CA2307322A1 - Regulation of epithelial tissue by hedgehog-like polypeptides, and formulations and uses related thereto - Google Patents

Regulation of epithelial tissue by hedgehog-like polypeptides, and formulations and uses related thereto Download PDF

Info

Publication number
CA2307322A1
CA2307322A1 CA002307322A CA2307322A CA2307322A1 CA 2307322 A1 CA2307322 A1 CA 2307322A1 CA 002307322 A CA002307322 A CA 002307322A CA 2307322 A CA2307322 A CA 2307322A CA 2307322 A1 CA2307322 A1 CA 2307322A1
Authority
CA
Canada
Prior art keywords
hedgehog
leu
seq
ala
gly
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.)
Granted
Application number
CA002307322A
Other languages
French (fr)
Other versions
CA2307322C (en
Inventor
Elizabeth A. Wang
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.)
Curis Inc
Original Assignee
Ontogeny, Inc.
Elizabeth A. Wang
Curis, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=26849158&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2307322(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Ontogeny, Inc., Elizabeth A. Wang, Curis, Inc. filed Critical Ontogeny, Inc.
Publication of CA2307322A1 publication Critical patent/CA2307322A1/en
Application granted granted Critical
Publication of CA2307322C publication Critical patent/CA2307322C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0625Epidermal cells, skin cells; Cells of the oral mucosa
    • C12N5/0629Keratinocytes; Whole skin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/60Sugars; Derivatives thereof
    • A61K8/606Nucleosides; Nucleotides; Nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/64Proteins; Peptides; Derivatives or degradation products thereof
    • 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
    • A61P1/02Stomatological preparations, e.g. drugs for caries, aphtae, periodontitis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • A61Q19/08Anti-ageing preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q7/00Preparations for affecting hair growth
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q7/00Preparations for affecting hair growth
    • A61Q7/02Preparations for inhibiting or slowing hair growth
    • 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
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43563Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from insects
    • C07K14/43577Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from insects from flies
    • C07K14/43581Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from insects from flies from Drosophila
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/461Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from fish
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/465Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from birds
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0625Epidermal cells, skin cells; Cells of the oral mucosa
    • C12N5/0627Hair cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/40Regulators of development
    • C12N2501/41Hedgehog proteins; Cyclopamine (inhibitor)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/70Enzymes

Abstract

The present application relates to a method for modulating the growth state of an epithelial cell by ectopically contacting the epithelial cell, in vitro or in vivo, with a hedgehog therapeutic or ptc therapeutic in an amount effective to alter the rate (promote or inhibit) of proliferation of the epithelial cell, e.g., relative to the absence of administration of the hedgehog therapeutic or ptc therapeutic. The subject method can be used, for example, to modulate the growth state of an epithelial tissue, such as for inducing the formation of skin or other cutaneous tissue, or for inducing growth of hair.

