<221> CDS
<222> (1)..(624) <400> 1 atg tgg aaa tgg ata ctg aca cat tgt gcc tca gcc ttt ccc cac ctg 48 Met Trp Lys Trp Ile Leu Thr His Cys Ala Ser Ala Phe Pro His Leu ccc ggc tgc tgc tgc tgc tgc ttt ttg ttg ctg ttc ttg gtg tct tcc 96 Pro Gly Cys Cys Cys Cys Cys Phe Leu Leu Leu Phe Leu Val Ser Ser gtc cct gtc acc tgc caa gcc ctt ggt cag gac atg gtg tca cca gag 149 Val Pro Val Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu gcc acc aac tct tct tcc tcc tcc ttc tcc tct cct tcc agc gcg gga 192 Ala Thr Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly agg cat gtg cgg agc tac aat cac ctt caa gga gat gtc cgc tgg aga 290 Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg aag cta ttc tct ttc acc aag tac ttt ctc aag att gag aag aac ggg 288 Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys ile Glu Lys Asn Gly aag gtc agc ggg acc aag aag gag aac tgc ccg tac agc atc ctg gag 336 Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu ata aca tca gta gaa atc gga gtt gtt gcc gtc aaa gcc att aac agc 384 Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser aac tat tac tta gcc atg aac aag aag ggg aaa ctc tat ggc tca aaa 932 Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys 130 135 lq0 gaa ttt aac aat gac tgt aag ctg aag gag agg ata gag gaa aat gga 480 Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly tac aat acc tat gca tca ttt aac tgg cag cat aat ggg agg caa atg 528 Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met tat gtg gca ttg aat gga aaa gga get eca agg aga gga cag aaa aca 576 Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr cga agg aaa aac acc tct get cac ttt ctt eca atg gtg gta cac tca 624 Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser tag 627 <210> 2 <211> 208 <222> PRT
<213> Homo sapiens <400> 2 Met Trp Lys Trp Ile Leu Thr His Cys Ala Ser Ala Phe Pro His Leu Pro Gly Cys Cys Cys Cys Cys Phe Leu Leu Leu Phe Leu Val Ser Ser Val Pro Val Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu WO 99/41282 PCf/US99103018 Ala Thr Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu IIe Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> 3 <211> 36 <212> DNA
<213> Homo Sapiens <400> 3 ccccacatgt ggaaatggat actgacacat tgtgcc 36 <210> 4 <211> 35 <212> DNA
<213> Homo sapiens <400> 4 cccaagcttc cacaaacgtt gccttcctct atgag 35 <210> 5 <211> 36 <212> DNA

<213> Homo Sapiens <400> 5 catgccatgg cgtgccaagc ccttggtcag gacatg 36 <210> 6 <211> 35 <212> DNA
<213> Homo Sapiens <900> 6 cccaagcttc cacaaacgtt gccttcctct atgag 35 <210> 7 <211> 35 <212> DNA
<213> Homo sapiens <400> 7 gcgggatccg ccatcatgtg gaaatggata ctcac 35 <210> 8 <211> 27 <212> DNA
<213> Homo sapiens <400> 8 gcgcggtacc acaaacgttg ccttcct 27 <210>9 <211>40 <212>DNA

<213>Homo Sapiens <400> 9 taacgaggat ccgccatcat gtggaaatgg atactgacac 40 <210> 10 <211> 38 <212> DNA
<213> Homo Sapiens <400> 10 taagcactcg agtgagtgta ccaccattgg aagaaatg 38 <210> 11 <211> 54 <212> DNA
<213> Homo Sapiens <400> 11 attaaccctc actaaaggga ggccatgtgg aaatggatac tgacacattg tgcc 59 <210> 12 <211> 35 <212> DNA
<213> Homo Sapiens '<400> 12 cccaagcttc cacaaacgtt gccttcctct atgag 35 <210>13 <211>206 <212>PRT