Description

wo ~noZ9s rcr~s9arnzz~
Regulation of Epithelial Tissue by Hedgehog like Polypeptides, and Formulations and Uses Related Thereto Background of the Invention Pattern formation is the activity by which embryonic cells form ordered spatial arrangements of differentiated tissues. The physical complexity of higher organisms arises during embryogenesis through the interplay of cell-intrinsic lineage and cell-extrinsic signaling. Inductive interactions are essential to embryonic patterning in vertebrate development from the earliest establishment of the body plan, to the patterning of the organ systems, to the generation of diverse cell types during tissue differentiation (Davidson. E., (1990) Development 108: 365-389; Gwdon, J. B., (1992) Cell 68: 185-199;
3esse1l, T. M. et al., (1992) Cell 68: 257-270). The effects of developmental cell interactions are varied.
Typically, responding cells are diverted from one route of cell differentiation to another by inducing cells that differ from both the uninduced and induced states of the responding cells I S (inductions). Sometimes cells induce their neighbors to differentiate like themselves (homoiogenetic induction); in other cases a cell inhibits its neighbors from differentiating like itself. Cell interactions in early development may be sequential. such that an initial induction between two cell types leads to a progressive amplification of diversiy.
Moreover, inductive interactions occw not only in embryos, but in adult cells as well. and can act to establish and maintain morphogenetic patterns as well as induce differentiation (J.B. Gurdon (1992) Cell 68:185-199).
Members of the Hedgehog family of signaling molecules mediate many important short- and long-range patterning processes during invertebrate and vertebrate development. In the fly a single hedgehog gene regulates segmental and imaginal disc patterning. In contrast. in vertebrates a hedgehog gene family is involved in the control of left-right asymmetry, polarity in the CNS, somites and limb, organogenesis, chondrogenesis and spermatogenesis.
The first hedgehog gene was identified by a genetic screen in the fruitfly Drosophila melanogaster (Niisslein-Volhard, C. and Wieschaus, E. ( 1980) Nature 287, 795-801 ). This screen identified a number of mutations affecting embryonic and larval development. In 1992 and 1993, the molecular natwe of the Drosophila hedgehog (hh) gene was reported (C.F., Lee et al. (1992) Cell 71, 33-50), and since then, several hedgehog homologues have been isolated wo ~noZSa rcrtus9snzzz~
-2-from various vertebrate species. While only one hedgehog gene has been found in Drosophila.
and other invertebrates, multiple Hedgehog genes are present in vertebrates.
The various Hedgehog proteins consist of a signal peptide, a highly conserved N-terminal region, and a more divergent C-terminal domain. In addition to signal sequence cleavage in the secretory pathway (Lee, J.J. et al. (1992) Cell 71:33-50;
Tabata, T. et al.
(1992) Genes Dev. 2635-2645; Chang, D.E. et al. (1994) Development 120:3339-3353), Hedgehog precursor proteins undergo an internal autoproteolytic cleavage which depends on conserved sequences in the C-terminal portion (Lee et al. (1994) Science 266:1528-1537;
Porter et al. (1995) Nature 374:363-366). This autocleavage leads to a 19 kD N-terminal peptide and a C-terminal peptide of 26-28 kD (Lee et al. (1992) supra; Tabata et al. (1992) supra; Chang et al. (1994) supra; Lee et al. (1994) supra; Bumcrot, D.A., et al. (1995) Mol.
Cell. Biol. 15:2294-2303; Porter et al. (1995) supra; Ekker, S.C. et al.
(1995) Curr. Biol.
5:944-955; Lai, C.J. et al. (1995) Development 121:2349-2360). The N-terminal peptide stays tightly associated with the surface of cells in which it was synthesized, while the C-terminal peptide is freely diffusible both in vitro and in vivo (Lee et al. ( 1994) supra; Bumcrot et al.
{1995) supra; Mart', E. et al. (1995) Development 121:2537-2547; Roelink, H.
et al. (1995) Cell 81:445-455). Interestingly, cell surface retention of the N-terminal peptide is dependent on autocleavage, as a truncated form of HH encoded by an RNA which terminates precisely at the normal position of internal cleavage is diffusible in vitro (Porter et al.
(1995) supra) and in vivo (Porter, J.A. et al. (1996) Cell 86, 21-34). Biochemical studies have shown that the autoproteolytic cleavage of the HH precursor protein proceeds through an internal thioester intermediate which subsequently is cleaved in a nucleophilic substitution. It is likely that the nucleophile is a small lipophilic molecule which becomes covalently bound to the C-terminal end of the N-peptide (Porter et al. {1996) supra), tethering it to the cell surface. The biological implications are profound. As a result of the tethering, a high local concentration of N-terminal Hedgehog peptide is generated on the surface of the Hedgehog producing cells.
It is this N-terminal peptide which is both necessary and sufficient for short and long range Hedgehog signaling activities in Drosophila and vertebrates (Porter et al. (1995) supra; Ekker et al.
(1995) supra; Lai et al. (1995) supra; Roelink, H. et al. (1995) Cell 81:445-4.55; Porter et al.
(1996) supra; Fietz, M.J. et al. (1995) Curr. Biol. 5:643-651; Fan, C.-M. et al. (1995) Cell 81:457-465; Mart', E., et al. (1995) Nature 375:322-325; Lopez-Martinez et al.
(1995) Curr.
-3-Biol 5:791-795; Ekker, S.C. et al. (1995) Development 121:2337-2347; Forbes, A.J. et.
al.(1996) Development 122:1125-1135).
HH has been implicated in short- and longe range patterning processes at various sites during Drosophila development. In the establishment of segment polarity in early embryos, it has short range effects which appear to be directly mediated, while in the patterning of the imaginal discs, it induces long range effects via the induction of secondary signals.
In vertebrates, several hedgehog genes have been cloned in the past few years (see Table 1). Of these genes, Shh has received most of the experimental attention, as it is expressed in different organizing centers which are the sources of signals that pattern neighbouring tissues. Recent evidence indicates that Shh is involved in these interactions.
The interaction of a hedgehog protein with one of its cognate receptor, patched, sets in motion a cascade involving the activation and inhibition of downstream effectors, the ultimate consequence of which is, in some instances, a detectable change in the transcription or translation of a gene. Transcriptional targets of hedgehog signaling are the patched gene itself (Hidalgo and Ingham, 1990 Development 110, 291-301; Marigo et al., 1996 ) and the vertebrate homologs of the drosophila cubitus interruptus (Ci) gene, the GLI
genes (Hui et al.
(1994) Dev Biol 162:402-413). Patched gene expression has been shown to be induced in cells of the limb bud and the neural plate that are responsive to Shh. (Marigo et al. ( 1996) Development 122:1225-1233). The GLI genes encode putative transcription factors having zinc finger DNA binding domains (Orenic et al. (1990) Genes & Dev 4:1053-1067;
Kinzler et al. (1990) Mol Cell Biol 10:634-642). Transcription of the GLI gene has been reported to be upregulated in response to hedgehog in limb buds, while transcription of the GLI3 gene is downregulated in response to hedgehog induction (Marigo et al. ( 1996}
Development 122:1225-1233). Moreover, it has been demonstrated that elevated levels of Ci are sufficient to activate patched (ptc) and other hedgehog target genes, even in the absence of hedgehog activity.
Summary ojthe Invention One aspect of the present application relates to a method for modulating the growth state of an epithelial cell by ectopically contacting the epithelial cell, in vitro or in vivo, with a hedgehog therapeutic or ptc therapeutic in an amount effective to alter the rate (promote or
-4-inhibit) of proliferation of the epithelial cell, e.g., relative to the absence of administeration of.
the hedgehog therapeutic or ptc therapeutic. The subject method can be used, for example, to modulate the growth state of an epithelial tissue, such as for inducing the formation of skin or other cutaneous tissue, or for inducing growth of hair.
Wherein the subject method is carried out using a hedgehog therapeutic, the hedgehog therapeutic preferably a polypeptide including a hedgehog portion comprising at least a bioactive extracellular portion of a hedgehog protein, e.g., the hedgehog portion includes at least 50, 100 or 150 (contiguous) amino acid residues of an N-terminal half of a hedgehog protein.In preferred embodiments, the hedgehog portion includes at least a portion of the hedgehog protein corresponding to a l9kd fragment of the extracellular domain of a hedgehog protein.
In certain preferred embodiments, the hedgehog portion has an amino acid sequence at least 60, 75, 85, or 95 percent identical with a hedgehog protein of any of SEQ ID Nos. 10-18 or 20, though sequences identical to those sequence listing entries are also contemplated as useful in the present method. The hedgehog portion can be encoded by a nucleic acid which hybridizes under stringent conditions to a nucleic acid sequence of any of SEQ
ID Nos. 1-9 or 19, e.g., the hedgehog portion can be encoded by a vertebrate hedgehog gene, especially a human hedgehog gene.
In certain embodiments, the hedgehog polypeptide is modified with one or more sterol moieties, e.g., cholesterol or a derivative thereof.
In certain embodiments, the hedgehog polypeptide is modified with one or more fatty acid moieties, such as a fatty acid moiety selected from the group consisting of myristoyl, pahnitoyl, stearoyl, and arachidoyl.
In certain embodiments, the hedgehog polypeptide is modified with one or more aromatic hydrocarbons, such as benzene, perylene, phenanthrene, anthracene, naphthalene, pyrene, chrysene, or naphthacene.
In certain embodiments, the hedgehog polypeptide is modified one or more times with a C7 - C30 alkyl or cycloalkyl.
-5-In other embodiments, the subject method can be carried out by administering a gene.
activation construct, wherein the gene activation construct is deigned to recombine with a genomic hedgehog gene of the patient to provide a heterologous transcriptional regulatory sequence operatively linked to a coding sequence of the hedgehog gene.
In still other embodiments, the subject method can be practiced with the administration of a gene therapy construct encoding a hedgehog polypeptide. For instance, the gene therapy construct can be provided in a composition selected from a group consisting of a recombinant viral particle, a liposome, and a poly-cationic nucleic acid binding agent, In yet other embodiments, the subject method can be carried out using a ptc therapeutic. An exemplary ptc therapeutic is a small organic molecule which binds to a patched protein and derepresses patched mediated inhibition of mitosis, e.g., a molecule which binds to patched and mimics hedgehog-mediated patched signal transduction, which binds to patched and regulates patched dependent gene expression. For instance, the binding of the ptc therapeutic to patched may result in upregulation of patched and/or gli expression.
In a more generic sense, the ptc therapeutic can be a small organic molecule which interacts with epithelial cells to induce hedgehog-mediated patched signal transduction, such as by altering the localization, protein-protein binding and/or enzymatic activity of an intracellular protein involved in a patched signal pathway. For instance, the ptc therapeutic may alter the level of expression of a hedgehog protein, a patched protein or a protein involved in the intracellular signal transduction pathway of patched.
In certain embodiments, the ptc therapeutic is an antisense construct which inhibits the expression of a protein which is involved in the signal transduction pathway of patched and the expression of which antagonizes hedgehog-mediated signals. The antisense construct is perferably an oligonucleotide of about 20-30 nucleotides in length and having a GC content of at least 50 percent.
In other embodiments, the ptc therapeutic is an inhibitor of protein kinase A
(PKA), such as a 5-isoquinolinesulfonamide. The PKA inhibitor can be a cyclic AMP
analog.
Exemplary PKA inhibitors include N-[2-((p-bromocinnamyl)amino)ethyl]-5-isoquinolinesulfonamide, 1-(5-isoquinoline-sulfonyl)-2-methylpiperazine, KT5720, 8-bromo-
-6-R2~N~R1 wherein, R1 and R2 each can independently represent hydrogen, and as valence and stability permit a lower alkyl, a lower alkenyl, a lower alkynyl, a carbonyl (such as a carboxyl, an ester, a fotmate, or a ketone), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an amino, an acylamino, an amido, a cyano, a nitro, an azido, a sulfate, a sulfonate, a sulfonamido, -(CH2)m R8, -(CH2)m OH, -(CH2)m-O-lower alkyl, -(CH2)"; O-lower alkenyl, (CH2)~-O-(CH2)m Rg, -(CH2)m-SH, -(CH2}m S-lower alkyl, -(CH2)m S-lower alkenyl, -(CHZ)ri S-(CH2)m Rg, or R1 and R2 taken together with N form a heterocycle (substituted or unsubstituted);
R3 is absent or represents one or more substitutions to the isoquinoline ring such as a lower alkyl, a lower alkenyl, a lower alkynyl, a carbonyl (such as a carboxyl, an ester, a formate, or a ketone), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an amino, an acylamino, an amido, a cyano, a nitro, an azido, a sulfate, a sulfonate, a sulfonamido, -(CH2)m Rg, -(CH2)m-OH, -(CH2)m O-lower alkyl, -(CH2)m O-lower alkenyh -(CH2)n-O-(CH2)m Rg, -(CH2)m-SH, -(CH2)m-S-lower alkyl, -(CH2)n; S-lower alkenyl, -(CH2}n S-(CH2)m R8;
Rg represents a substituted or unsubstituted aryl, aralkyl, cycloalkyl, cycloalkenyl, or heterocycle; and n and m are independently for each occurrence zero or an integer in the range of 1 to 6.
The subject method can be used to treat, e.g., a epithelial disorder, such as in the control of a wound healing process. For instance, the subect method can be used as part of such treatments as burn treatment; skin regeneration, skin grafting, pressure sore treatment, wo ~no~s rcT~rs9snzz2~
dermal ulcer treatment, post surgery scar reduction and treatment of ulcerative colitis. In the contml of hair growth, the subject method can used as part of a treatment of alopecia.
Yet another aspect of the present invention concerns preparations of a hedgehog or ptc therapeutic formulated for topical application to epithelial tissue, e.g., to skin. For example, such formulations may include a polypeptide comprising a hedgehog polypeptide sequence including a bioactive fragment of a hedgehog protein, which polypeptide is formulated for topical application to epithelial tissue.
Detailed Description of the Invention Figures lA, B and C illustrate the induction of hair growth on mice treated with various hedgehog formulations.
Detailed Description of the Invention Normal skin epidermis is a complex epithelial tissue containing keratinocytes that are proliferating, differentiating and desquamating, and is stratified such that morphological and functional changes in the keratinocytes occur in an orderly progression. The normal epidermis is maintained in a dynamic steady state as proliferation of keratinocytes continually compensates for the loss of cells which are shed from the surface of the skin.
Within the epidermis, proliferation takes place in the basal layer of keratinocytes that are attached to the underlying basement membrane, and cells undergo terminal differentiation as they migrate through the suprabasal layers, finally being shed from the tissue surface as dead, cornified squames. Three subpopulations of basal keratinocytes have been defined by cell kinetic analysis: stem cells, transit-amplifying cells, and committed cells. Stem cells retain a high capacity for self renewal throughout adult life and are ultimately responsible for epidermal maintenance and repair. The progeny of stem cells can either be stem cells themselves or cells known as transit-amplifying cells. Transit-amplifying cells divide a small number of times, but have a high probability of producing daughters that withdraw irreversibly from the cell cycle and are committed to differentiate terminally.

WO 99/Z029$ PCTNS98/Z2227 _g_ 1. Overview The present application is directed to the discovery that preparations of hedgehog polypeptides can be used to control the formation and/or maintenance of epithelial tissue. As described in the appended examples, hedgehog proteins are implicated in the proliferation of epithelial stem cells and may provide early signals that regulate the differentiation of the stem cells into epithelial tissues. In general, the method of the present invention comprises contacting an epithelial cell with an amount of a hedgehog therapeutic (defined infra which produces a non-toxic response by the cell of (i) induction of epithelial tissue formation or (ii) inhibition of epithelial tissue formation, depending on the whether the hedgehog therapeutic is a sufficient hedgehog agonist or hedgehog antagonist. The subject method can be carried out on epithelial cells which may be either dispersed in culture or a part of an intact tissue or organ. Moreover, the method can be performed on cells which are provided in culture (in vitro), or on cells in a whole animal (in vivo).
In one aspect, the present invention provides pharmaceutical preparations and methods for controlling the proliferation of epithelially-derived tissue utilizing, as an active ingredient, a hedgehog polypeptide or a mimetic thereof. The invention also relates to methods of controlling proliferation of epithelial-derived tissue by use of the pharmaceutical preparations of the invention.
The hedgehog formulations of the present invention may be used as part of regimens in the treatment of disorders of, or surgical or cosmetic repair of, such epithelial tissues as skin and skin organs; corneal, lens and other ocular tissue; mucosal membranes; and periodontal epithelium. The methods and compositions disclosed herein provide for the treatment or prevention of a variety of damaged epithelial and mucosal tissues. For instance, the subject method can be used to control wound healing processes, as for example may be desirable in connection with any surgery involving epithelial tissue, such as from dermatological or periodontal surgeries. Exemplary surgical repair for which hedgehog therapy is a candidate treatment include severe burn and skin regeneration, skin grafts, pressure sores, dermal ulcers, fissures, post surgery scar reduction, and ulcerative colitis.
In another aspect of the present invention, hedgehog preparations can be used to effect the growth of hair, as for example in the treatment of alopecia whereby hair growth is potentiated, or for example in cosemetic removal of hair (depilation) whereby hair growth is inhibited.
In certain embodiments, the subject compositions can be used to inhibit, rather than promote, growth of epithelial-derived tissue. For instance, certain of the compositions disclosed herein may be applied to the treatment or prevention of a variety hyperplastic or neoplastic conditions. The method can find application for the treatment or prophylaxis of, e.g., psoriasis; keratosis; acne; comedogenic lesions; folliculitis and pseudofolliculitis;
keratoacanthoma; callosities; Darier's disease; ichthyosis; lichen planus;
molluscous contagiosum; melasma; Fordyce disease; and keloids or hypertrophic scars.
Certain of the formulations of the present invention may also be used as part of treatment regimens in auto-immune diseases for affecting healing of proliferative manifestations of the disorder, as for example, part of a treatment for aphthous ulcers, pemphigus such as pemphigus vulgaris, pemphigus foliaceus, pemphigus vegetans or pemphigus erythematous, epidermolysis, lupus lesions or desquamative lesions.
The subject hedgehog treatments are effective on both human and animal subjects afflicted with these conditions. Animal subjects to which the invention is applicable extend to both domestic animals and livestock, raised either as pets or for commercial purposes.
Examples are dogs, cats, cattle, horses, sheep, hogs and goats.
Still another aspect of the present invention provides a method of stimulating the growth and regulating the differentiation of epithelial tissue in tissue culture.
Without wishing to be bound by any particular theory, the induction of stem cell proliferation by hedgehog proteins may be due at least in part to the ability of these proteins to antagonize (directly or indirectly) patched-mediated regulation of gene expression and other physiological effects mediated by that protein. The patched gene product, a cell surface protein, is understood to signal through a pathway which causes transcriptional repression of members of the Wnt and DppBMP families of morphogens, proteins which impart positional information. In development of the CNS and patterning of limbs in vertebrates, the introduction of hedgehog relieves (derepresses) this inhibition conferred by patched, allowing expression of particular gene programs.