<213>Homo Sapiens <400> 13 Met Ser Gly Pro Gly Thr Ala Ala Val Ala Leu Leu Pro Ala Val Leu Leu Ala Leu Leu Ala Pro Trp Ala Gly Arg Gly Gly Ala Ala Ala Pro Thr Ala Pro Asn Gly Thr Leu Glu Ala Glu Leu Glu Arg Arg Trp Glu Ser Leu Val Ala Leu Ser Leu Ala Arg Leu Pro Val Ala Ala Gln Pro Lys Glu Ala Ala Val Gln Ser Gly Ala Gly Asp Tyr Leu Leu Gly Ile 65 70 75 8p Lys Arg Leu Arg Arg Leu Tyr Cys Asn Val Gly Ile Gly Phe His Leu Gln Ala Leu Pro Asp Gly Arg Ile Gly Gly Ala His Ala Asp Thr Arg Asp Ser Leu Leu Glu Leu Ser Pro Val Glu Arg Gly Val Val Ser Ile Phe Gly Val Ala Ser Arg Phe Phe Val Ala Met Ser Ser Lys Gly Lys Leu Tyr Gly Ser Pro Phe Phe Thr Asp Glu Cys Thr Phe Lys Glu Ile Leu Leu Pro Asn Asn Tyr Asn Ala Tyr Glu Ser Tyr Lys Tyr Pro Gly Met Phe Ile Ala Leu Ser Lys Asn Gly Lys Thr Lys Lys Gly Asn Arg Val Ser Pro Thr Met Lys Val Thr His Phe Leu Pro Arg Leu <210> 14 <211> 198 <212> PRT
<213> Homo sapiens <900> 14 Met Ser Arg Gly Ala Gly Arg Leu Gln Gly Thr Leu Trp Ala Leu Val Phe Leu Gly Ile Leu Val Gly Met Val Val Pro Ser Pro Ala Gly Thr Arg Ala Asn Asn Thr Leu Leu Asp Ser Arg Gly Trp Gly Thr Leu Leu Ser Arg Ser Arg Ala Gly Leu Ala Gly Glu Ile Ala Gly Val Asn Trp Glu Ser Gly Tyr Leu Val Gly Ile Lys Arg Gln Arg Arg Leu Tyr Cys Asn Val Gly Ile Gly Phe His Leu Gln Val Leu Pro Asp Gly Arg Ile Ser Gly Thr His Glu Glu Asn Pro Tyr Ser Leu Leu Glu Ile Ser Thr Val Glu Arg Gly Val Val Ser Leu Phe Gly Val Arg Ser Ala Leu Phe Val Ala Met Asn Ser Lys Gly Arg Leu Tyr Ala Thr Pro Ser Phe Gln Glu Glu Cys Lys Phe Arg Glu Thr Leu Leu Pro Asn Asn Tyr Asn Ala Tyr Glu Ser Asp Leu Tyr Gln Gly Thr Tyr Ile Ala Leu Ser Lys Tyr Gly Arg Val Lys Arg Gly Ser Lys Val Ser Pro Ile Met Thr Val Thr _'7_ His Phe Leu Pro Arg Ile <210> 15 <211> 268 <212> PRT
<213> Homo sapiens <400> 15 Met Ser Leu Ser Phe Leu Leu Leu Leu Phe Phe Ser His Leu ile Leu Ser Ala Trp Ala His Gly Glu Lys Arg Leu Ala Pro Lys Gly Gln Pro Gly Pro Ala Ala Thr Asp Arg Asn Pro Arg Gly Ser Ser Ser Arg Gln Ser Ser Ser Ser Ala Met Ser Ser Ser Ser Ala Ser Ser Ser Pro Ala Ala Ser Leu Gly Ser Gln Gly Ser Gly Leu Glu Gln Ser Ser Phe Gln 65 70 75 g0 Trp Ser Pro Ser Gly Arg Arg Thr Gly Ser Leu Tyr Cys Arg Val Gly Ile Gly Phe His Leu Gln Ile Tyr Pro Asp Gly Lys Val Asn Gly Ser His Glu Ala Asn Met Leu Ser Val Leu Glu Ile Phe Ala Val Ser Gln Gly Ile Val Gly Ile Arg Gly Val Phe Ser Asn Lys Phe Leu Ala Met Ser Lys Lys Gly Lys Leu His Ala Ser Ala Lys Phe Thr Asp Asp Cys Lys Phe Arg Glu Arg Phe Gln Glu Asn Ser Tyr Asn Thr Tyr Ala Ser Ala Ile His Arg Thr Glu Lys Thr Gly Arg Glu Trp Tyr Val Ala Leu Asn Lys Arg Gly Lys Ala Lys Arg Gly Cys Ser Pro Arg Val Lys Pro Gln His Ile Ser Thr His Phe Leu Pro Arg Phe Lys Gln Ser Glu Gln _g_ Pro Glu Leu Ser Phe Thr Val Thr Val Pro Glu Lys Lys Asn Pro Pro Ser Pro Ile Lys Ser Lys Ile Pro Leu Ser Ala Pro Arg Lys Asn Thr Asn Ser Val Lys Tyr Arg Leu Lys Phe Arg Phe Gly <210> 16 <211> 155 <212> PRT
<213> Homo sapiens <400> 16 Met Ala Glu Gly Glu Ile Thr Thr Phe Thr Ala Leu Thr Glu Lys Phe Asn Leu Pro Pro Gly Asn Tyr Lys Lys Pro Lys Leu Leu Tyr Cys Ser 2p 25 30 Asn Gly Gly His Phe Leu Arg Ile Leu Pro Asp Gly Thr Val Asp Gly Thr Arg Asp Arg Ser Asp Gln His Ile Gln Leu Gln Leu Ser Ala Glu Ser Val Gly Glu Val Tyr Ile Lys Ser Thr Glu Thr Gly Gln Tyr Leu Ala Met Asp Thr Asp Gly Leu Leu Tyr Gly Ser Gln Thr Pro Asn Glu Glu Cys Leu Phe Leu Glu Arg Leu Glu Glu Asn His Tyr Asn Thr Tyr Ile Ser Lys Lys His Ala Glu Lys Asn Trp Phe Val Gly Leu Lys Lys Asn Gly Ser Cys Lys Arg Gly Pro Arg Thr His Tyr Gly Gln Lys Ala Ile Leu Phe Leu Pro Leu Pro Val Ser Ser Asp <210> 17 <211> 155 <212> PRT
<213> Homo sapiens <400> 17 Met Ala Ala Gly Ser Ile Thr Thr Leu Pro Ala Leu Pro Glu Asp Gly Gly Ser Gly Ala Phe Pro Pro Gly His Phe Lys Asp Pro Lys Arg Leu Tyr Cys Lys Asn Gly Gly Phe Phe Leu Arg Ile His Pro Asp Gly Arg Val Asp Gly Val Arg Glu Lys Ser Asp Pro His Ile Lys Leu Gln Leu Gln Ala Glu Glu Arg Gly Val Val Ser Ile Lys Gly Val Cys Ala Asn Arg Tyr Leu Ala Met Lys Glu Asp Gly Arg Leu Leu Ala Ser Lys Cys Val Thr Asp Glu Cys Phe Phe Phe Glu Arg Leu Glu Ser Asn Asn Tyr Asn Thr Tyr Arg Ser Arg Lys Tyr Thr Ser Trp Tyr Val Ala Leu Lys Arg Thr Gly Gln Tyr Lys Leu Gly Ser Lys Thr Gly Pro Gly Gln Lys Ala Ile Leu Phe Leu Pro Met Ser Ala Lys Ser <210> 18 <211> 208 <212> PRT
<213> Homo sapiens <900> 18 Met Ala Pro Leu Gly Glu Val Gly Asn Tyr Phe Gly Val Gln Asp Ala Val Pro Phe Gly Asn Val Pro Val Leu Pro Val Asp Ser Pro Val Leu Leu Ser Asp His Leu Gly Gln Ser Glu Ala Gly Gly Leu Pro Arg Gly Pro Ala Val Thr Asp Leu Asp His Leu Lys Gly Ile Leu Arg Arg Arg Gln Leu Tyr Cys Arg Thr Gly Phe His-Leu Glu Ile Phe Pro Asn Gly Thr Ile Gln Gly Thr Arg Lys Asp His Ser Arg Phe Gly Ile Leu Glu Phe Iie Ser Ile Ala Val Gly Leu Val Ser Ile Arg Gly Val Asp Ser Gly Leu Tyr Leu Gly Met Asn Glu Lys Gly Glu Leu Tyr Gly Ser Glu Lys Leu Thr Gln Glu Cys Val Phe Arg Glu Gln Phe Glu Glu Asn Trp Tyr Asn Thr Tyr Ser Ser Asn Leu Tyr Lys His Val Asp Thr Gly Arg Arg Tyr Tyr Val Ala Leu Asn Lys Asp Gly Thr Pro Arg Glu Gly Thr Arg Thr Lys Arg His Gln Lys Phe Thr His Phe Leu Pro Arg Pro Val Asp Pro Asp Lys Val Pro Glu Leu Tyr Lys Asp Ile Leu Ser Gln Ser <210> 19 <211> 194 <212> PRT
<213> Homo sapiens <900> 19 Met His Lys Trp Ile Leu Thr Trp Ile Leu Pro Thr Leu Leu Tyr Arg Ser Cys Phe His Ile Ile Cys Leu Val Gly Thr Ile Ser Leu Ala Cys Asn Asp Met Thr Pro Glu Gln Met Ala Thr Asn Val Asn Cys Ser Ser Pro Glu Arg His Thr Arg Ser Tyr Asp Tyr Met Glu Gly Gly Asp Ile Arg Val Arg Arg Leu Phe Cys Arg Thr Gln Trp Tyr Leu Arg Ile Asp Lys Arg Gly Lys Val Lys Gly Thr Gln Glu Met Lys Asn Asn Tyr Asn Ile Met Glu Ile Arg Thr Val Ala Val Gly Ile Val Ala Ile Lys Gly Val Glu Ser Glu Phe Tyr Leu Ala Met Asn Lys Glu Gly Lys Leu Tyr Ala Lys Lys Glu Cys Asn Glu Asp Cys Asn Phe Lys Glu Leu Ile Leu Glu Asn His Tyr Asn Thr Tyr Ala Ser Ala Lys Trp Thr His Asn Gly Gly Glu Met Phe Val Ala Leu Asn Gln Lys Gly Ile Pro Val Arg Gly Lys Lys Thr Lys Lys Glu Gln Lys Thr Ala His Phe Leu Pro Met Ala Ile Thr <210> 20 <211> 208 <212> PRT
<213> Homo Sapiens <400> 20 Met Trp Lys Trp Ile Leu Thr His Cys Ala Ser Ala Phe Pro His Leu Pro Gly Cys Cys Cys Cys Cys Phe Leu Leu Leu Phe Leu Val Ser Ser Val Pro Val Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu Ala Thr Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> 21 <211> 239 <212> PRT
<213> Homo sapiens <400> 21 Met Gly Leu Ile Trp Leu Leu Leu Leu Ser Leu Leu Glu Pro Gly Trp Pro Ala Ala Gly Pro Gly Ala Arg Leu Arg Arg Asp Ala Gly Gly Arg Gly Gly Val Tyr Glu His Leu Gly Gly Ala Pro Arg Arg Arg Lys Leu Tyr Cys Ala Thr Lys Tyr His Leu Gln Leu His Pro Ser Gly Arg Val Asn Gly Ser Leu Glu Asn Ser Ala Tyr Ser Ile Leu Glu Ile Thr Ala Val Glu Val Gly Ile Val Ala Ile Arg Gly Leu Phe Ser Gly Arg Tyr Leu Ala Met Asn Lys Arg Gly Arg Leu Tyr Ala Ser Glu His Tyr Ser Ala Glu Cys Glu Phe Val Glu Arg Ile His Glu Leu Gly Tyr Asn Thr Tyr Ala Ser Arg Leu Tyr Arg Thr Val Ser Ser Thr Pro Gly Ala Arg Arg Gln Pro Ser Ala Glu Arg Leu Trp Tyr Val Ser Val Asn Gly Lys Gly Arg Pro Arg Arg Gly Phe Lys Thr Arg Arg Thr Gln Lys Ser Ser Leu Phe Leu Pro Arg Val Leu Asp His Arg Asp His Glu Met Val Arg Gln Leu Gln Ser Gly Leu Pro Arg Pro Pro Gly Lys Gly Val Gln Pro Arg Arg Arg Arg Gln Lys Gln Ser Pro Asp Asn Leu Glu Pro Ser His Val Gln Ala Ser Arg Leu Gly Ser Gln Leu Glu Ala Ser Ala His <210> 22 <211> 268 <212> PRT
<213> Homo Sapiens -<900> 22 Met Gly Ser Pro Arg Ser Ala Leu Ser Cys Leu Leu Leu His Leu Leu Val Leu Cys Leu Gln Ala Gln Val Arg Ser Ala Ala Gln Lys Arg Gly Pro Gly Ala Gly Asn Pro Ala Asp Thr Leu Gly Gln Gly His Glu Asp Arg Pro Phe Gly Gln Arg Ser Arg Ala Gly Lys Asn Phe Thr Asn Pro Ala Pro Asn Tyr Pro Glu Glu Gly Ser Lys Glu Gln Arg Asp Ser Val Leu Pro Lys Val Thr Gln Arg His Val Arg Glu Gln Ser Leu Val Thr 85 90 g5 Asp Gln Leu Ser Arg Arg Leu Ile Arg Thr Tyr Gln Leu Tyr Ser Arg Thr Ser Gly Lys His Val Gln Val Leu Ala Asn Lys Arg Ile Asn Ala Met Ala Glu Asp Gly Asp Pro Phe Ala Lys Leu Ile Val Glu Thr Asp Thr Phe Gly Ser Arg Val Arg Val Arg Gly Ala Glu Thr Gly Leu Tyr Ile Cys Met Asn Lys Lys Gly Lys Leu Ile Ala Lys Ser Asn Gly Lys Gly Lys Asp Cys Val Phe Thr Glu Ile Val Leu Glu Asn Asn Tyr Thr Ala Leu Gln Asn Ala Lys Tyr Glu Gly Trp Tyr Met Ala Phe Thr Arg Lys Gly Arg Pro Arg Lys Gly Ser Lys Thr Arg Gln His Gln Arg Glu Val His Phe Met Lys Arg Leu Pro Arg Gly His His Thr Thr Glu Gln Ser Leu Arg Phe Glu Phe Leu Asn Tyr Pro Pro Phe Thr Arg Ser Leu Arg Gly Ser Gln Arg Thr Trp Ala Pro Glu Pro Arg <210> 23 <211> 4177 <212> DNA
<213> Homo Sapiens <220>
<221> CDS
<222> (593)..(1216) <400> 23 ggaattccgg gaagagaggg aagaaaacaa cggcgactgg gcagctgcct ccacttctga 60 caactccaaa gggatatact tgtagaagtg gctcgcaggc tggggctccg cagagagaga 120 ccagaaggtg ccaaccgcag aggggtgcag atatctcccc ctattcccca ccccacctcc I80 cttgggtttt gttcaccgtg ctgtcatctg tttttcagac ctttttggca tctaacatgg 240 tgaagaaagg agtaaagaag agaacaaagt aactcctggg ggagcgaaga gcgctggtga 300 ccaacaccac caacgccacc accagctcct gctgctgcgg ccacccacgt ccaccattta 360 ccgggaggct ccagaggcgt aggcagcgga tccgagaaag gagcgagggg agtcagccgg 420 cttttccgag gagttatgga tgttggtgca ttcacttctg gccagatccg cgcccagagg 480 gagctaacca gcagccacca cctcgagctc tctccttgcc ttgcatcggg tcttaccctt 540 ccagtatgtt ccttctgatg agacaatttc cagtgccgag agtttcagta ca atg tgg 598 Met Trp aaa tgg ata ctg aca cat tgt gcc tca gcc ttt ccc cac ctg ccc ggc 646 Lys Trp Ile Leu Thr His Cys Ala Ser Ala Phe Pro His Leu Pro Gly tgc tgc tgc tgc tgc ttt ttg ttg ctg ttc ttg gtg tct tcc gtc cct 694 Cys Cys Cys Cys Cys Phe Leu Leu Leu Phe Leu Val Ser Ser Val Pro gtc acc tgc caa gcc ctt ggt cag gac atg gtg tca cca gag gcc acc 742 Val Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu Ala Thr aac tct tct tcc tcc tcc ttc tcc tct cct tcc agc gcg gga agg cat 790 Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His gtg cgg agc tac aat cac ctt caa gga gat gtc cgc tgg aga aag cta 838 Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu ttc tct ttc acc aag tac ttt ctc aag att gag aag aac ggg aag gtc 886 Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val agc ggg acc aag aag gag aac tgc ccg tac agc atc ctg gag ata aca 934 Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu I1e Thr tca gta gaa atc gga gtt gtt gcc gtc aaa gcc att aac agc aac tat 982 Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr tac tta gcc atg aac aag aag ggg aaa ctc tat ggc tca aaa gaa ttt 1030 Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe aac aat gac tgt aag ctg aag gag agg ata gag gaa aat gga tac aat 1078 Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn acc tat gca tca ttt aac tgg cag cat aat ggg agg caa atg tat gtg 1126 Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val gca ttg aat gga aaa gga get cca agg aga gga cag aaa aca cga agg 1179 Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg aaa aac acc tct get cac ttt ctt cca atg gtg gta cac tca 1216 Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser tagaggaagg caacgtttgt ggatgcagta aaaccaatgg ctcttttgcc aagaatagtg 1276 gatattcttc atgaagacag tagattgaaa ggcaaagaca cgttgcagat gtctgcttgc 1336 ttaaaagaaa gccagccttt gaaggttttt gtattcactg ctgacatatg atgttctttt 1396 aattagttct gtgtcatgtc ttataatcaa gatataggca gatcgaatgg gatagaagtt 1456 attcccaagt gaaaaacatt gtggctgggt tttttgttgt tgttgtcaag tttttgtttt 1516 taaacctctg agatagaact taaaggacat agaacaatct gttgaaagaa cgatcttcgg 1576 gaaagttatt tatggaatac gaactcatat caaagacttc attgctcatt caagcctaat 1636 gaatcaatga acagtaatac gtgcaagcat ttactggaaa gcacttgggt catatcatat 1696 gcacaaccaa aggagttctg gatgtggtct catggaataa ttgaatagaa tttaaaaata 1756 taaacatgtt agtgtgaaac tgttctaaca atacaaatag tatggtatgc ttgtgcattc 1816 tgccttcatc cctttctatt tctttctaag ttatttattt aataggatgt taaatatctt 1876 ttggggtttt aaagagtatc tcagcagctg tcttctgatt tatcttttct ttttattcag 1936 cacaccacat gcatgttcac gacaaagtgt ttttaaaact tggcgaacac ttcaaaaata 1996 ggagttggga ttagggaagc agtatgagtg cccgtgtgct atcagttgac ttaatttgca 2056 cttctgcagt aataaccatc aacaataaat atggcaatgc tgtgccatgg cttgagtgag 2116 agatgtctgc tatcatttga aaacatatat tactctcgag gcttcctgtc tcaagaaata 2176 gaccagaagg ccaaattctt ctctttcaat acatcagttt gcctccaaga atatactaaa 2236 aaaaggaaaa ttaattgcta aatacattta aatagcctag cctcattatt tactcatgat 2296 ttcttgccaa atgtcatggc ggtaaagagg ctgtccacat ctctaaaaac cctctgtaaa 2356 ttccacataa tgcatctttc ccaaaggaac tataaagaat ttggtatgaa gcgcaactct 2416 cccaggggct taaactgagc aaatcaaata tatactggta tatgtgtaac catatacaaa 2976 aacctgttct agctgtatga tctagtcttt acaaaaccaa ataaaacttg ttttctgtaa 2536 atttaaagag ctttacaagg ttccataatg taaccatatc aaaattcatt ttgttagagc 2596 acgtatagaa aagagtacat aagagtttac caatcatcat cacattgtat tccactaaat 2656 aaatacataa gccttatttg cagtgtctgt agtgatttta aaaatgtaga aaaatactat 2716 ttgttctaaa tacttttaag caataactat aatagtatat tgatgctgca gttttatctt 2776 catatttctt gttttgaaaa agcattttat tgtttggaca cagtattttg gtacaaaaaa 2636 aaagactcac taaatgtgtc ttactaaagt ttaacctttg gaaatgctgg cgttctgtga 2896 ttctccaaca aacttatttg tgtcaatact taaccagcac ttccagttaa tctgttattt 2956 ttaaaaattg ctttattaag aaattttttg tataatccca taaaaggtca tatttttccc 3016 attcttcaaa aaaactgtat ttcagaagaa acacatttga ggcactgtct tttggcttat 3076 agtttaaatt gcatttcatc atactttgct tccaacttgc tttttggcaa atgagattat 3136 aaaaatgttt aatttttgtg gttggaatct ggatgttaaa atttaattgg taactcagtc 3196 tgtgagctat aatgtaatgc attcctatcc aaactaggta tctttttttc ctttatgttg 3256 aaataataat ggcacctgac acatagacat agaccaccca caacctaaat taaatgtttg 3316 gtaagacaaa tacacattgg atgaccacag taacagcaaa cagggcacaa actggattct 3376 tatttcacat agacatttag attactaaag agggctatgt gtaaacagtc atcattatag 3436 tactcaagac actaaaacag cttctagcca aatatattaa agcttgcaga ggccaaaaat 3996 agaaaacatc tcccctgtct ctcccacatt tccctcacag aaagacaaaa aacctgcctg 3556 gtgcagtagc tcacacctgt aatcccagca gtttgggaga ctgtgggaag atggcttgag 3616 tccaggagtt ctagacaggc ctgagaaacc tagtgagaca tccttctctt aaacaaaaca 3676 aaacaaaaca aatgtagcca tgcgtggtgg catatacctg tggtcccaac tactcaggag 3736 gctgaaacgg aaggatctct tgggccccag gagtttgagg ctgcagtgag ctataatctt 3796 gccattgcac tccagcctgg gtgaaaaaga gccagaaaga aaggaaagag agaaaagaga 3856 aaagaaagag agaaaagaca gaaagacagg aaggaaggaa ggaaggaagg aaggaaggaa 3916 ggaagcaagg aaagaaggaa ggaaggaaag.aagggaggga aggaaggaga gagaaagaaa 3976 gattgtttgg taaggagtaa tgacattctc ttgcatttaa aagtggcata tttgcttgaa 4036 atggaaatag aattctggtc ccttttgcaa ctactgaaga aaaaaaaaag cagtttcagc 4096 cctgaatgtt gtagatttga aaaaaaaaaa aaaaaaactc gagggggggc ccgtacccaa 4156 ttcgccctat agtgagtcgt a 4177 <210>24 <211>208 <212>PRT