Recently, it has been reported that mutations in the human version of patched, a gene first identified in a fruit fly developmental pathway, cause a hereditary skin cancer and may contribute to sporadic skin cancers. See, for example, Hahn et al. { 1996) Cell 86:841-851; and Johnson et al. (1996) Science 272:1668-1671. The demonstraction that nevoid basal-cell carcinoma (NBCC) results from mutations in the human patched gene provided an example of the roles patched plays in post-embryonic deveolpment. These observations have led the art to understand one activity of patched to be a tumor suppressor gene, which may act by inhibiting proliferative signals from hedgehog. Our observations set forth below reveal potential new roles for the hedgehoglpatched pathway in maintenance of epithelial cell proliferation and differentiation. Accordingly, the present invention contemplates the use of other agents which are capable of mimicking the effect of the hedgehog protein on patched signalling, e.g., as may be identified from the drug screening assays described below.
ll. Definitions For convience, certain terms employed in the specfication, examples, and appended claims are collected here.
The term "hedgehog therapeutic" refers to various farms of hedgehog polypeptides, as well as peptidomimetics, which can modulate the proliferation/differentiation state of epithelial cells by, as will be clear from the context of individual examples, mimicing or potentiating (agonizing) or inhibiting (antagonizing) the effects of a naturally-occurring hedgehog protein. A hedgehog therapeutic which mimics or potentiates the activity of a wild-type hedgehog protein is a "hedgehog agonist". Conversely, a hedgehog therapeutic which inhibits the activity of a wild-type hedgehog protein is a "hedgehog antagonist".
In particular, the term "hedgehog polypeptide" encompasses preparations of hedgehog proteins and peptidyl fragments thereof, both agonist and antagonist forms as the specific context will make clear.
As used herein the term "bioactive fragment of a hedgehog protein" refers to a fragment of a full-length hedgehog polypeptide, wherein the fragment specifically agonizes or antagonizes inductive events mediated by wild-type hedgehog proteins. The hedgehog biactive fragment preferably is a soluble extracellular portion of a hedgehog protein, where solubility is with reference to physiologically compatible solutions.
Exemplary bioactive fragments are described in PCT publications WO 95/18856 and WO 96/17924.
The term "patched" or "ptc" refers to a family of related transmembrane proteins which have been implicated in the signal transduction induced by contacting a cell with a hedgehog protein. For example, the mammalian ptc family includes ptcl and ptc2. In addition to references set out below, see also Takabatake et al. ( 1997) FEBS Lett 410:485 and GenBank AB000847 for examples of ptc2. Unless otherwise evident from the context, it will be understood that embodiments described in the context of ptc 1 (or just ptc) also refer to equivalent embodiments involving other ptc homologs like ptc2.
The term "ptc therapeutic" refers to agents which either (i) mimic the effect of hedgehog proteins on patched signalling, e.g., which antagonize the cell-cycle inhibitory activity of patched, or (ii) activate or potentiate patched signalling. In other embodiments, the ptc therapeutic can be a hedgehog antagonist. The ptc therapeutic can be, e.g., a peptide. a nucleic acid, a carbohydrate, a small organic molecule, or natural product extract (or fraction thereof j.
A "proliferative" form of a hedgehog or ptc therapeutic is one which induces proliferation of epithelial cells, particularly epithelial stem cells.
Conversely, an "antiproliferative" form of a hedgehog or ptc therapeutic is one which inhibits proliferation of an epithelial cells, preferably in a non-toxic manner, e.g., by promoting or maintaining a differentiated phenotype or otherwise promoting quiescence.
By way of example, though not wishing to be bound by a particular theory, proliferative hedgehog polypeptide will generally be a form of the protein which derepresses patched-mediated cell-cycle arrest, e.g., the polypeptide mimics the effect of a naturally occurring hedgehog protein effect on epithelial cells. A proliferative ptc therapeutic includes other agents which depress patched-mediated cell-cycle arrest, and may act extracellularly or intracellularly.
An illustrative antiproliferative ptc therapeutic agent may potentiate patched-mediated cell-cycle arrest. Such agents can be small molecules that inhibit. e.g., hedgehog binding to patched, as well as agents which stimulate and/or potentiate a signal transduction pathway of.
the patched protein.
The terms "epithelia", "epithelial" and "epithelium" refer to the cellular covering of internal and external body surfaces (cutaneous, mucous and serous), including the glands and other structures derived therefrom, e.g., corneal, esophegeal, epidermal, and hair follicle epithelial cells. Other exemplary epithlelial tissue includes: olfactory epithelium, which is the pseudostratified epithelium lining the olfactory region of the nasal cavity, and containing the receptors for the_sense of smell; glandular epithelium, which refers to epithelium composed of secreting cells; squamous epithelium, which refers to epithelium composed of flattened plate-like cells. The term epithelium can also refer to transitional epithelium, which that characteristically found lining hollow organs that are subject to great mechanical change due to contraction and distention, e.g. tissue which represents a transition between stratified squamous and columnar epithelium.
The term "epithelialization" refers to healing by the growth of epithelial tissue over a denuded surface.
The term "skin" refers to the outer protective covering of the body, consisting of the corium and the epidermis, and is understood to include sweat and sebaceous glands, as well as hair follicle structures. Throughout the present application; the adjective "cutaneous" may be used, and should be understood to refer generally to attributes of the skin, as appropriate to the context in which they are used.
The term "epidermis" refers to the outermost and nonvascular layer of the skin, derived from the embryonic ectoderm, varying in thickness from 0.07-1.4 mm. On the palmar and plantar surfaces it comprises, from within outward, five layers: basal layer composed of columnar cells arranged perpendicularly; prickle-cell or spinous layer composed of flattened polyhedral cells with short processes or spines; granular layer composed of flattened granular cells; clear layer composed of several layers of clear, transparent cells in which the nuclei are indistinct or absent; and horny layer composed of flattened, cornified non-nucleated cells. In the epidermis of the general body surface, the clear layer is usually absent.
The "corium" or "dermis" refers to the layer of the skin deep to the epidermis.
consisting of a dense bed of vascular connective tissue, and containing the nerves and terminal organs of sensation. The hair roots, and sebaceous and sweat glands are structures of the epidermis which are deeply embedded in the dermis.
The term "nail" refers to the horny cutaneous plate on the dorsal surface of the distal end of a finger or toe.
The term "epidermal gland" refers to an aggregation of cells associated with the epidermis and specialized to secrete or excrete materials not related to their ordinary metabolic needs. For example, "sebaceous glands" are holocrine glands in the corium that secrete an oily substance and sebum. The term "sweat glands" refers to glands that secrete sweat, situated in the corium or subcutaneous tissue, opening by a duct on the body surface.
The term "hair" refers to a threadlike structure, especially the specialized epidermal structure composed of keratin and developing from a papilla sunk in the corium, produced only by mammals and characteristic of that group of animals. Also, the aggregate of such hairs. A
"hair follicle" refers to one of the tubular-invaginations of the epidermis enclosing the hairs, and from which the hairs grow; and "hair follicle epithelial cells" refers to epithelial cells which surround the dermal papilla in the hair follicle, e.g., stem cells, outer root sheath cells, matrix cells, and inner root sheath cells. Such cells may be normal non-malignant cells, or transformed/immortalized cells.
The term "nasal epithelial tissue" refers to nasal and olfactory epithelium.
"Excisional wounds" include tears, abrasions, cuts, punctures or lacerations in the epithelial layer of the skin and may extend into the dermal layer and even into subcutaneous fat and beyond. Excisional wounds can result from surgical procedures or from accidental penetration of the skin.
"Burn wounds" refer to cases where large surface areas of skin have been removed or lost from an individual due to heat and/or chemical agents.
"Dermal skin ulcers" refer to lesions on the skin caused by superficial loss of tissue, usually with inflammation. Dermal skin ulcers which can be treated by the method of the present invention include decubitus ulcers, diabetic ulcers, venous stasis ulcers and arterial ulcers. Decubitus wounds refer to chronic ulcers that result from pressure applied to areas of the skin for extended periods of time. Wounds of this type are often called bedsores or WO 99/20298 PCT/tJS98IZ2227 pressure sores. Venous stasis ulcers result from the stagnation of blood or other fluids from-defective veins. Arterial ulcers refer to necrotic skin in the area around arteries having poor blood flow.
"Dental tissue" refers to tissue in the mouth which is similar to epithelial tissue, for example gum tissue. The method of the present invention is useful for treating periodontal disease.
"Internal epithelial tissue" refers to tissue inside the body which has characteristics similar to the epidermal layer in the skin. Examples include the lining of the intestine. The method of the present invention is useful for promoting the healing of certain internal wounds, for example wounds resulting from surgery.
A "wound to eye tissue" refers to severe dry eye syndrome, corneal ulcers and abrasions and ophthalmic surgical wounds.
Throughout this application, the term "proliferative skin disorder" refers to any disease/disorder of the skin marked by unwanted or aberrant proliferation of cutaneous tissue.
These conditions are typically characterized by epidermal cell proliferation or incomplete cell differentiation, and include, for example, X-linked ichthyosis, psoriasis, atopic dermatitis, allergic contact dermatitis, epidermolytic hyperkeratosis, and seborrheic dermatitis. For example, epidermodysplasia is a form of faulty development of the epidermis..
Another example is "epidermolysis", which refers to a loosened state of the epidermis with formation of blebs and bullae either spontaneously or at the site of trauma.
The term "carcinoma" refers to a malignant new growth made up of epithelial cells tending to infiltrate surrounding tissues and to give rise to metastases.
Exemplary carcinomas include: "basal cell carcinoma", which is an epithelial tumor of the skin that, while seldom metastasizing, has potentialities for local invasion and destruction;
"squamous cell carcinoma", which refers to carcinomas arising from squamous epithelium and having cuboid cells;
"carcinosarcoma". which include malignant tumors composed of carcinomatous and sarcomatous tissues; "adenocystic carcinoma", carcinoma marked by cylinders or bands of hyaline or mucinous stroma separated or surrounded by nests or cords of small epithelial cells, occurring in the mammary and salivary glands, and mucous glands of the respiratory tract;
"epidermoid carcinoma", which refers to cancerous cells which tend to differentiate in the same way as those of the epidermis; i.e., they tend to form prickle cells and undergo cornification; "nasopharyngeal carcinoma", which refers to a malignant tumor arising in the epithelial lining of the space behind the nose; and "renal cell carcinoma", which pertains to carcinoma of the renal parenchyma composed of tubular cells in varying arrangements.
Another carcinomatous epithelial growth is "papillomas", which refers to benign tumors derived from epithelium and having a papillomavirus as a causative agent; and "epiderznoidomas", which refers to a cerebral or meningeal tumor formed by inclusion of ectodermal elements at the time of closure of the neural groove.
As used herein, the term "psoriasis" refers to a hyperproliferative skin disorder which alters the skin's regulatory mechanisms. In particular, lesions are formed which involve primary and secondary alterations in epidermal proliferation, inflammatory responses of the skin, and an expression of regulatory molecules such as lymphokines and inflammatory factors. Psoriatic skin is morphologically characterized by an increased turnover of epidermal cells, thickened epidermis, abnormal keratinization, inflammatory cell infiltrates into the I S dermis layer and polymorphonuclear leukocyte infiltration into the epidermis layer resulting in an increase in the basal cell cycle. Additionally, hyperkeratotic and parakeratotic cells are present.
The term "keratosis" refers to proliferative skin disorder characterized by hyperplasia of the horny layer of the epidermis. Exemplary keratotic disorders include keratosis follicularis, keratosis palmaris et plantaris, keratosis pharyngea, keratosis pilaris, and actinic keratosis.
As used herein, "proliferating" and "proliferation" refer to cells undergoing mitosis.
As used herein, "transformed cells" refers to cells which have spontaneously converted to a state of unrestrained growth, i.e., they have acquired the ability to grow through an indefinite number of divisions in culture. Transformed cells may be characterized by such terms as neoplastic, anaplastic and/or hyperplastic, with respect to their loss of growth control.
As used herein, "immortalized cells" refers to cells which have been altered via chemical and/or recombinant means such that the cells have the ability to grow through an indefinite number of divisions in culture.

A "patient" or "subject" to be treated by the subject method can mean either a human or.
non-human animal.
The term "cosmetic preparation" refers to a form of a pharmaceutical preparation which is formulated for topical administration.
An "effective amount" of, e.g., a hedgehog therapeutic, with respect to the subject method of treatment, refers to an amount of, e.g., a hedgehog polypeptide in a preparation which, when applied as part of a desired dosage regimen brings about a change in the rate of cell proliferation and/or the state of differentiation of a cell so as to produce an amount of epithelial cell proliferation according to clinically acceptable standards for the disorder to be treated or the cosmetic purpose.
The "growth state" of a cell refers to the rate of proliferation of the cell and the state of differentiation of the cell.
"Homology" and "identity" each refer to sequence similarity between two polypeptide sequences, with identity being a more strict comparison. Homology and identity can each be determined by comparing a position in each sequence which may be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same amino acid residue, then the polypeptides can be referred to as identical at that position; when the equivalent site is occupied by the same amino acid (e.g., identical) or a similar amino acid (e.g., similar in steric and/or electronic nature), then the molecules can be refered to as homologous at that position. A percentage of homology or identity between sequences is a function of the number of matching or homologous positions shared by the sequences. An "unrelated" or "non-homologous" sequence shares less than 40 percent identity, though preferably less than 25 percent identity, with an AR sequence of the present invention.
The term "corresponds to", when referring to a particular polypeptide or nucleic acid sequence is meant to indicate that the sequence of interest is identical or homologous to the reference sequence to which it is said to correspond.
The terms "recombinant protein", "heterologous protein" and "exogenous protein" are used interchangeably throughout the specification and refer to a polypeptide which is produced by recombinant DNA techniques, wherein generally, DNA encoding the polypeptide is inserted into a suitable expression construct which is in turn used to transform a host cell to-produce the heterologous protein. That is, the polypeptide is expressed from a heterologous nucleic acid.
A "chimeric protein" or "fusion protein" is a fusion of a first amino acid sequence encoding a hedgehog polypeptide with a second amino acid sequence defining a domain foreign to and not substantially homologous with any domain of hh protein. A
chimeric protein may present a foreign domain which is found (albeit in a different protein) in an organism which also expresses the first protein, or it may be an "interspecies", "intergenic", etc. fusion of protein structures expressed by different kinds of organisms. In general, a fusion protein can be represented by the general formula (X)"{hh)m-(Y)n, wherein hh represents all or a portion of the hedgehog protein, X and Y each independently represent an amino acid sequences which are not naturally found as a polypeptide chain contiguous with the hedgehog sequence, m is an integer greater than or equal to 1, and each occurrence of n is, independently, 0 or an integer greater than or equal to 1 (n and m are preferably no greater than S or 10).
lll. Exemplary Applications of Method and Compositions The subject method has wide applicability to the treatment or prophylaxis of disorders afflicting epithelial tissue, as well as in cosmetic uses. In general, the method can be characterized as including a step of administering to an animal an amount of a ptc or hedgehog therapeutic effective to alter the proliferative state of a treated epithelial tissue. The mode of administration and dosage regimens will vary depending on the epithelial tissues) which is to be treated. For example, topical formulations will be preferred where the treated tissue is epidermal tissue, such as dermal or mucosal tissues. Likewise, as described in further detail below, the use of a particular ptc or hedgehog therapeutic, e.g., an agonist or antagonist, will depend on whether proliferation of cells of the treated tissue is desired or intended to be prevented.
A method which "promotes the healing of a wound" results in the wound healing more quickly as a result of the treatment than a similar wound heals in the absence of the treatment.

WO 99/20298 PCTlUS98122227 "Promotion of wound healing" can also mean that the method causes the proliferation and growth of, inter alia, keratinocytes, or that the wound heals with less scarring, less wound contraction, less collagen deposition and more superficial surface area. In certain instances, "promotion of wound healing" can also mean that certain methods of wound healing have improved success rates, (e.g. the take rates of skin grafts,) when used together with the method of the present invention.
Complications are a constant risk with wounds that have not fully healed and remain open. Although most wounds heal quickly without treatment, some types of wounds resist healing. Wounds which cover large surface areas also remain open for extended periods of time. In one embodiment of the present invention, the subject method can be used to accelerate the healing of wounds involving epithelial tissues, such as resulting from surgery, burns, inflammation or irritation. Certain of the hedgehog and ptc therapeutic formulations (e.g., proliferative forms) of the present invention can also be applied prophylactically, such as in the form of a cosmetic preparation, to enhance tissue regeneration processes, e.g., of the skin, hair and/or fingernails.
Despite significant progress in reconstructive surgical techniques, scarnng can be an important obstacle in regaining normal function and appearance of healed skin.
This is particularly true when pathologic scarring such as keloids or hypertrophic scars of the hands or face causes functional disability or physical deformity. In the severest circumstances, such scarring may precipitate psychosocial distress and a life of economic deprivation. Wound repair includes the stages of hemostasis, inflammation, proliferation, and remodeling. The proliferative stage involves multiplication of fibroblasts and endothelial and epithelial cells.
Through the use of the subject method, the rate of proliferation of epithelial cells in and proximal to the wound can be controlled in order to accelerate closure of the wound and/or minimize the formation of scar tissue.
Full and partial thickness burns are an example of a wound type which often covers large surface areas and therefore requires prolonged periods of time to heal.
As a result, life-threatening complications such as infection and loss of bodily fluids often arise. In addition, healing in burns is often disorderly, resulting in scarring and disfigurement.
In some cases wound contraction due to excessive collagen deposition results in reduced mobility of muscles in the vicinity of the wound. The compositions and method of the present invention can be used to accelerate the rate of healing of burns and to promote healing processes that result in more desirable cosmetic outcomes and less wound contraction and scarring.
Severe burns which cover large areas are often treated by skin autografts taken from undamaged areas of the patient's body. The subject method can also be used in conjunction with skin grafts to impove "take" rates of the graft by accelerating growth of both the grafted skin and the patient's skin that is proximal to the graft.
Dermal ulcers are yet another example of wounds that are amenable to treatment by the subject method, e.g., to cause healing of the ulcer and/or to prevent the ulcer from becoming a chronic wound. For example, one in seven individuals with diabetes develop dermal ulcers on their extremities, which are susceptible to infection. Individuals with infected diabetic ulcers often require hospitalization, intensive services, expensive antibiotics, and, in some cases, amputation. Dermal ulcers, such as those resulting from venous disease (venous stasis ulcers), excessive pressure (decubitus ulcers) and arterial ulcers also resist healing.
The prior art treatments are generally limited to keeping the wound protected, free of infection and, in some cases, to restore blood flow by vascular surgery. According to the present method, the afflicted area of skin can be treated by a therapy which includes a hedgehog or ptc therapeutic which promotes epithelization of the wound, e.g., accelerates the rate of the healing of the skin ulcers.
The present treatment can also be effective as part of a therapeutic regimen for treating ora! and paraoral ulcers, e.g. resulting from radiation and/or chemotherapy.
Such ulcers commonly develop within days after chemotherapy or radiation therapy. These ulcers usually begin as small, painful irregularly shaped lesions usually covered by a delicate gray necrotic membrane and surrounded by inflammatory tissue. In many instances, lack of treatment results in proliferation of tissue around the periphery of the lesion on an inflammatory basis.
For instance, the epithelium bordering the ulcer usually demonstrates proliferative activity, resulting in loss of continuity of surface epithelium. These lesions, because of their size and loss of epithelial integrity, lend the body to potential secondary infection.
Routine ingestion of food and water becomes a very painful event and, if the ulcers proliferate throughout the alimentary canal, diarrhea usually is evident with all its complicating factors. According to the WO 99/Z0298 PCT/US98/22Z2~