<213>Homo Sapiens <900> 29 Met Trp Lys Trp Ile Leu Thr His Cys Ala Ser Ala Phe Pro His Leu Pro Gly Cys Cys Cys Cys Cys Phe Leu Leu Leu Phe Leu Val Ser Ser Val Pro Val Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu Ala Thr Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> 25 <211> 31 <212> PRT
<213> Homo sapiens <400> 25 Gly Gln Asp Met Val Ser Pro Glu Ala Thr Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His Val Arg Ser Tyr Asn <210> 26 <211> 19 <212> PRT
<213> Homo Sapiens <400> 26 Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser <210>27 <211>30 <212>PRT

<213>Homo sapiens <400> 27 Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr <210>28 <211>19 <212>PRT

<213>Homo Sapiens <400> 28 Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala <210> 29 <211> 555 <212> DNA
<213> Homo Sapiens <220>
<221> CDS
<222> (1)..(552) <400> 29 atg aga gga tcg cat cac cat cac cat cac gga tcc tgc cag get ctg 98 Met Arg Gly Ser His His His His His His Gly Ser Cys Gln Ala Leu ggt cag gac atg gtt tct ccg gaa get acc aac tct tcc tct tcc tct 96 Gly Gln Asp Met Val Ser Pro Glu Ala Thr Asn Ser Ser Ser Ser Ser ttc tct tcc ccg tct tcc get ggt cgt cac gtt cgt tct tac aac cac 144 Phe Ser Ser Pro Ser Ser Ala Gly Arg His Val Arg Ser Tyr Asn His ctg cag ggt gac gtt cgt tgg cgt aaa ctg ttc tct ttc acc aaa tac 192 Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr ttc ctg aaa atc gaa aaa aac ggt aaa gtt tct ggg acc aag aag gag 240 Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu aac tgc ccg tac agc atc ctg gag ata aca tca gta gaa atc gga gtt 288 Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val gtt gcc gtc aaa gcc att aac agc aac tat tac tta gcc atg aac aag 336 Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys aag ggg aaa ctc tat ggc tca aaa gaa ttt aac aat gac tgt aag ctg 389 Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu aag gag agg ata gag gaa aat gga tac aat acc tat gca tca ttt aac 432 Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn tgg cag cat aat ggg agg caa atg tat gtg gca ttg aat gga aaa gga 980 Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly get cca agg aga gga cag aaa aca cga agg aaa aac acc tct get cac 528 Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His ttt ctt cca atg gtg gta cac tca tag 555 Phe Leu Pro Met Val Val His Ser <210> 30 <211> 184 <212> PRT
<213> Homo Sapiens <900> 30 Met Arg Gly Ser His His His His His His Gly Ser Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu Ala Thr Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210>31 <211>84 <212>DNA

<213>Homo sapiens WO 99/41282 PC'T/US99/03018 <400> 31 atgtggaaat ggatactgac ccactgcgct tctgctttcc cgcacctgcc gggttgctgc 60 tgctgctgct tcctgctgct gttc gq <210> 32 <211> 82 <212> DNA
<213> Homo sapiens <400> 32 ccggagaaac catgtcctga cccagagcct ggcaggtaac cggaacagaa gaaaccagga 60 acagcagcag gaagcagcag ca 82 <210>33 <211>80 <212>DNA

<213>Homo Sapiens <400> 33 gggtcaggac atggtttctc cggaagctac caactcttct tcttcttctt tctcttctcc 60 gtcttctgct ggtcgtcacg g0 <210> 34 <211> 81 <212> DNA
<213> Homo Sapiens <900> 39 ggtgaaagag aacagtttac gccaacgaac gtcaccctgc aggtggttgt aagaacgaac 60 gtgacgacca gcagaagacg g 81 <210> 35 <211> 75 <212> DNA
<213> Homo Sapiens <400> 35 cgttggcgta aactgttctc tttcaccaaa tacttcctga aaatcgaaaa aaacggtaaa 60 gtttctggga ccaaa <210> 36 <211> 39 <212> DNA
<213> Homo sapiens <400> 36 tttggtccca gaaactttac cgtttttttc gattttcag 39 <210> 37 <211> 36 <212> DNA
<213> Homo sapiens <900> 37 aaaggatcca tgtggaaatg gatactgacc cactgc 36 <210> 38 <211> 627 <212> DNA
<213> Homo Sapiens <220>
<221> CDS
<222> (1)..(624) <400> 3B
atg tgg aaa tgg ata ctg acc cac tgc get tct get ttc ccg cac ctg 48 Met Trp Lys Trp Ile Leu Thr His Cys Ala Ser Ala Phe Pro His Leu ccg ggt tgc tgc tgc tgc tgc ttc ctg ctg ctg ttc ctg gtt tct tct 96 Pro Gly Cys Cys Cys Cys Cys Phe Leu Leu Leu Phe Leu Val Ser Ser gtt ccg gtt acc tgc cag get ctg ggt cag gac atg gtt tct ccg gaa 144 Val Pro Val Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu get acc aac tct tcc tct tcc tct ttc tct tcc ccg act tcc get ggt 192 Ala Thr Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Thr Ser Ala Gly cgt cac gtt cgt tct tac aac cac ctg cag ggt gac gtt cgt tgg cgt 290 Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg aaa ctg ttc tct ttc acc aaa tac ttc ctg aaa atc gaa aaa aac ggt 288 Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly aaa gtt tct ggg acc aag aag gag aac-tgc ccg tac agc atc ctg gag 336 WO 99/41282 PC'f/US99103018 Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu ata aca tca gta gaa atc gga gtt gtt gcc gtc aaa gcc att aac agc 384 Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser aac tat tac tta gcc atg aac aag aag ggg aaa ctc tat ggc tca aaa 432 Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys gaa ttt aac aat gac tgt aag ctg aag gag agg ata gag gaa aat gga 480 Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly tac aat acc tat gca tca ttt aac tgg cag cat aat ggg agg caa atg 528 Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met tat gtg gca ttg aat gga aaa gga get cca agg aga gga cag aaa aca 576 Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr cga agg aaa aac acc tct get cac ttt ctt cca atg gtg gta cac tca 624 Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser tag 627 <210>39 <211>208 <212>PRT

<213>Homo sapiens <400> 39 Met Trp Lys Trp Ile Leu Thr His Cys Ala Ser Ala Phe Pro His Leu Pro Gly Cys Cys Cys Cys Cys Phe Leu Leu Leu Phe Leu Val Ser Ser Val Pro Val Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu Ala Thr Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Thr Ser Ala Gly Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe-Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> 40 -<211> 38 <212> DNA
<213> Homo sapiens <400> 40 tttcatgact tgtcaagctc tgggtcaaga tatggttc 3g <210>41 <211>28 <212>DNA

<213>Homo sapiens <900> 41 gcccaagctt ccacaaacgt tgccttcc 2g <210> 42 <211> 525 <212> DNA
<213> Homo sapiens <220>
<221> CDS
<222> (1)..(522) <400> 42 atg acc tgc cag get ctg ggt cag gac atg gtt tct ccg gaa get acc 48 Met Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu Ala Thr aac tct tcc tct tcc tct ttc tct tcc ccg tct tcc get ggt cgt cac 96 Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His gtt cgt tct tac aac cac ctg cag ggt gac gtt cgt tgg cgt aaa ctg 144 Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu ttc tct ttc acc aaa tac ttc ctg aaa atc gaa aaa aac ggt aaa gtt 192 Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val tct ggg acc aag aag gag aac tgc ccg tac agc atc ctg gag ata aca 240 Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr 65 70 75 gp tca gta gaa atc gga gtt gtt gcc gtc aaa gcc att aac agc aac tat 288 Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr tac tta gcc atg aac aag aag ggg aaa ctc tat ggc tca aaa gaa ttt 336 Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe aac aat gac tgt aag ctg aag gag agg ata gag gaa aat gga tac aat 384 Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn acc tat gca tca ttt aac tgg cag cat aat ggg agg caa atg tat gtg 432 Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val gca ttg aat gga aaa gga get cca agg aga gga cag aaa aca cga agg 480 Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg aaa aac acc tct get cac ttt ctt cca atg gtg gta cac tca tag 525 Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> 43 <211> 174 <212> PRT
<213> Homo Sapiens <400> 43 Met Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu Ala Thr Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr 85 . 90 95 Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> 44 <211> 45 <212> DNA
<213> Homo Sapiens <900> 99 tcagtgaatt cattaaagag gagaaattaa tcatgacttg ccagg 45 <210>95 <211>48 <212>DNA

<213>Homo Sapiens <400> 45 tcatgacttg ccaggcactg ggtcaagaca tggtttcccc ggaagcta 98 <210> 46 <211> 48 <212> DNA
<213> Homo Sapiens <400> 46 gcttcagcag cccatctagc gcaggtcgtc acgttcgctc ttacaacc 48 <210> 47 <211> 48 <212> DNA
<213> Homo Sapiens <400> 47 gttcgttggc gcaaactgtt cagctttacc aagtacttcc tgaaaatc 98 <210>48 <211>28 <212>DNA

<213>Homo Sapiens <400> 48 tcgaaaaaaa cggtaaagtt tctgggac 2g <210>49 <211>48 <212>ANA

<213>Homo sapiens <400> 49 gatgggctgc tgaagctaga gctggagctg ttggtagctt ccggggaa 48 <210>50 <211>45 <212>DNA

<213>Homo Sapiens <400> 50 aacagtttgc gccaacgaac atcaccctgt aagtggttgt aagag 45 <210>51 <211>47 <212>DNA

<213>Homo sapiens <400> 51 ttcttggtcc cagaaacttt accgtttttt tcgattttca ggaagta 47 <210>52 <211>24 <212>DNA

<213>Homo Sapiens <400> 52 ttcttggtcc cagaaacttt accg 24 <210> 53 <2I1> 45 <212> DNA
<213> Homo Sapiens <900> 53 agatcaggct tctattatta tgagtgtacc accattggaa gaaag 45 <210>59 <211>525 <212>DNA

<213>Homo sapiens <220>
<221> CDS
<222> (1)..(522) <400> 54 atg act tgc cag gca ctg ggt caa gac atg gtt tcc ccg gaa get acc 48 Met Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu Ala Thr aac agc tcc agc tct agc ttc agc agc cca tct agc gca ggt cgt cac 96 Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His gtt cgc tct tac aac cac tta cag ggt gat gtt cgt tgg cgc aaa ctg 144 Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu ttc agc ttt acc aag tac ttc ctg aaa atc gaa aaa aac ggt aaa gtt 192 Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val tct ggg acc aag aag gag aac tgc ccg tac agc atc ctg gag ata aca 240 Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr tca gta gaa atc gga gtt gtt gcc gtc aaa gcc att aac agc aac tat 2B8 Ser Val Glu Ile Gly Val Val Ala Val Lys AIa Ile Asn Ser Asn Tyr tac tta gcc atg aac aag aag ggg aaa ctc tat ggc tca aaa gaa ttt 336 Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe aac aat gac tgt aag ctg aag gag agg ata gag gaa aat gga tac aat 389 Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn acc tat gca tca ttt aac tgg cag cat aat ggg agg caa atg tat gtg 432 Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val gca ttg aat gga aaa gga get cca agg aga gga cag aaa aca cga agg 480 Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg aaa aac acc tct get cac ttt ctt cca atg gtg gta cac tca tag 525 Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210>55 <211>179 <212>PRT