present invention, a treatment for such ulcers which includes application of an hedgehog.
therapeutic can reduce the abnormal proliferation and differentiation of the affected epithelium, helping to reduce the severity of subsequent inflammatory events.
In another exemplary embodiment, the subject method is provided for treating or preventing gastrointestinal diseases. Briefly, a wide variety of diseases are associated with disruption of the gastrointestinal epithelium or villi, including chemotherapy-and radiation-therapy-induced enteritis (i.e. gut toxicity) and mucositis, peptic ulcer disease, gastroenteritis and colitis, villus atrophic disorders, and the like. For example, chemotherapeutic agents and radiation therapy used in bone marrow transplantation and cancer therapy affect rapidly proliferating cells in both the hematopoietic tissues and small intestine, leading to severe and often dose-limiting toxicities. Damage to the small intestine mucosal barrier results in serious complications of bleeding and sepsis. The subject method can be used to promote proliferation of gastrointenstinal epithelium and thereby increase the tolerated doses for radiation and chemotherapy agents. Effective treatment of gastrointestinal diseases may be determined by several criteria, including an enteritis score, other tests well known in the art.
The subject method and compositions can also be used to treat wounds resulting from dermatological diseases, such as lesions resulting from autoimmune disorders such as psoriasis. Atopic dermititis refers to skin trauma resulting from allergies associated with an immune response caused by allergens such as pollens, foods, dander, insect venoms and plant toxins.
With age, the epidermis thins and the skin appendages atrophy. Hair becomes sparse and sebaceous secretions decrease, with consequent susceptibility to dryness, chapping, and fissuring. The dermis diminishes with loss of elastic and collagen fibers.
Moreover, keratinocyte proliferation (which is indicative of skin thickness and skin proliferative capacity) decreases with age. An increase in keratinocyte proliferation is believed to conteract skin aging, i.e., wrinkles, thickness, elasticity and repair. According to the present invention. a proliferative form of a hedgehog or ptc therapeutic can be used either therapeutically or cosmetically to counteract, at least for a time, the effects of aging on skin.
The subject method can also be used in treatment of a wound to eye tissue.
Generally, damage to corneal tissue, whether by disease, surgery or injury, may affect epithelial and/or WO 99/20298 PCTNS98l22227 endothelial cells, depending on the nature of the wound. Corneal epithelial cells are the non-keratinized epithelial cells lining the external surface of the cornea and provide a protective barrier against the external environment. Corneal wound healing has been of concern to both clinicians and researchers. Opthomologists are frequently confronted with corneal dystrophies and problematic injuries that result in persistent and recurrent epithelial erosion, often leading to permanent endothelial loss. The use of proliferative forms of the subject hedgehog and/or other ptc therapeutics can be used in these instances to promote epithelialization of the affected corneal tissue.
To further illustrate, specific disorders typically associated with epithelial cell damage in the eye, and for which the subject method can provide beneficial treatment, include persistent corneal epithelial defects, recurrent erosions, neurotrophic corneal ulcers, keratoconjunctivitis sicca, microbial corneal ulcers, viral cornea ulcers, and the like. Surgical procedures typically causing injury to the epithelial cell layers include laser procedures performed on the ocular surface, any refractive surgical procedures such as radial keratotomy and astigmatic keratotomy, conjunctiva) flaps, conjunctiva) transplants, epikeratoplasty, and corneal scraping. Moreover, superficial wounds such as scrapes, surface erosion, inflammation, etc. can cause lose of epithelial cells. According to the present invention, the corneal epithelium is contacted with an amount of a ptc or hedgehog therapeutic effective to cause proliferation of the corneal epithelial cells to appropriately heal the wound.
In other embodiments, antiproliferative preparations of hedgehog or ptc therapeutics can be used to inhibit lens epithelial cell proliferation to prevent post-operative complications of extracapsular cataract extraction. Cataract is an intractable eye disease and various studies on a treatment of cataract have been made. But at present, the treatment of cataract is attained by surgical operations. Cataract surgery has been applied for a long time and various operative methods have been examined. Extracapsular lens extraction has become the method of choice for removing cataracts. The major medical advantages of this technique over intracapsular extraction are lower incidence of aphakic cystoid macular edema and retinal detachment.
Extracapsular extraction is also required for implantation of posterior chamber type intraocular lenses which are now considered to be the lenses of choice in most cases.

However, a disadvantage of extracapsular cataract extraction is the high incidence of posterior lens capsule opacification, often called after-cataract, which can occur in up to 50%
of cases within three years after surgery. After-cataract is caused by proliferation of equatorial and anterior capsule lens epithelial cells which remain after extracapsular lens extraction.
These cells proliferate to cause Sommerling rings, and along with fibroblasts which also deposit and occur on the posterior capsule, cause opacification of the posterior capsule, which interferes with vision. Prevention of after-cataract would be preferable to treatment. To inhibit secondary cataract formation, the subject method provides a means for inhibiting proliferation of the remaining lens epithelial cells. For example, such cells can be induced to remain quiescent by instilling a solution .containing an antiproliferative hedgehog or ptc therapeutic preparation into the anterior chamber of the eye after lens removal.
Furthermore, the solution can be osmotically balanced to provide minimal effective dosage when instilled into the anterior chamber of the eye, thereby inhibiting subcapsular epithelial growth with some specificity.
The subject method can also be used in the treatment of corneopathies marked by corneal epithelial cell proliferation, as for example in ocular epithelial .disorders such as epithelial downgrowth or squamous cell carcinomas of the ocular surface.
The maintenance of tissues and organs ex vivo is also highly desirable. Tissue replacement therapy is well established in the treatment of human disease. For example, more than 40,000 corneal transplants were performed in the United States in 1996.
Human epidermal cells can be grown in vitro and used to populate burn sites and chronic skin ulcers and other dermal wounds. The subject method can be used to accelerate the growth of epithelial tissue in vitro, as well as to accelerate the grafting of the cultured epithelial tissue to an animal host The present method can be used for improving the "take rate" of a skin graft.
Grafts of epidermal tissue can, if the take rate of the graft is to long, blister and shear, decreasing the likelihood that the autograft will "take", i.e. adhere to the wound and form a basement membrane with the underlying granulation tissue. Take rates can be increased by the subject method by inducing proliferation of the keratinocytes. The method of increasing take rates comprises contacting the skin autograft with an effective wound healing amount of a hedgehog or ptc therapeutic compositions described in the method of promoting wound healing and in the method of promoting the growth and proliferation of keratinocytes, as described above.
Skin equivalents have many uses not only as a replacement for human or animal skin for skin grafting, but also as test skin for determining the effects of pharmaceutical substances and cosmetics on skin. A major difficulty in pharmacological, chemical and cosmetic testing is the difficulties in determining the efficacy and safety of the products on skin. One advantage of the skin equivalents of the invention is their use as an indicator of the effects produced by such substances through in vitro testing on test skin.
Thus, in one embodiment of the subject method can be used as part of a protocol for skin grafting of, e.g., denuded areas, granulating wounds and burns. The use of proliferative hedgehog and/or ptc therapeutics can enhance such grafting techniques as split thickness autografts and epidermal autografts (cultured autogenic keratinocytes) and epidermal allografts (cultured allogenic keratinocytes). In the instance of the allograft, the use of the subject method to enhance the formation of skin equivalents in culture helps to provide/maintain a ready supply of such grafts (e.g., in tissue banks) so that the patients might be covered in a single procedure with a material which allows permanent healing to occur.
In this regard, the present invention also concerns composite living skin equivalents comprising an epidermal layer of cultured keratinocyte cells which have been expanded by treatment with a hedgehog or other ptc therapeutic. The subject method can be used as part of a process for the preparation of composite living skin equivalents. In an illustrative embodiment, such a method comprises obtaining a skin sample. treating the skin sample enzymically to separate the epidermis from the dermis, treating the epidermis enzymically to release the keratinocyte cells, culturing, in the presence of a hedgehog or ptc therapeutic, the epidermal keratinocytes until confluence, in parallel, or separately, treating the dermis enzymatically to release the fibroblast cells. culturing the fibroblasts cells until sub-confluence, inoculating a porous, cross-linked collagen sponge membrane with the cultured fibroblast cells, incubating the inoculated collagen sponge on its surface to allow the growth of the fibroblast cells throughout the collagen sponge, and then inoculating it with cultured keratinocyte cells, and further incubating the composite skin equivalent complex in the presence of a hedgehog or ptc therapeutic to promote the growth of the cells.

In other embodiments, skin sheets containing both epithelial and mesenchymal layers can be isolated in culture and expanded with culture media supplemented with a proliferative form of a hedgehog or ptc therapeutic.
Any skin sample amenable to cell culture techniques can be used in accordance with the present invention. The skin samples may be autogenic or allogenic.
In another aspect of the invention, the subject method can be used in conjunction with various periodontal procedures in which control of epithelial cell proliferation in and around periodontal tissue is desired.
In one embodiment, proliferative forms of the hedgehog and ptc therapeutics can be used to enhance reepithelialization around natural and prosthetic teeth, e.g., to promote formation of gum tissue.
In another embodiment, antiproliferative ptc therapeutics can find application in the treatment of peridontal disease. It is estimated that in the United States alone, there are in excess of 125 million adults with periodontal disease in varying forms.
Periodontal disease starts as inflammatory lesions because of specific bacteria localizing in the area where the gingiva attaches to the tooth. Usually first to occur is a vascular change in the underlying connective tissue. Inflammation in the connective tissue stimulates the following changes in the epithelial lining of the sulcus and in the epithelial attachment:
increased mitotic activity in the basal epithelial layer; increased producing of keratin with desquamation;
cellular desquamation adjacent to the tooth surface tends to deepen the pocket;
epithelial cells of the basal layer at the bottom of the sulcus and in the area of attachment proliferate into the connective tissue and break up of the gingival fibers begins to occur, wherein dissolution of the connective tissue results in the fonmation of an open lesion. The application of hedgehog preparations to the periodontium can be used to inhibit proliferation of epithelial tissue and thus prevent further periodontoclastic development.
In yet another aspect, the subject method can be used to help control guided tissue regeneration, such as when used in conjunction with bioresorptable materials.
For example, incorporation of periodontal implants, such as prosthetic teeth, can be facilitated by the instant method. Reattachment of a tooth involves both formation of connective tissue fibers and re-epithelization of the tooth pocket. The subject methodtreatment can be used to accelerate wo ~noz98 rcrius9snzzz~