<213>Homo Sapiens <900> 55 Met Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu Ala Thr Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val VaI Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210>56 <211>35 <212>DNA

<213>Homo sapiens <400> 56 ggaccctcat gacctgccag gctctgggtc aggac 35 <210> 57 <211> 28 <212> DNA
<213> Homo sapiens <400> 57 ggacagccat ggctggtcgt cacgttcg 2g <210>58 <211>29 <212>DNA

<213>Homo sapiens <400> 58 ggacagccat ggttcgttgg cgtaaactg 2g <210>59 <211>31 <212>DNA

<213>Homo sapiens <400> 59 ggacagccat ggaaaaaaac ggtaaagttt c 31 <210> 60 <211> 29 <212> DNA
<213> Homo sapiens <400> 60 ggacccccat ggagaactgc ccgtagagc 29 <210> 61 <211> 32 <212> DNA
<213> Homo sapiens <900> 61 ggacccccat ggtcaaagcc attaacagca ac 32 <210>62 <211>33 <212>DNA

<213>Homo Sapiens <400> 62 ggacccccat ggggaaactc tatggctcaa aag 33 <210> 63 <211> 37 <212> DNA
<213> Homo Sapiens <400> 63 ctgcccaagc ttattatgag tgtaccacca ttggaag 37 <210> 64 <211> 36 <212> DNA
<213> Homo Sapiens <400> 64 ctgcccaagc ttattacttc agcttacagt cattgt 36 <210> 65 <212> 525 <212> DNA
<213> Homo sapiens <220>
<221> CDS
<222> (1) .. (522) <400> 65 atg acc tgc cag get ctg ggt cag gac atg gtt tct ccg gaa get acc 48 Met Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu Ala Thr aac tct tcc tct tcc tct ttc tct tcc ccg tct tcc get ggt cgt cac 96 Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His gtt cgt tct tac aac cac ctg cag ggt gac gtt cgt tgg cgt aaa ctg 144 Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu ttc tct ttc acc aaa tac ttc ctg aaa atc gaa aaa aac ggt aaa gtt 192 Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val tct ggg acc aag aag gag aac tgc ccg tac agc atc ctg gag ata aca 240 Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr 65 70 75 g0 tca gta gaa atc gga gtt gtt gcc gtc aaa gcc att aac agc aac tat 288 Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr tac tta gcc atg aac aag aag ggg aaa ctc tat ggc tca aaa gaa ttt 336 Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe aac aat gac tgt aag ctg aag gag agg ata gag gaa aat gga tac aat 389 Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn acc tat gca tca ttt aac tgg cag cat aat ggg agg caa atg tat gtg 432 Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val gca ttg aat gga aaa gga get cca agg aga gga cag aaa aca cga agg 480 Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg aaa aac acc tct get cac ttt ctt cca atg gtg gta cac tca tag 525 Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> 66 <211> 174 <212> PRT
<213> Homo Sapiens <900> 66 Met Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu Ala Thr Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <2I0> 67 <211> 499 <212> DNA
<213> Homo Sapiens <220>
<221> CDS
<222> (1)..(441) <400> 67 atg get ggt cgt cac gtt cgt tct tac aac cac ctg cag ggt gac gtt 48 Met Ala Gly Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val cgt tgg cgt aaa ctg ttc tct ttc acc aaa tac ttc ctg aaa atc gaa 96 Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu aaa aac ggt aaa gtt tct ggg acc aag aag gag aac tgc ccg tac agc 144 Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser atc ctg gag ata aca tca gta gaa atc gga gtt gtt gcc gtc aaa gcc 192 Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala att aac agc aac tat tac tta gcc atg aac aag aag ggg aaa ctc tat 290 Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr ggc tca aaa gaa ttt aac aat gac tgt aag ctg aag gag agg ata gag 288 Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu gaa aat gga tac aat acc tat gca tca ttt aac tgg cag cat aat ggg 336 Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly agg caa atg tat gtg gca ttg aat gga aaa gga get cca agg aga gga 384 Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly cag aaa aca cga agg aaa aac acc tct get cac ttt ctt cca atg gtg 432 Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val gta cac tca tag 444 Val His Ser <210> 68 <211> 147 <212> PRT
<213> Homo Sapiens <400> 68 Met Ala Gly Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Giu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> 69 <211> 402 <212> DNA
<213> Homo Sapiens <220>
<221> CDS
<222> (1)..(399) <400> 69 atg gtt cgt tgg cgt aaa ctg ttc tct ttc acc aaa tac ttc ctg aaa 48 Met Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys atc gaa aaa aac ggt aaa gtt tct ggg acc aag aag gag aac tgc ccg 96 Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro tac agc atc ctg gag ata aca tca gta gaa atc gga gtt gtt gcc gtc 149 Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val aaa gcc att aac agc aac tat tac tta gcc atg aac aag aag ggg aaa 192 Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys ctc tat ggc tca aaa gaa ttt aac aat gac tgt aag ctg aag gag agg 240 Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg ata gag gaa aat gga tac aat acc tat gca tca ttt aac tgg cag cat 288 Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His aat ggg agg caa atg tat gtg gca ttg aat gga aaa gga get cca agg 336 Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg WO 99!41282 PCT/US99103018 aga gga cag aaa aca cga agg aaa aac acc tct get cac ttt ctt cca 384 Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro atg gtg gta cac tca tag 402 Met Val Val His Ser <210> 70 <211> 133 <212> PRT
<213> Homo sapiens <400> 70 Met Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210>71 <211>354 <212>DNA

<213>Homo sapiens <220>
<221> CDS
<222> (1)..(351) <400> 71 atg gaa aaa aac ggt aaa gtt tct ggg acc aag aag gag aac tgc ccg 48 Met Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro tac agc atc ctg gag ata aca tca gta gaa atc gga gtt gtt gcc gtc 96 Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val aaa gcc att aac agc aac tat tac tta gcc atg aac aag aag ggg aaa 144 Lys Ala Ile_ Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys ctc tat ggc tca aaa gaa ttt aac aat gac tgt aag ctg aag gag agg 192 Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg ata gag gaa aat gga tac aat acc tat gca tca ttt aac tgg cag cat 290 Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His aat ggg agg caa atg tat gtg gca ttg aat gga aaa gga get cca agg 288 Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg aga gga cag aaa aca cga agg aaa aac acc tct get cac ttt ctt cca 336 Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro atg gtg gta cac tca tag 354 Met Val Val His Ser <210> 72 <211> 117 <212> PRT
<213> Homo Sapiens <400> 72 Met Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His 65 70 75 g0 Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210>73 <211>321 <212>DNA

<213>Homo Sapiens <220>
<221> CDS
<222> (1)..(318) <400> 73 atg gag aac tgc ccg tac agc atc ctg gag ata aca tca gta gaa atc 48 Met Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile gga gtt gtt gcc gtc aaa gcc att aac agc aac tat tac tta gcc atg 96 Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met aac aag aag ggg aaa ctc tat ggc tca aaa gaa ttt aac aat gac tgt 149 Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys aag ctg aag gag agg ata gag gaa aat gga tac aat acc tat gca tca 192 Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser ttt aac tgg cag cat aat ggg agg caa atg tat gtg gca ttg aat gga 290 Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly aaa gga get cca agg aga gga cag aaa aca cga agg aaa aac acc tct 288 Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser get cac ttt ctt cca atg gtg gta cac tca tag 321 Ala His Phe Leu Pro Met Val Val His Ser WO 99/41282 PC'T/US99/03018 <2I0> 74 <211> 106 <212> PRT
<213> Homo sapiens <400> 74 Met Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Aia Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly 65 70 75 gp Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser 85 90 g5 Ala His Phe Leu Pro Met Val Val His Ser <210> 75 <211> 264 <212> DNA
<213> Homo sapiens <220>
<221> CDS
<222> (1)..(261) <900> 75 atg gtc aaa gcc att aac agc aac tat tac tta gcc atg aac aag aag 98 Met Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys ggg aaa ctc tat ggc tca aaa gaa ttt aac aat gac tgt aag ctg aag 96 Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys gag agg ata gag gaa aat gga tac aat acc tat gca tca ttt aac tgg 194 Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp cag cat aat ggg agg caa atg tat gtg gca ttg aat gga aaa gga get 192 Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala WO 99/41282 PC'TIUS99/03018 cca agg aga gga cag aaa aca cga agg aaa aac acc tct get cac ttt 240 Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe ctt cca atg gtg gta cac tca tag 264 Leu Pro Met Val Val His Ser <210> 76 <211> 87 <212> PRT
<213> Homo Sapiens <400> 76 Met Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys GlyLys LeuTyrGly SerLysGlu PheAsnAsnAsp CysLys Leu Lys GluArg IleGluGlu AsnGlyTyr AsnThrTyrAla SerPhe Asn Trp GlnHis AsnGlyArg GlnMetTyr ValAlaLeuAsn GlyLys Gly Ala ProArg ArgGlyGln LysThrArg ArgLysAsnThr SerAla His Phe 65 70 75 g0 LeuPro MetValVal HisSer <210> 77 <211> 219 <212> DNA
<213> Homo sapiens <220>
<221> CDS
<222> (1)..(216) <400> 77 atg ggg aaa ctc tat ggc tca aaa gaa ttt aac aat gac tgt aag ctg 48 Met Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu aag gag agg ata gag gaa aat gga tac aat acc tat gca tca ttt aac 96 Lys Glu Arg Ile Glu Glu Asn Gly Tyr-Asn Thr Tyr Ala Ser Phe Asn WO 99/41282 ' PCTIUS99/03018 tgg cag cat aat ggg agg caa atg tat gtg gca ttg aat gga aaa gga 144 Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly get cca agg aga gga cag aaa aca cga agg aaa aac acc tct get cac 192 Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His ttt ctt cca atg gtg gta cac tca tag 219 Phe Leu Pro Met Val Val His Ser <210> 78 <211> 72 <212> PRT
<213> Homo sapiens <400> 78 Met Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys-Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> 79 <211> 357 <212> DNA
<213> Homo sapiens <220>
<221> CDS
<222> (1)..(357) <400> 79 atg acc tgc cag get ctg ggt cag gac atg gtt tct ccg gaa get acc 48 Met Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu Ala Thr aac tct tcc tct tcc tct ttc tct tcc ccg tct tcc get ggt cgt cac 96 Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His gtt cgt tct tac aac cac ctg cag ggt gac gtt cgt tgg cgt aaa ctg 144 Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu ttc tct ttc acc aaa tac ttc ctg aaa atc gaa aaa aac ggt aaa gtt 192 Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val tct ggg acc aag aag gag aac tgc ccg tac agc atc ctg gag ata aca 290 Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr tca gta gaa atc gga gtt gtt gcc gtc aaa gcc att aac agc aac tat 288 Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr tac tta gcc atg aac aag aag ggg aaa ctc tat ggc tca aaa gaa ttt 336 Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe aac aat gac tgt aag ctg aag 357 Asn Asn Asp Cys Lys Leu Lys <210> 80 <211> 119 <212> PRT
<213> Homo sapiens <400> 80 Met Thr Cys Gln Ala Leu Gly Gln Asp Met Val Ser Pro Glu Ala Thr Asn Ser Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys <210>81 <211>276 <212>DNA

<213>Homo Sapiens <220>
<221> CDS
<222> (1}..(276}
<400> 81 atg get ggt cgt cac gtt cgt tct tac aac cac ctg cag ggt gac gtt 48 Met Ala Gly Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val cgt tgg cgt aaa ctg ttc tct ttc acc aaa tac ttc ctg aaa atc gaa 96 Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu aaa aac ggt aaa gtt tct ggg acc aag aag gag aac tgc ccg tac agc 149 Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser 35 40 , 45 atc ctg gag ata aca tca gta gaa atc gga gtt gtt gcc gtc aaa gcc 192 Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala att aac agc aac tat tac tta gcc atg aac aag aag ggg aaa ctc tat 240 Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr 65 70 75 gp ggc tca aaa gaa ttt aac aat gac tgt aag ctg aag 276 Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys <210>82 <211>92 <212>PRT

<213>Homo Sapiens <400> 82 Met Ala Gly Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys <210> 83 <211> 525 <212> DNA
<213> Homo Sapiens <400> B3 atgacctctc aggctctggg tcaggacatg gtttctccgg aagctaccaa ctcttcctct 60 tcctctttct cttccccgtc ttccgctggt cgtcacgttc gttcttacaa ccacctgcag 120 ggtgacgttc gttggcgtaa actgttctct ttcaccaaat acttcctgaa aatcgaaaaa 180 aacggtaaag tttctgggac caagaaggag aactctccgt acagcatcct ggagataaca 240 tcagtagaaa tcggagttgt tgccgtcaaa gccattaaca gcaactatta cttagccatg 300 aacaagaagg ggaaactcta tggctcaaaa gaatttaaca atgactgtaa gctgaaggag 360 aggatagagg aaaatggata caatacctat gcatcattta actggcagca taatgggagg 420 caaatgtatg tggcattgaa tggaaaagga gctccaagga gaggacagaa aacacgaagg 980 aaaaacacct ctgctcactt tcttccaatg gtggtacact catag 525 <210> 84 <211> 525 <212> DNA
<213> Homo sapiens <400> 84 atgacctgcc aggctctggg tcaggacatg gtttctccgg aagctaccaa ctcttcctct 60 tcctctttct cttccccgtc ttccgctggt cgtcacgttc gttcttacaa ccacctgcag 120 ggtgacgttc gttggcgtaa actgttctct ttcaccaaat acttcctgaa aatcgaaaaa 180 WO 99!41282 PCT/US99/03018 -4b-aacggtaaag tttctgggac caagaaggag aactctccgt acagcatcct ggagataaca 240 tcagtagaaa tcggagttgt tgccgtcaaa gccattaaca gcaactatta cttagccatg 300 aacaagaagg ggaaactcta tggctcaaaa gaatttaaca atgactgtaa gctgaaggag 360 aggatagagg aaaatggata caatacctat gcatcattta actggcagca taatgggagg 420 caaatgtatg tggcattgaa tggaaaagga gctccaagga gaggacagaa aacacgaagg 480 aaaaacacct ctgctcactt tcttccaatg gtggtacact catag 525 <210>85 <211>29 <212>DNA

<213>Homo Sapiens <900> 85 ggaccctcat gacctctcag gctctgggt 2g <210> 86 <211> 21 <212> DNA
<213> Homo Sapiens <400> 86 aaggagaact ctccgtacag c 21 <210> 87 <211> 21 <212> DNA
<213> Homo Sapiens <400> 87 gctgtacggt ctgttctcct t 21 <210>88 <211>35 <212>DNA

<213>Homo Sapiens <400> 88 ggaccctcat gacctgccag gctctgggtc aggac <210> 89 <211> 37 <212> DNA

<213> Homo Sapiens <900> 89 ctgcccaagc ttattatgag tgtaccacca ttggaag 3~
<210> 90 <211> 33 <212> DNA
<213> Homo sapiens <400> 90 aaaggatcct gccaggctct gggtcaggac atg 33 <210>91 <211>32 <212>DNA

<213>Homo sapiens <900> 91 gcggcacatg tcttacaacc acctgcaggg tg 32 <210> 92 <211> 28 <212> DNA
<213> Homo Sapiens <400> 92 gggcccaagc ttatgagtgt accaccat 2g <210> 93 <211> 36 <212> DNA
<213> Homo Sapiens <400> 93 ccggcggatc ccatatgtct tacaaccacc tgcagg 36 <210>94 <211>35 <212>DNA

<213>Homo Sapiens <400> 99 ccggcggtac cttattatga gtgtaccacc attgg 35 <210> 95 <211> 426 <212> DNA
<213> Homo sapiens <400> 95 atgtcttaca accacctgca gggtgacgtt cgttggcgta aactgttctc tttcaccaaa 60 tacttcctga aaatcgaaaa aaacggtaaa gtttctggga ccaagaagga gaactgcccg 120 tacagcatcc tggagataac atcagtagaa atcggagttg ttgccgtcaa agccattaac 180 agcaactatt acttagccat gaacaagaag gggaaactct atggctcaaa agaatttaac 240 aatgactgta agctgaagga gaggatagag gaaaatggat acaataccta tgcatcattt 300 aactggcagc ataatgggag gcaaatgtat gtggcattga atggaaaagg agctccaagg 360 agaggacaga aaacacgaag gaaaaacacc tctgctcact ttcttccaat ggtggtacac 920 tcataa 426 <210>96 <211>141 <212>PRT

<213>Homo Sapiens <400> 96 Met Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr 85 ' 90 95 Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> 97 <211> 20 <212> DNA
<213> Homo Sapiens <400> 97 caaccacctg cagggtgacg <210>98 <211>78 <212>DNA

<213>Homo Sapiens <900> 98 aacggtcgac aaatgtatgt ggcactgaac ggtaaaggtg ctccacgtcg tggtcagaaa 60 acccgtcgta aaaacacc 78 <210> 99 -<211> 76 <212> DNA
<213> Homo Sapiens <400> 99 gggcccaagc ttaagagtgt accaccattg gcagaaagtg agcagaggtg tttttacgac 60 gggttttctg accacg 76 <210> 100 <211> 23 <212> DNA
<213> Homo sapiens <400> 100 gccacataca tttgtcgacc gtt 23 <210>101 <211>19 <212>DNA

<213>Homo sapiens <400> 101 gggcccaagc ttaagagtg lg -SO-<210> 102 <211> 23 <212> DNA
<213> Homo sapiens <900> 102 gccacataca tttgtcgacc gtt 23 <210>103 <211>90 <212>DNA

<213>Homo Sapiens <400> 103 ctgcagggtg acgttcgttg gcgtaaactg ttctccttca ccaaatactt cctgaaaatc 60 gaaaaaaacg gtaaagtttc tggtaccaag g0 <210>104 <211>90 <212>DNA

<213>Homo sapiens <400> 104 agctttaaca gcaacaacac cgatttcaac ggaggtgatt tccaggatgg agtacgggca 60 gttttctttc ttggtaccag aaactttacc g0 <210>105 <211>90 <212>DNA

<213>Homo Sapiens <400> 105 ggtgttgttg ctgttaaagc tatcaactcc aactactacc tggctatgaa caagaaaggt 60 aaactgtacg gttccaaaga atttaacaac g0 <210>106 <211>100 <212>DNA

<213>Homo Sapiens <400> 106 gtcgaccgtt gtgctgccag ttgaaggaag cgtaggtgtt gtaaccgttt tcttcgatac 60 gttctttcag tttacagtcg ttgttaaatt ctttggaacc 100 <210>107 <211>25 <212>DNA

<213>Homo Sapiens <400> 107 gcggcgtcga ccgttgtgct gccag <210> 108 <211> 26 <212> DNA
<213> Homo Sapiens <900> 108 gcggcctgca gggtgacgtt cgttgg 26 <210>109 <211>36 <212>DNA

<213>Homo Sapiens <400> 109 ccggcggatc ccatatgtct tacaaccacc tgcagg 36 <210>110 <211>39 <212>DNA

<213>Homo Sapiens <400> 110 cgcgcgatat cttattaaga gtgtaccacc attg 34 <210>111 <211>426 <212>DNA

<213>Homo Sapiens <400> 111 atgtcttaca accacctgca gggtgacgtt cgttggcgta aactgttctc cttcaccaaa 60 tacttcctga aaatcgaaaa aaacggtaaa gtttctggta ccaagaaaga aaactgcccg 120 tactccatcc tggaaatcac ctccgttgaa atcggtgttg ttgctgttaa agctatcaac 180 tccaactact acctggctat gaacaagaaa ggtaaactgt acggttccaa agaatttaac 240 WO 99141282 PC'T/US99/03018 aacgactgta aactgaaaga acgtatcgaa gaaaacggtt acaacaccta cgcttccttc 300 aactggcagc acaacggtcg acaaatgtat gtggcactga acggtaaagg tgctccacgt 360 cgtggtcaga aaacccgtcg taaaaacacc tctgctcact ttctgccaat ggtggtacac 420 tcttaa 426 <210>112 <211>141 <212>PRT

<213>Homo Sapiens <400> 112 Met Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser 20 25 ~ 30 G1y Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> 113 <211> 28 <212> DNA
<213> Homo sapiens <400> 113 cgcggccatg gctctgggtc aggacatg 2g <210> 114 <211> 28 <212> DNA
<213> Homo Sapiens <400> 114 gggcccaagc ttatgagtgt accaccat 28 <210> 115 <211> 516 <212> DNA
<213> Homo Sapiens <400> 115 atggctctgg gtcaagatat ggtttctccg gaagctacca actcttcctc ttcctctttc 60 tcttccccgt cttccgctgg tcgtcacgtt cgttcttaca accacctgca gggtgacgtt 120 cgttggcgta aactgttctc tttcaccaaa tacttcctga aaatcgaaaa aaacggtaaa 180 gtttctggga ccaagaagga gaactgcccg tacagcatcc tggagataac atcagtagaa 240 atcggagttg ttgccgtcaa agccattaac agcaactatt acttagccat gaacaagaag 300 gggaaactct atggctcaaa agaatttaac aatgactgta agctgaagga gaggatagag 360 gaaaatggat acaataccta tgcatcattt aactggcagc ataatgggag gcaaatgtat 420 gtggcattga atggaaaagg agctccaagg agaggacaga aaacacgaag gaaaaacacc 480 tctgctcact ttcttccaat ggtggtacac tcataa 516 <210> 116 <211> 171 <212> PRT
<213> Homo sapiens <400> 116 Met Ala Leu Gly Gln Asp Met Val Ser Pro Glu Ala Thr Asn Sex Ser Ser Ser Ser Phe Ser Ser Pro Ser Ser Ala Gly Arg His Val Arg Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210>117 <211>32 <212>DNA

<213>Homo sapiens <400> 117 gcggcacatg tcttacaacc acctgcaggg tg 32 <210> 118 <211> 75 <212> DNA
<213> Homo Sapiens <400> 118 ctgcccaagc ttttatgagt gtaccaccat tggaagaaag tgagcagagg tgtttttttc 60 tcgtgttttc tgtcc 7S
<210>119 <211>426 <212>DNA

<213>Homo Sapiens <400> 119 atgtcttaca accacctgca gggtgacgtt cgttggcgta aactgttctc tttcaccaaa 60 tacttcctga aaatcgaaaa aaacggtaaa gtttctggga ccaagaagga gaactgcccg 120 tacagcatcc tggagataac atcagtagaa atcggagttg ttgccgtcaa agccattaac 180 agcaactatt acttagccat gaacaagaag gggaaactct atggctcaaa agaatttaac 240 aatgactgta agctgaagga gaggatagag gaaaatggat acaataccta tgcatcattt 300 aactggcagc ataatgggag gcaaatgtat gtggcattga atggaaaagg agctccaagg 360 agaggacaga aaacacgaga aaaaaacacc tctgctcact ttcttccaat ggtggtacac 420 tcatag 926 <210>120 <211>191 <212>PRT

<213>Homo sapiens <900> 120 Met Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Glu Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210>121 <211>32 <2I2>DNA

<213>Homo Sapiens WO 99141282 PC1'/US99/03018 <400> 121 gcggcacatg tcttacaacc acctgcaggg tg 32 <210>122 <211>75 <212>DNA

<213>Homo Sapiens <400> 122 ctgcccaagc ttttatgagt gtaccaccat tggaagaaag tgagcagagg tgtttttctg 60 tcgtgttttc tgtcc 75 <210>123 <211>426 <212>DNA

<213>Homo Sapiens <900> 123 atgtcttaca accacctgca gggtgacgtt cgttggcgta aactgttctc tttcaccaaa 60 tacttcctga aaatcgaaaa aaacggtaaa gtttctggga ccaagaagga gaactgcccg 120 tacagcatcc tggagataac atcagtagaa atcggagttg ttgccgtcaa agccattaac 180 agcaactatt acttagccat gaacaagaag gggaaactct atggctcaaa agaatttaac 290 aatgactgta agctgaagga gaggatagag gaaaatggat acaataccta tgcatcattt 300 aactggcagc ataatgggag gcaaatgtat gtggcattga atggaaaagg agctccaagg 360 agaggacaga aaacacgaca gaaaaacacc tctgctcact ttcttccaat ggtggtacac 920 tcatag 426 <210>124 <211>191 <212>PRT

<213>Homo Sapiens <400> 129 Met Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr 85 90 g5 Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Gln Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210>125 <211>32 <212>DNA

<213>Homo sapiens <400> 125 gcggcacatg tcttacaacc acctgcaggg tg 32 <210>126 <211>84 <212>DNA