tissue reattachment by controlling the mitotic function of basal epithelial cells in early stages of wound healing.
Yet another aspect of the present invention relates to the use of hedgehog therapeutic preparations to control hair growth. Hair is basically composed of keratin, a tough and insoluble protein; its chief strength lies in its disulphide bond of cystine.
Each individual hair comprises a cylindrical shaft and a root, and is contained in a follicle, a flask-like depression in the skin. The bottom of the follicle contains a finger-like projection termed the papilla, which consists of connective tissue from which hair grows, and through which blood vessels supply the cells with nourishment. The shaft is the part that extends outwards from the skin surface, whilst the root has been described as the buried part of the hair. The base of the root expands into the hair bulb, which rests upon the papilla. Cells from which the hair is produced grow in the bulb of the follicle; they are extruded in the form of fibers as the cells proliferate in the follicle. Hair "growth" refers to the formation and elongation of the hair fiber by the dividing cells.
As is well known in the art, the common hair cycle is divided into three stages: anagen, catagen and telogen. During the active phase (anagen) , the epidermal stem cells of the denmal papilla divide rapidly. Daughter cells move upward and differentiate to form the concentric layers of the hair itself. The transitional stage, catagen, is marked by the cessation of mitosis of the stem cells in the follicle. The resting stage is known as telogen, where the hair is retained within the scalp for several weeks before an emerging new hair developing below it dislodges the telogen-phase shaft from its follicle. From this model it has become clear that the larger the pool of dividing stem cells that differentiate into hair cells, the more hair growth occurs. Accordingly, methods for increasing or reducing hair growth can be carried out by potentiating or inhibiting, respectively, the proliferation of these stem cells.
In one embodiment, the subject method provides a means for altering the dynamics of the hair growth cycle to induce proliferation of hair follicle cells, particularly stem cells of the hair follicle. The subject compositions and method can be used to increase hair follicle size and the rate of hair growth in wanm-blooded animals, such as humans, e.g., by promoting proliferation of hair follicle stem cells. In one embodiment, the method comprises administering to the skin in the area in which hair growth is desired an amount of hedgehog or wo ~noz9s pc~r~rs9snzzr ptc therapeutic sufficient to increase hair follicle size and/or the rate of hair growth in the animal. Typically, the composition will be administered topically as a cream, and will be applied on a daily basis until hair growth is observed and for a time thereafter sufficient to maintain the desired amount of hair growth. This method can have applications in the promotion of new hair growth or stimulation of the rate of hair growth, e.g., following chemotherapeutic treatment or for treating various forms of alopecia, e.g., male pattern baldness. For instance, one of several biochemical cellular and molecular disturbances that occur during the anagen phase or catagen phase of subjects with androgenic alopecia can be corrected or improved by treatment using the subject method, e.g., in the functioning or formation of the stem cells, their migration process or during the mitosis phase of keratin production within the follicular papilla and matrix.
In other embodimemts, cerain of the hedgehog and ptc therapeutics (e.g., antiproliferative forms) can be employed as a way of reducing the growth of human hair as opposed to its conventional removal by cutting, shaving, or depilation. For instance, the present method can be used in the treatment of trichosis characterized by abnormally rapid or dense growth of hair, e.g. hypertrichosis. In an exemplary embodiment, hedgehog antagonists can be used to manage hirsutism, a disorder marked by abnormal hairiness. The subject method can also provide a process for extending the duration of depilation.
Moreover, because a hedgehog antagonist (or ptc agonist) will often be cytostatic to epithelial cells, rather than cytotoxic, such agents can be used to protect hair follicle cells from cytotoxic agents which require progression into S-phase of the cell-cycle for efficacy, e.g.
radiation-induced death. Treatment by the subject method can provide protection by causing the hair follicle cells to become quiescent, e.g., by inhibiting the cells from entering S phase, and thereby preventing the follicle cells from undergoing mitotic catastrophe or programmed cell death. For instance, hedgehog antagonists can be used for patients undergoing chemo- or radiation-therapies which ordinarily result in hair loss. By inhibiting cell-cycle progression during such therapies, the subject treatment can protect hair follicle cells from death which might otherwise result from activation of cell death programs. After the therapy has concluded, the hedgehog or ptc treatment can also be removed with concommitant relief of the inhibition of follicle cell proliferation.

The subject method can also be used in the treatment of folliculitis, such as folliculitis.
decalvans, folliculitis ulerythematosa reticulata or keloid folliculitis. For example, a cosmetic prepration of an hedgehog therapeutic can be applied topically in the treatment of pseudofolliculitis, a chronic disorder occurring most often in the submandibular region of the neck and associated with shaving, the characteristic lesions of which are erythematous papules and pustules containing buried hairs.
In another aspect of the invention, antiproliferative forms of the subject hedgehog and ptc therapeutics can be used to induce differentiation of epithelially-derived tissue. Such forms of these molecules can provide a basis for differentiation therapy for the treatment of hyperplastic and/or neoplastic conditions involving epithelial tissue. For example, such preparations can be used for the treatment of cutaneous diseases in which there is abnormal proliferation or growth of cells of the skin.
For instance, the pharmaceutical preparations of the invention are intended for the treatment of hyperplastic epidermal conditions, such as keratosis, as well as for the treatment of neoplastic epidermal conditions such as those characterized by a high proliferation rate for various skin cancers, as for example basal cell carcinoma or squamous cell carcinoma. The subject method can also be used in the treatment of autoimmune diseases affecting the skin, in particular, of dermatological diseases involving morbid proliferation and/or keratinization of the epidermis, as for example, caused by psoriasis or atopic dermatosis.
Many common diseases of the skin, such as psoriasis, squamous cell carcinoma, keratoacanthoma and actinic keratosis are characterized by localized abnormal proliferation and growth. For example, in psoriasis, which is characterized by scaly, red, elevated plaques on the skin, the keratinocytes are known to proliferate much more rapidly than normal and to differentiate less completely.
In one embodiment, the preparations of the present invention are suitable for the treatment of dermatological ailments linked to keratinization disorders causing abnormal proliferation of skin cells, which disorders may be marked by either inflammatory or non-inflammatory components. To illustrate, therapeutic preparations of a ptc agonist, e.g., which promotes quiescense or differentiation can be used to treat varying forms of psoriasis, be they cutaneous, mucosal or ungual. Psoriasis, as described above, is typically characterized by epidermal keratinocytes which display marked proliferative activation and differentiation along a "regenerative" pathway. Treatment with an antiproliferative embodiment of the subject method can be used to reverse the pathological epidermal activiation and can provide a basis for sustained remission of the disease.
S A variety of other keratotic lesions are also candidates for treatment with the subject antiproliferative preparations. Actinic keratoses, for example, are superFcial inflanunatory premalignant tumors arising on sun-exposed and irradiated skin. The lesions are erythematous to brown with variable scaling. Current therapies include excisional and cryosurgery. These treatments are painful, however, and often produce cosmetically unacceptable scarring.
Accordingly, treatment of keratosis, such as actinic keratosis, can include application, preferably topical, of a ptc agonist composition in amounts sufficient to inhibit hyperproliferation of epidermal/epidermoid cells of the lesion.
Acne represents yet another dermatologic ailment which may be treated with an antiproliferative embodiment of the subject method. Acne vulgaris, for instance, is a multifactorial disease most commonly occurring in teenagers and young adults, and is characterized by the appearance of inflammatory and noninflammatory lesions on the face and upper trunk. The basic defect which gives rise to acne vulgaris is hypercornification of the duct of a hyperactive sebaceous gland. Hypercornification blocks the normal mobility of skin and follicle microorganisms, and in so doing, stimulates the release of lipases by Propinobacterium acnes and Staphylococcus epidermidis bacteria and Pitrosporum ovate, a yeast. Treatment with an antiproliferative form of a hedgehog or ptc therapeutic, particularly topical preparations, may be useful for preventing the transitional features of the ducts, e.g.
hypercornification, which lead to lesion formation. The subject treatment may further include, for example, antibiotics, retinoids and antiandrogens.
The present invention also provides a method for treating various forms of dermatitis.
Dermatitis is a descriptive term referring to poorly demarcated lesions which are either pruritic, erythematous, scaley, blistered, weeping, fissured or crusted. These lesions arise from any of a wide variety of causes. The most common types of dermatitis are atopic, contact and diaper dermatitis. For instance, seborrheic dermatitis is a chronic, usually pruritic, dermatitis with erythema, dry, moist, or greasy scaling, and yellow crusted patches on various areas, especially the scalp, with exfoliation of an excessive amount of dry scales stasis dermatitis, an often chronic, usually eczematous dermatitis. Actinic dermatitis is dermatitis that due to exposure to actinic radiation such as that from the sun, ultraviolet waves or x- or gamma-radiation. According to the present invention, the subject hedgehog or ptc therapeutic preparations can be used in the treatment and/or prevention of certain symptoms of dermatitis caused by unwanted proliferation of epithelial cells. Such therapies for these various forms of dermatitis can also include topical and systemic corticosteroids, antipuritics, and antibiotics.
Also included in ailments which may be treated by the subject method are disorders specific to non-humans, such as mange.
IV. Exemplary hedgehog therapeutic compounds.
The hedgehog therapeutic compositions of the subject method can be generated by any of a variety of techniques, including purification of naturally occurring proteins, recombinantly produced proteins and synthetic chemistry. Polypeptide forms of the hedgehog therapeutics 1 S are preferably derived from vertebrate hedgehog proteins, e.g., have sequences corresponding to naturally occurring hedgehog proteins, or fragments thereof, from vertebrate organisms.
However, it will be appreciated that the hedgehog polypeptide can correspond to a hedgehog protein (or fragment thereof) which occurs in any metazoan organism.
The various naturally-occurring hedgehog proteins from which the subject therapeutics can be derived are characterized by a signal peptide, a highly conserved N-terminal region, and a more divergent C-terminal domain. In addition to signal sequence cleavage in the secretory pathway (Lee, J.J. et al. (1992) Ce1171:33-50; Tabata, T. et al. (1992) Genes Dev. 2635-2645;
Chang, D.E. et al. (1994) Development 120:3339-3353), hedgehog precursor proteins naturally undergo an internal autoproteolytic cleavage which depends on conserved sequences in the C-terminal portion (Lee et al. (1994) Science 266:1528-1537; Porter et al.
(1995) Nature 374:363-366). This autocleavage leads to a 19 kD N-terminal peptide and a C-terminal peptide of 26-28 kD (Lee et al. ( 1992) supra; Tabata et al. ( 1992) supra; Chang et al. ( 1994) supra;
Lee et al. (1994) supra; Bumcrot, D.A., et al. (1995) Mol. Cell. Biol. 15:2294-2303; Porter et al. (1995) supra; Ekker, S.C. et al. (1995) Curr. Biol. 5:944-955; Lai, C.J.
et al. (1990 Development 121:2349-2360). The N-terminal peptide stays tightly associated with the surface of cells in which it was synthesized, while the C-terminal peptide is freely diffusible both in vitro and in vivo (Lee et al. ( 1994) supra; Bumcrot et al. ( 1995) supra; Mart', E. et al.
(1995) Development 121:2537-2547; Roelink, H. et al. (1995) Cell 81:445-455).
Cell surface retention of the N-terminal peptide is dependent on autocleavage, as a truncated form of S hedgehog encoded by an RNA which terminates precisely at the normal position of internal cleavage is diffusible in vitro (Porter et al. (1995) supra) and in vivo (Porter, J.A. et al. {1996) Cell 86, 21-34). Biochemical studies have shown that the autoproteolytic cleavage of the hedgehog precursor protein proceeds through an internal thioester intermediate which subsequently is cleaved in a nucleophilic substitution. It is suggested that the nucleophile is a small lipophilic molecule, more particularly cholesterol, which becomes covalently bound to the C-terminal end of the N-peptide (Porter et al. ( 1996) supra), tethering it to the cell surface.
The vertebrate family of hedgehog genes includes at least four members, e.g., paralogs of the single drosophila hedgehog gene (SEQ ID No. 19). Three of these members, herein referred to as Desert hedgehog (Dhh), Sonic hedgehog (Shh) and Indian hedgehog (Ihh), apparently exist in all vertebrates, including fish, birds, and mammals. A
fourth member, herein refer ed to as tiggie-winkle hedgehog (Thh), appears specific to fish..
According to the appended sequence listing, (see also Table 1 ) a chicken Shh polypeptide is encoded by SEQ ID
NO:I; a mouse Dhh polypeptide is encoded by SEQ ID NO:2; a mouse Ihh polypeptide is encoded by SEQ ID N0:3; a mouse Shh polypeptide is encoded by SEQ ID N0:4 a zebrafish Shh polypeptide is encoded by SEQ ID NO:S; a human Shh polypeptide is encoded by SEQ ID
N0:6; a human Ihh polypeptide is encoded by SEQ ID N0:7; a human Dhh polypeptide is encoded by SEQ ID No. 8; and a zebrafish Thh is encoded by SEQ ID No. 9.

Table 1 Guide to hedgehog sequences in Sequence Listing Nucleotide Amino Acid Chicken Shh SEQ ID No. SEQ ID No.

Mouse Dhh SEQ ID No. SEQ ID No.

Mouse Ihh SEQ~D No. 3 SEQ ID No.

Mouse Shh SEQ ID No. SEQ ID No.

Zebrafish SEQ ID No. SEQ ID No.
Shh 5 14 Human Shh SEQ ID No. SEQ ID No.

Human Ihh SEQ ID No. SEQ ID No.
7 16 Human Dhh SEQ ID No. SEQ ID No.
8 17 Zebrafish SEQ ID No. SEQ ID No.
Thh 9 18 Drosophila SEQ ID No. SEQ ID No.

In addition to the sequence variation between the various hedgehog homologs, the hedgehog proteins are apparently present naturally in a number of different forms, including a pro-form, a full-length mature form, and several processed fragments thereof.
The pro-form includes an N-terminal signal peptide for directed secretion of the extracellular domain, while the full-length mature form lacks this signal sequence.
As described above, further processing of the mature form occurs in some instances to yield biologically active fragments of the protein. For instance, sonic hedgehog undergoes additional proteolytic processing to yield two peptides of approximately 19 kDa and 27 kDa, the l9kDa fragment corresponding to an proteolytic N-terminal portion of the mature protein.
In addition to proteolytic fragmentation, the vertebrate hedgehog proteins can also be modified post-translationally, such as by glycosylation and/or addition of lipophilic moieties, such as stents, fatty acids, etc., though bacterially produced (e.g.
unmodified) forms of the proteins still maintain certain of the bioactivities of the native protein.
Bioactive fragments of hedgehog polypeptides of the present invention have been generated and are described in great detail in, e.g., PCT publications WO 95/18856 and WO 96/17924.
There are a wide range of lipophilic moieties with which hedgehog polypeptides can be derivatived. The term "lipophilic group", in the context of being attached to a hedgehog polypeptide, refers to a group having high hydrocarbon content thereby giving the group high affinity to lipid phases. A lipophilic group can be, for example, a relatively long chain alkyl or cycloalkyl (preferably n-alkyl) group having approximately 7 to 30 carbons.
The alkyl group WO 99/20298 PCT/US98n2227 may terminate with a hydroxy or primary amine "tail". To further illustrate, lipophilic molecules include naturally-occurring and synthetic aromatic and non-aromatic moieties such as fatty acids, sterols, esters and alcohols, other lipid molecules, cage structures such as adamantane and buckminsterfullerenes, and aromatic hydrocarbons such as benzene, perylene, phenanthrene, anthracene, naphthalene, pyrene, chrysene, and naphthacene.
In one embodiment, the hedgehog polypeptide is modified with one or more sterol moieties, such as cholesterol. See, for example, PCT publication WO 96/17924.
In certain embodiments, the cholesterol is preferably added to the C-terminal glycine were the hedgehog polypeptide corresponds to the naturally-occurring N-terminal proteolytic fragment.
In another embodiment, the hedgehog polypeptide can be modified with a fatty acid moiety, such as a myrostoyl, palmitoyl, stearoyl, or arachidoyl moiety. See, e.g., Pepinsky et al. (1998) J Biol. Chem 273: 14037.
In addition to those effects seen by cholesterol-addition to the C-terminus or fatty acid addition to the N-terminus of extracellular fragments of the protein, at least certain of the biological activities of the hedgehog gene products are unexpectedly potentiated by derivativation of the protein with lipophilic moieties at other sites on the protein and/or by moieties other than cholesterol or fatty acids. Certain aspects of the invention are directed to the use of preparations of hedgehog polypeptides which are modified at sites other than N-terminal or C-terminal residues of the natural processed form of the protein, and/or which are modified at such terminal residues with lipophilic moieties other than a sterol at the C-terminus or fatty acid at the N-terminus.
Particularly useful as lipophilic molecules are alicyclic hydrocarbons, saturated and unsaturated fatty acids and other lipid and phospholipid moieties, waxes, cholesterol, isoprenoids, terpenes and polyalicyclic hydrocarbons including adamantane and buckminsterfullerenes, vitamins, polyethylene