<213>Homo Sapiens <900> 126 ctgcccaagc ttttatgagt gtaccaccat tggaagaaag tgagcagagg tgtttttcct 60 tcgtgtttcc tgtcctctcc ttgg g4 <210> 127 <211> 926 <212> DNA
<213> Homo Sapiens <400> 127 atgtcttaca accacctgca gggtgacgtt cgttggcgta aactgttctc tttcaccaaa 60 tacttcctga aaatcgaaaa aaacggtaaa gtttctggga ccaagaagga gaactgcccg 120 tacagcatcc tggagataac atcagtagaa atcggagttg ttgccgtcaa agccattaac 180 agcaactatt acttagccat gaacaagaag gggaaactct atggctcaaa agaatttaac 290 aatgactgta agctgaagga gaggatagag gaaaatggat acaataccta tgcatcattt 300 aactggcagc ataatgggag gcaaatgtat gtggcattga atggaaaagg agctccaagg 360 agaggacagg aaacacgaag gaaaaacacc tctgctcact ttcttccaat ggtggtacac 920 tcatag <210>128 <211>141 <212>PRT

<213>Homo sapiens <400> 128 Met Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser 35 40 q5 Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Glu Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> 129 <211> 32 <212> DNA
<213> Homo Sapiens <900> 129 gcggcacatg tcttacaacc acctgcaggg tg 32 WO 99/41282 PC'T/US99/03018 <230>130 <211>84 <212>DNA

<213>Homo Sapiens <400> 130 ctgcccaagc ttttatgagt gtaccaccat tggaagaaag tgagcagagg tgtttttcct 60 tcgtgtctgc tgtcctctcc ttgg 84 <210> 131 <211> 426 <212> DNA
<213> Homo sapiens <400> 131 atgtcttaca accacctgca gggtgacgtt cgttggcgta aactgttctc tttcaccaaa 60 tacttcctga aaatcgaaaa aaacggtaaa gtttctggga ccaagaagga gaactgcccg 120 tacagcatcc tggagataac atcagtagaa atcggagttg ttgccgtcaa agccattaac 180 agcaactatt acttagccat gaacaagaag gggaaactct atggctcaaa agaatttaac 240 aatgactgta agctgaagga gaggatagag gaaaatggat acaataccta tgcatcattt 300 aactggcagc ataatgggag gcaaatgtat gtggcattga atggaaaagg agctccaagg 360 agaggacagc agacacgaag gaaaaacacc tctgctcact ttcttccaat ggtggtacac 420 tcatag 426 <210> 132 <211> 141 <212> PRT
<213> Homo Sapiens <400> 132 Met Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Arg Gly Gln Gln Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> i33 <211> 32 <212> DNA
<2I3> Homo Sapiens <400> 133 gcggcacatg tcttacaacc acctgcaggg tg 32 <210>134 <211>93 <212>DNA

<213>Homo sapiens <900> 139 ctgcccaagc ttttatgagt gtaccaccat tggaagaaag tgagcagagg tgtttttcct 60 tcgtgttttc tgtccttccc ttggagctcc ttt g3 <210> 135 <211> 426 <212> DNA
<213> Homo Sapiens <400> 135 atgtcttaca accacctgca gggtgacgtt cgttggcgta aactgttctc tttcaccaaa 60 tacttcctga aaatcgaaaa aaacggtaaa gtttctggga ccaagaagga gaactgcccg 120 tacagcatcc tggagataac atcagtagaa atcggagttg ttgccgtcaa agccattaac 180 agcaactatt acttagccat gaacaagaag gggaaactct atggctcaaa agaatttaac 290 aatgactgta agctgaagga gaggatagag gaaaatggat acaataccta tgcatcattt 300 aactggcagc ataatgggag gcaaatgtat gtggcattga atggaaaagg agctccaagg 360 gaaggacaga aaacacgaag gaaaaacacc tctgctcact ttcttccaat ggtggtacac 420 tcatag <210> 136 <211> 140 <212> PRT
<213> Homo sapiens <400> 136 Met Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Glu Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> 137 <211> 32 <212> DNA
<213> Homo sapiens <400> 137 gcggcacatg tcttacaacc acctgcaggg tg 32 <210> 138 <211> 93 <212> DNA
<213> Homo sapiens <400> 138 ctgcccaagc ttttatgagt gtaccaccat tggaagaaag tgagcagagg tgtttttcct 60 tcgtgttttc tgtccctgcc ttggagctcc ttt g3 <210> 139 <211> 426 <212> DNA
<213> Homo sapiens <900> 139 atgtcttaca accacctgca gggtgacgtt cgttggcgta aactgttctc tttcaccaaa 60 tacttcctga aaatcgaaaa aaacggtaaa gtttctggga ccaagaagga gaactgcccg 120 tacagcatcc tggagataac atcagtagaa atcggagttg ttgccgtcaa agccattaac 180 agcaactatt acttagccat gaacaagaag gggaaactct atggctcaaa agaatttaac 240 aatgactgta agctgaagga gaggatagag gaaaatggat acaataccta tgcatcattt 300 aactggcagc ataatgggag gcaaatgtat gtggcattga atggaaaagg agctccaagg 360 cagggacaga aaacacgaag gaaaaacacc tctgctcact ttcttccaat ggtggtacac 420 tcatag 426 <210> 190 <211> 141 <212> PRT
<213> Homo sapiens <900> 140 Met Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Lys Gly Ala Pro Arg Gln Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> 141 <211> 32 <212> DNA
<213> Homo sapiens <400> 141 gcggcacatg tcttacaacc acctgcaggg tg 32 <210>142 <211>21 <212>DNA

<213>Homo Sapiens <400> 142 ttgaatggag aaggagctcc a 21 <210> 143 <211> 21 <212> DNA
<213> Homo Sapiens <400> 143 tggagctcct tctccattca a 21 <210> 199 <211> 33 <212> DNA
<213> Homo Sapiens <400> 194 ctgcccaagc ttttatgagt gtaccaccat tgg 33 <210> 195 <211> 426 <212> DNA
<213> Homo Sapiens <400> 145 atgtcttaca accacctgca gggtgacgtt cgttggcgta aactgttctc tttcaccaaa 60 tacttcctga aaatcgaaaa aaacggtaaa gtttctggga ccaagaagga gaactgcccg 120 tacagcatcc tggagataac atcagtagaa atcggagttg ttgccgtcaa agccattaac i80 agcaactatt acttagccat gaacaagaag gggaaactct atggctcaaa agaatttaac 240 aatgactgta agctgaagga gaggatagag gaaaatggat acaataccta tgcatcattt 300 aactggcagc ataatgggag gcaaatgtat gtggcattga atggagaagg agctccaagg 360 agaggacaga aaacacgaag gaaaaacacc tctgctcact ttcttccaat ggtggtacac 420 tcatag <210> 146 <211> 141 <212> PRT
<213> Homo Sapiens <400> 146 Met Ser Tyr Asn His Leu Gln Gly Asp Val Arg Trp Arg Lys Leu Phe Ser Phe Thr Lys Tyr Phe Leu Lys Ile Glu Lys Asn Gly Lys Val Ser Gly Thr Lys Lys Glu Asn Cys Pro Tyr Ser Ile Leu Glu Ile Thr Ser Val Glu Ile Gly Val Val Ala Val Lys Ala Ile Asn Ser Asn Tyr Tyr Leu Ala Met Asn Lys Lys Gly Lys Leu Tyr Gly Ser Lys Glu Phe Asn Asn Asp Cys Lys Leu Lys Glu Arg Ile Glu Glu Asn Gly Tyr Asn Thr Tyr Ala Ser Phe Asn Trp Gln His Asn Gly Arg Gln Met Tyr Val Ala Leu Asn Gly Glu Gly Ala Pro Arg Arg Gly Gln Lys Thr Arg Arg Lys Asn Thr Ser Ala His Phe Leu Pro Met Val Val His Ser <210> 147 <211> 3974 <212> DNA
<213> Homo sapiens <400> 147 ggtacctaag tgagtagggc gtccgatcga cggacgcctt ttttttgaat tcgtaatcat 60 ggtcatagct gtttcctgtg tgaaattgtt atccgctcac aattccacac aacatacgag 120 ccggaagcat aaagtgtaaa gcctggggtg cctaatgagt gagctaactc acattaattg 180 cgttgcgctc actgcccgct ttccagtcgg gaaacctgtc gtgccagctg cattaatgaa 240 tcggccaacg cgcggggaga ggcggtttgc gtattgggcg ctcttccgct tcctcgctca 300 ctgactcgct gcgctcggtc gttcggctgc ggcgagcggt atcagctcac tcaaaggcgg 360 taatacggtt atccacagaa tcaggggata acgcaggaaa gaacatgtga gcaaaaggcc 420 agcaaaaggc caggaaccgt aaaaaggccg cgttgctggc gtttttccat aggctccgcc 480 cccctgacga gcatcacaaa aatcgacgct caagtcagag gtggcgaaac ccgacaggac 540 tataaagata ccaggcgttt ccccctggaa gctccctcgt gcgctctcct gttccgaccc 600 tgccgcttac cggatacctg tccgcctttc tcccttcggg aagcgtggcg ctttctcata 660 gctcacgctg taggtatctc agttcggtgt aggtcgttcg ctccaagctg ggctgtgtgc 720 acgaaccccc cgttcagccc gaccgctgcg ccttatccgg taactatcgt cttgagtcca 780 acccggtaag acacgactta tcgccactgg cagcagccac tggtaacagg attagcagag 840 cgaggtatgt aggcggtgct acagagttct tgaagtggtg gcctaactac ggctacacta 900 gaagaacagt atttggtatc tgcgctctgc tgaagccagt taccttcgga aaaagagttg 960 gtagctcttg atccggcaaa caaaccaccg ctggtagcgg tggttttttt gtttgcaagc 1020 agcagattac gcgcagaaaa aaaggatctc aagaagatcc tttgatcttt tctacggggt 1080 ctgacgctca gtggaacgaa aactcacgtt aagggatttt ggtcatgaga ttatcgtcga 1140 caattcgcgc gcgaaggcga agcggcatgc atttacgttg acaccatcga atggtgcaaa 1200 acctttcgcg gtatggcatg atagcgcccg gaagagagtc aattcagggt ggtgaatgtg 1260 aaaccagtaa cgttatacga tgtcgcagag tatgccggtg tctcttatca gaccgtttcc 1320 cgcgtggtga accaggccag ccacgtttct gcgaaaacgc gggaaaaagt ggaagcggcg 1380 atggcggagc tgaattacat tcccaaccgc gtggcacaac aactggcggg caaacagtcg 1440 ttgctgattg gcgttgccac ctccagtctg gccctgcacg cgccgtcgca aattgtcgcg 1500 gcgattaaat ctcgcgccga tcaactgggt gccagcgtgg tggtgtcgat ggtagaacga 1560 agcggcgtcg aagcctgtaa agcggcggtg cacaatcttc tcgcgcaacg cgtcagtggg 1620 ctgatcatta actatccgct ggatgaccag gatgccattg ctgtggaagc tgcctgcact 1680 aatgttccgg cgttatttct tgatgtctct gaccagacac ccatcaacag tattattttc 1740 tcccatgaag acggtacgcg actgggcgtg gagcatctgg tcgcattggg tcaccagcaa 1800 atcgcgctgt tagcgggccc attaagttct gtctcggcgc gtctgcgtct ggctggctgg 1860 cataaatatc tcactcgcaa tcaaattcag ccgatagcgg aacgggaagg cgactggagt 1920 gccatgtccg gttttcaaca aaccatgcaa atgctgaatg agggcatcgt tcccactgcg 1980 atgctggttg ccaacgatca gatggcgctg ggcgcaatgc gcgccattac cgagtccggg 2040 ctgcgcgttg gtgcggatat ctcggtagtg ggatacgacg ataccgaaga cagctcatgt 2100 tatatcccgc cgttaaccac catcaaacag gattttcgcc tgctggggca aaccagcgtg 2160 gaccgcttgc tgcaactctc tcagggccag gcggtgaagg gcaatcagct gttgcccgtc 2220 tcactggtga aaagaaaaac caccctggcg cccaatacgc aaaccgcctc tccccgcgcg 2280 ttggccgatt cattaatgca gctggcacga caggtttccc gactggaaag cgggcagtga 2340 gcgcaacgca attaatgtaa gttagcgcga attgtcgacc aaagcggcca tcgtgcctcc 2400 ccactcctgc agttcggggg catggatgcg cggatagccg ctgctggttt cctggatgcc 2460 gacggatttg cactgccggt agaactccgc gaggtcgtcc agcctcaggc agcagctgaa 2520 ccaactcgcg aggggatcga gcccggggtg ggcgaagaac tccagcatga gatccccgcg 2580 ctggaggatc atccagccgg cgtcccggaa aacgattccg aagcccaacc tttcatagaa 2640 ggcggcggtg gaatcgaaat ctcgtgatgg caggttgggc gtcgcttggt cggtcatttc 2700 gaaccccaga gtcccgctca gaagaactcg tcaagaaggc gatagaaggc gatgcgctgc 2760 gaatcgggag cggcgatacc gtaaagcacg aggaagcggt cagcccattc gccgccaagc 2820 tcttcagcaa tatcacgggt agccaacgct atgtcctgat agcggtccgc cacacccagc 2880 cggccacagt cgatgaatcc agaaaagcgg ccattttcca ccatgatatt cggcaagcag 2940 gcatcgccat gggtcacgac gagatcctcg ccgtcgggca tgcgcgcctt gagcctggcg 3000 aacagttcgg ctggcgcgag cccctgatgc tcttcgtcca gatcatcctg atcgacaaga 3060 ccggcttcca tccgagtacg tgctcgctcg atgcgatgtt tcgcttggtg gtcgaatggg 3120 caggtagccg gatcaagcgt atgcagccgc cgcattgcat cagccatgat ggatactttc 3180 tcggcaggag caaggtgaga tgacaggaga tcctgccccg gcacttcgcc caatagcagc 3240 cagtcccttc ccgcttcagt gacaacgtcg agcacagctg cgcaaggaac gcccgtcgtg 3300 gccagccacg atagccgcgc tgcctcgtcc tgcagttcat tcagggcacc ggacaggtcg 3360 gtcttgacaa aaagaaccgg gcgcccctgc gctgacagcc ggaacacggc ggcatcagag 3420 cagccgattg tctgttgtgc ccagtcatag ccgaatagcc tctccaccca agcggccgga 3480 gaacctgcgt gcaatccatc ttgttcaatc atgcgaaacg atcctcatcc tgtctcttga 3540 tcagatcttg atcccctgcg ccatcagatc cttggcggca agaaagccat ccagtttact 3600 ttgcagggct tcccaacctt accagagggc gccccagctg gcaattccgg ttcgcttgct 3660 gtccataaaa ccgcccagtc tagctatcgc catgtaagcc cactgcaagc tacctgcttt 3720 ctctttgcgc ttgcgttttc ccttgtccag atagcccagt agctgacatt catccggggt 3780 cagcaccgtt tctgcggact ggctttctac gtgttccgct tcctttagca gcccttgcgc 3840 cctgagtgct tgcggcagcg tgaagcttaa aaaactgcaa aaaatagttt gacttgtgag 3900 cggataacaa ttaagatgta cccaattgtg agcggataac aatttcacac attaaagagg 3960 agaaattaca tatg 3974 <210> 148 <211> 112 <212> DNA
<213> Homo sapiens <400> 198 aagcttaaaa aactgcaaaa aatagtttga cttgtgagcg gataacaatt aagatgtacc 60 caattgtgag cggataacaa tttcacacat taaagaggag aaattacata tg 112

Claims (68)

What Is Claimed Is:
1. A method of increasing the level of platelets in blood comprising administering to an individual a polypeptide selected from the group consisting of:

(a) a polypeptide comprising an amino acid sequence at least 95%
identical to Arg (80) - Ser (208) of SEQ ID NO:2; and (b) a polypeptide comprising an amino acid sequence identical to Arg (80) - Ser (208) of SEQ ID NO:2 except for at least one but not more than twenty amino acid substitutions;

wherein said polypeptide is administered in an amount effective to increase the platelet level in said individual's blood.
2. The method of claim 1, wherein said polypeptide is administered to alleviate thrombocytopenia.
3. The method of claim 2, wherein said thrombocytopenia is caused by disease or condition selected from the group consisting of: drug induced hypersensitivity, idiopathic thrombocytopenia pupura (ITP), posttransfusion purpura, neonatal thrombocytopenia, metastatic tumors in the bone, aplastic anemia, myelofibrosis, leukemia, microangiopathic hemolytic anemia, thrombotic thrombocytopenic purpura (TTP), hemolytic-uremic syndrome, prosthetic valve hemolytic syndrome, cancer chemotherapy, Zieve's syndrome, sepsis, HELLP
preeclamptic syndrome, megaloblastic anemia due to B21 and folic acid deficiency, peritonitis, congenital rubella syndrome, HIV-1 virus infection, Epstein-Barr infectious mononucleosis, systemic lupus, preeclampsia, thyrotoxicosis, uremia, and disseminated intravascular coagulation (DIC).
4. A method of increasing the level of plasma fibrinogen comprising administering to an individual a polypeptide selected from the group consisting of:

(a) a polypeptide comprising an amino acid sequence at least 95%
identical to Arg (80) - Ser (208) of SEQ ID NO:2; and (b) a polypeptide comprising an amino acid sequence identical to Arg (80) - Ser (208) of SEQ ID NO:2 except for at least one but not more than twenty amino acid substitutions;

wherein said polypeptide is administered in an amount effective to increase the level of plasma fibrinogen.
5. The method of claim 4, wherein said polypeptide is administered to alleviate hypofibrinogenemia.
6. The method of claim 5, wherein said hypofibrinogenemia is caused by a disease or condition selected from the group consisting of: hepatitis, cirrhosis, and disseminated intravascular coagulation (DIC)
7. A method of increasing the level of serum albumin comprising administering to an individual a polypeptide selected from the group consisting of:

(a) a polypeptide comprising an amino acid sequence at least 95%
identical to Arg (80) - Ser (208) of SEQ ID O0:2; and (b) a polypeptide comprising an amino acid sequence identical to Arg (80) - Ser (208) of SEQ ID NO:2 except for at least one but not more than twenty amino acid substitutions;

wherein said polypeptide is administered in an amount effective to increase the level of serum albumin.
8. The method of claim 7, wherein said polypeptide is administered to alleviate hypoalbuminemia.
9. The method of claim 8, wherein said hypoalbuminemia is caused by a disease or condition selected from the group consisting of: hemorrhages, burns, exudates, rheumatic diseases, granulomatous processes, bacterial infections, viral infections accompanied by tissue destruction, tissue necrosis, vasculitis, ulcerative bowel disease, serositis, subacute bacterial endocarditis, parasitic infestations, acute and chronic liver disease, amyloidosis, malnutrition, malignancy, congestive heart failure, constrictive pericarditis, cardiac valvular disease, nephrotic syndrome, trauma and crush injuries, gastrointestinal and lymphatic fistulae, and protein-losing gastroenteropathies.
10. A method of increasing the level of serum globulin comprising administering to an individual a polypeptide selected from the group consisting of:

(a) a polypeptide comprising an amino acid sequence at least 95%
identical to Arg (80) - Ser (208) of SEQ ID NO:2; and (b) a polypeptide comprising an amino acid sequence identical to Arg (80) - Ser (208) of SEQ ID NO:2 except for at least one but not more than twenty amino acid substitutions;

wherein said polypeptide is administered in an amount effective to increase the level of serum globulin.
11. The method of claim 10, wherein said polypeptide is administered to alleviate hypoglobulinemia.
12. The method of claim 11, wherein said hypoglobulinemia is caused by a disease or condition selected from the group consisting of: alpha-1 antitrypsin deficiencies, severe liver disease, estrogen therapy, megaloblastic anemia, hypogammaglobulinemia and aggammaglobuinemia.
13. A method of increasing the level of total serum protein comprising administering to an individual a polypeptide selected from the group consisting of:

(a) a polypeptide comprising an amino acid sequence at least 95%
identical to Arg (80) - Ser (208) of SEQ ID NO:2; and (b) a polypeptide comprising an amino acid sequence identical to Arg (80) - Ser (208) of SEQ ID NO:2 except for at least one but not more than twenty amino acid substitutions;

wherein said polypeptide is administered in an amount effective to increase the level of total serum protein.
14. The method of claim 13, wherein said polypeptide is administered to alleviate total protein loss.
15. The method of claim 14, wherein said total serum protein loss is caused by a disease or condition selected from the group consisting of:
protein-losing gastroenteropathies, acute burns, nephrotic syndrome, chronic liver disease, malabsorption syndrome, malnutrition, and agammaglobulinemia.
16. A method of reducing damage caused by an inhibition of normal proliferation of epithelial cells of the bladder or prostate comprising administering to an individual a polypeptide selected from the group consisting of:

(a) a polypeptide comprising an amino acid sequence at least 95%
identical to Arg (80) - Ser (208) of SEQ ID NO:2; and (b) a polypeptide comprising an amino acid sequence identical to Arg (80) - Ser (208) of SEQ ID NO:2 except for at least one but not more than twenty amino acid substitutions;

wherein said polypeptide is administered in an amount effective to reduce damage caused by an inhibition of normal proliferation of epithelial cells of the bladder or prostate.
17. The method of claim 16, wherein normal proliferation of epithelial cells of the bladder or prostate is inhibited by radiation treatment or by administration of an antineoplastic drug.
18. The method of claim 17, wherein said antineoplastic drug is cyclophosphamide.
19. The method of claim 16, wherein said polypeptide is administered to prevent or reduce damage caused by hemorrhagic cystitis.
20. The method of claim 17, wherein said polypeptide is administered prior to said radiation treatment or administration of an antineoplastic drug.
21. The method of claim 17, wherein said polypeptide is administered to increase bladder load.
22. The method of claim 17, further comprising administering an effective amount of mesna.
23. A method of stimulating proliferation of salivary gland cells comprising administering to an individual a polypeptide selected from the group consisting of:

(a) a polypeptide comprising an amino acid sequence at least 95%
identical to Arg (80) - Ser (208) of SEQ ID NO:2; and (b) a polypeptide comprising an amino acid sequence identical to Arg (80) - Ser (208) of SEQ ID NO:2 except for at least one but not more than twenty amino acid substitutions;

wherein said polypeptide is administered in an amount effective to stimulate proliferation of salivary gland cells.
24. The method of claim 23, wherein said polypeptide is administered in an amount effective to stimulate proliferation of the submandibular salivary gland cells.
25. The method of claim 23, wherein said polypeptide is administered in an amount effective to stimulate proliferation of the parotid salivary gland cells.
26. The method of claim 23, wherein said polypeptide is administered in an amount effective to stimulate proliferation of the sublingual salivary gland cells.
27. The method of claim 24, wherein said submandibular salivary gland cells are serous secretory cells.
28. The method of claim 24, wherein said submandibular salivary gland cells are mucous secretory cells.
29. The method of claim 24, wherein said parotid salivary gland cells are serous secretory acini cells.
30. The method of claim 23, wherein said polypeptide is administered to treat or reduce salivary gland dysfunction.
31. A method of stimulating proliferation of lacrimal gland cells comprising administering to an individual a polypeptide selected from the group consisting of:

(a) a polypeptide comprising an amino acid sequence at least 95%
identical to Arg (80) - Ser (208) of SEQ ID NO:2; and (b) a polypeptide comprising an amino acid sequence identical to Arg (80) - Ser (208) of SEQ ID NO:2 except for at least one but not more than twenty amino acid substitutions;

wherein said polypeptide is administered in an amount effective to stimulate proliferation of lacrimal gland cells.
32. The method of claim 31, wherein said lacrimal gland cells are secretory cells.
33. The method of claim 31, wherein said polypeptide is administered in an amount effective to treat or reduce lacrimal gland dysfunction.
34. The method of claim 33, wherein said polypeptide is administered to treat or reduce lacrimal gland dysfunction caused by radiation therapy, autoimmune disease, aging, HIV, keratoconjuctivitis sicca, burns or other medical conditions.
35. A method of stimulating proliferation of the sinus epithelium comprising administering to an individual a polypeptide selected from the group consisting of:

(a) a polypeptide comprising an amino acid sequence at least 95%
identical to Arg (80) - Ser (208) of SEQ ID NO:2; and (b) a polypeptide comprising an amino acid sequence identical to Arg (80) - Ser (208) of SEQ ID NO:2 except for at least one but not more than twenty amino acid substitutions;

wherein said polypeptide is administered in an amount effective to stimulate the sinus epithelium.
36. The method of claim 35, wherein said polypeptide is administered in an amount effective to treat or reduce damage to the sinus epithelium.
37. The method of claim 36, wherein said polypeptide is administered to treat or reduce damage to the sinus epithelium caused by surgery, wounds, or other medical conditions.
38. The method of claim 36, wherein said polypeptide is administered to reduce infection, scarring, polyps, cyst formation and recurrence.
39. A method of stimulating proliferation of Goblet cells comprising administering to an individual a polypeptide selected from the group consisting of:

(a) a polypeptide comprising an amino acid sequence at least 95%
identical to Arg (80) - Ser (208) of SEQ ID NO:2; and (b) a polypeptide comprising an amino acid sequence identical to Arg (80) - Ser (208) of SEQ ID NO:2 except for at least one but not more than twenty amino acid substitutions;
wherein said polypeptide is administered in an amount effective to stimulate proliferation of Goblet cells.
40. The method of claim 39, wherein said Goblet cells are in the respiratory epithelium of the nasal air passage way.
41. The method of claim 39, wherein said Goblet cells are in the large or small intestine.
42. The method of claim 39, wherein said Goblet cells are in the conjunctiva.
43. The method of claim 39, wherein said polypeptide is administered for treating or preventing keratoconjuctivitis sicca.
44. The method of claim 1, wherein said polypeptide is administered with a pharmaceutically acceptable carrier or excipient.
45. The method of claim 1, wherein said polypeptide is administered by a route selected from the group consisting of intravenously, subcutaneously and intraperitoneally.
46. The method of claim 1, wherein said polypeptide is (a).
47. The method of claim 46, wherein said polypeptide comprises Arg (80) - Ser (208) of SEQ ID NO:2.
48. The method of claim 46, wherein said polypeptide comprises an amino acid sequence at least 95% identical to Val(77) to Ser(208) of SEQ ID
NO:2.
49. The method of claim 48, wherein said polypeptide comprises Val(77) to Ser(208) of SEQ ID NO:2.
50. The method of claim 46, wherein said polypeptide comprises an amino acid sequence at least 95% identical to Ser(69) to Ser(208) of SEQ ID
NO:2.
51. The method of claim 50, wherein said polypeptide comprises Ser(69) to Ser(208) of SEQ ID NO:2.
52. The method of claim 46, wherein said polypeptide comprises an amino acid sequence at least 95% identical to Ala (63) - Ser (208) of SEQ ID
NO:2.
53. The method of claim 52, wherein said polypeptide comprises Ala (63) - Ser (208) of SEQ ID NO:2.
54. The method of claim 46, wherein said polypeptide comprises an amino acid sequence at least 95% identical to Cys (37) - Ser (208) of SEQ ID
NO:2.
55. The method of claim 54, wherein said polypeptide comprises Cys (37) - Ser (208) of SEQ ID NO:2.
56. The method of claim 46, wherein said polypeptide comprises an amino acid sequence at least 95% identical to Thr (36) - Ser (208) of SEQ ID
NO:2.
57. The method of claim 56, wherein said polypeptide comprises Thr (36) - Ser (208) of SEQ ID NO:2.
58. The method of claim 46, wherein said polypeptide comprises an amino acid sequence at least 95% identical to Trp (2) - Ser (208) of SEQ ID
NO:2.
59. The method of claim 58, wherein said polypeptide comprises Trp (2) - Ser (208) of SEQ ID NO:2.
60. The method of claim 46, wherein said polypeptide comprises an amino acid sequence at least 95% identical to Met (1) - Ser (208) of SEQ ID
NO:2.
61. The method of claim 60, wherein said polypeptide comprises Met (1) - Ser (208) of SEQ ID NO:2.
62. The method of claim 47, 49, 51, 53, 55, or 57, having a Met residue at the N-terminus.
63. The method of claim 51, wherein said polypeptide consists essentially of Ser (69) - Ser (208) of SEQ ID NO:2.
64. The method of claim 63, having a Met residue at the N-terminus.
65. The method of claim 51, wherein said polypeptide consists of Ser (69) - Ser (208) of SEQ ID NO:2.
66. The method of claim 51, wherein said polypeptide consists of Met (1) - Ser (141) of SEQ ID NO:96.
67. The method of claim 1, wherein said polypeptide is (b).
68. The method of claim 67, wherein said substitution is selected from the group consisting of Arg (80) Lys, Lys (87) Arg, Tyr (88) Trp, Phe (89) Tyr, Lys (91) Arg, Ser (99) Lys, Lys (102) Gln, Lys 103(Glu), Glu (104) Met, Asn (105) Lys, Pro (107) Asn, Ser (109) Asn, Leu (111) Met, Thr (114) Arg, Glu(117) Ala, Val (120) Ile, Val (123) Ile, Ala (125) Gly, Ile (126) Val, Asn (127) Glu, Asn (127) Gln, Tyr (130) Phe, Met (134) Thr, Lys (136) Glu, Lys (137) Glu, Gly (142) Ala, Ser (143) Lys, Phe (146) Ser, Asn (148) Glu, Lys (151) Asn, Leu (152) Phe, Glu (154) Gly, Glu (154) Asp, Arg (155) Leu, Glu (157) Leu, Gly (160) His, Phe (167) Ala, Asn (168) Lys, Gln (170) Thr, Arg (174) Gly, Tyr (177) Phe, Gly (182) Gln, Ala (185) Val, Ala (185) Leu, Ala (185) Ile, Arg (187) Gln (190) Lys, Lys (195) Glu, Thr (197) Lys, Ser (198) Thr, Arg (194) Glu, Arg (194) Gln, Lys (191) Glu, Lys (191) Gln, Arg (188) Glu, Arg (188) Gln, and Lys (183) Glu.
CA002320515A 1998-02-13 1999-02-12 Therapeutic uses of keratinocyte growth factor-2 Abandoned CA2320515A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US7458598P 1998-02-13 1998-02-13
US60/074,585 1998-02-13
US11438798P 1998-12-30 1998-12-30
US60/114,387 1998-12-30
PCT/US1999/003018 WO1999041282A1 (en) 1998-02-13 1999-02-12 Therapeutic uses of keratinocyte growth factor-2

Publications (1)

Publication Number Publication Date
CA2320515A1 true CA2320515A1 (en) 1999-08-19

Family

ID=26755823

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002320515A Abandoned CA2320515A1 (en) 1998-02-13 1999-02-12 Therapeutic uses of keratinocyte growth factor-2

Country Status (9)

Country Link
US (1) US6599879B1 (en)
EP (1) EP1054900A4 (en)
JP (1) JP2002507546A (en)
KR (1) KR20010040906A (en)
CN (1) CN1295579A (en)
AU (1) AU762519B2 (en)
CA (1) CA2320515A1 (en)
NZ (1) NZ506422A (en)
WO (1) WO1999041282A1 (en)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6693077B1 (en) 1995-02-14 2004-02-17 Human Genome Sciences, Inc. Keratinocyte growth factor-2
US6077692A (en) 1995-02-14 2000-06-20 Human Genome Sciences, Inc. Keratinocyte growth factor-2
US7232667B2 (en) * 1995-02-14 2007-06-19 Human Genome Sciences, Inc. Keratinocyte growth factor-2 polynucleotides
US6869927B1 (en) 1997-12-22 2005-03-22 Human Genome Sciences, Inc. Keratinocyte growth factor-2 formulations
EP1041996A4 (en) 1997-12-22 2003-05-14 Human Genome Sciences Inc Keratinocyte growth factor-2 formulations
CA2320515A1 (en) 1998-02-13 1999-08-19 Human Genome Sciences, Inc. Therapeutic uses of keratinocyte growth factor-2
AU2002309473A1 (en) * 2001-01-08 2002-10-08 Human Genome Sciences, Inc. Keratinocyte growth factor-2
EP1421974B1 (en) * 2002-06-10 2006-12-06 D.M.G. Italia Srl Carboxy methyl beta-1, 3 glucan containing ophthalmic solution
CN1302109C (en) * 2004-04-21 2007-02-28 中国人民解放军军事医学科学院基础医学研究所 Horny cell growth factor mutant and its preparation method and use thereof
US7223737B1 (en) 2004-08-13 2007-05-29 Alcon, Inc. Method of treating dry eye disorders using glycosides
WO2006057859A1 (en) * 2004-11-24 2006-06-01 Therakine Corporation An implant for intraocular drug delivery
JP5637551B2 (en) * 2005-12-28 2014-12-10 キッセイ薬品工業株式会社 Pharmaceuticals for the treatment of salivary gland disorders
CN100434117C (en) * 2006-06-06 2008-11-19 武汉大学 Suppository for treating chronic cervicitis
CN101489569A (en) * 2006-06-12 2009-07-22 席拉坎有限责任公司 Topical treatment for diseases of eye surface
US20100068259A1 (en) * 2008-05-30 2010-03-18 Annette Tobia Natural product inhibitors of 3dg
CN107893081B (en) * 2017-12-27 2021-01-12 温州大学 Gene sequence, expression vector and production method of human keratinocyte growth factor-2
US20230285536A1 (en) * 2020-06-24 2023-09-14 Trustees Of Tufts College Edible cell based vaccines

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HU218090B (en) * 1989-01-31 2000-05-28 Stuart A. Aaronson Process for producing epidermis-specific growth factor, the dns coding this, the antibodies produced against them, and pharmaceutical compositions containing the growth factor or the antibody
US5703047A (en) * 1992-09-21 1997-12-30 Board Of Regents, The University Of Texas System Methods and treatments for corneal healing with growth factors
KR100390340B1 (en) 1993-03-26 2003-09-26 암겐 인코포레이티드 Pharmaceutical composition containing assets of keratinocyte growth
US5965530A (en) * 1993-03-26 1999-10-12 Amgen Inc. Therapeutic uses of keratinocyte growth factor
ES2227527T3 (en) * 1993-06-29 2005-04-01 Chiron Corp A TRACTED KERATINOCITS (KGF) GROWTH FACTOR THAT HAS INCREASED BIOLOGICAL ACTIVITY.
CA2185671A1 (en) 1994-03-15 1995-09-21 Prizm Pharmaceuticals, Inc. Heparin-binding growth factors for gene therapy and anterior eye disorders
EP1323824A3 (en) * 1995-02-14 2004-01-21 Human Genome Sciences, Inc. Use of human keratinocyte growth factor-2 for the preparation of pharmaceutical compositions
US6077692A (en) 1995-02-14 2000-06-20 Human Genome Sciences, Inc. Keratinocyte growth factor-2
US5773252A (en) 1995-06-05 1998-06-30 Human Genome Sciences, Inc. Fibroblast growth factor 15
WO1998006844A1 (en) * 1996-08-13 1998-02-19 Human Genome Sciences, Inc. Keratinocyte growth factor-2 (kgf-2 or fibroblast growth factor-12, kgf-12)
EP1041996A4 (en) 1997-12-22 2003-05-14 Human Genome Sciences Inc Keratinocyte growth factor-2 formulations
CA2320515A1 (en) 1998-02-13 1999-08-19 Human Genome Sciences, Inc. Therapeutic uses of keratinocyte growth factor-2
EP1196187A4 (en) 1999-06-02 2003-02-05 Human Genome Sciences Inc Keratinocyte growth factor-2 formulations
EP1196441A1 (en) 1999-07-02 2002-04-17 Human Genome Sciences, Inc. Keratinocyte growth factor-2

Also Published As

Publication number Publication date
AU762519B2 (en) 2003-06-26
EP1054900A1 (en) 2000-11-29
CN1295579A (en) 2001-05-16
AU2867799A (en) 1999-08-30
US6599879B1 (en) 2003-07-29
KR20010040906A (en) 2001-05-15
EP1054900A4 (en) 2004-12-22
JP2002507546A (en) 2002-03-12
NZ506422A (en) 2002-10-25
WO1999041282A1 (en) 1999-08-19
WO1999041282A8 (en) 2000-07-06

Similar Documents

Publication Publication Date Title
US6077692A (en) Keratinocyte growth factor-2
US6599879B1 (en) Therapeutic uses of keratinocyte growth factor-2
AU739773B2 (en) Keratinocyte growth factor-2 (KGF-2 or fibroblast growth factor-12, FGF-12)
US20030008357A1 (en) Vascular endothelial growth factor 2
US6303338B1 (en) Pancreas-derived plasminogen activator inhibitor
US6143498A (en) Antimicrobial peptide
KR20010034576A (en) Vascular endothelial growth factor 2
US6419917B1 (en) Human chemotactic protein
MXPA02003434A (en) Vascular endothelial growth factor 2.
US6011012A (en) Human cystatin E
JPH07103156B2 (en) Osteogenic growth polypeptides identified from regenerated bone marrow
MXPA00007884A (en) Therapeutic uses of keratinocyte growth factor-2
US20020110867A1 (en) Cardiac and pancreatic protein and gene
JP2001511648A (en) Dendritic cell-derived growth factor
WO1998044112A9 (en) Human muscle derived growth factor - cardiac and pancreatic protein (capp) and gene
EP1247862A2 (en) Keratinocyte growth factor-2 (KGF-2 or fibroblast growth factor-12, FGF-12)
KR19980703534A (en) Human cryptin growth factor
WO2023175585A1 (en) Hybrid promoters and their use in osteolysis syndrome
AU1017202A (en) Keratinocyte growth factor-2 (kgf-2 or fibroblast growth fact or-12, fgf-12)
WO1999009161A1 (en) Pancreas-derived plasminogen activator inhibitor
MXPA00008920A (en) Vascular endothelial growth factor 2
NZ513514A (en) Antimicrobial peptide
MXPA97006854A (en) Factor 14 of fibroblas growth

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued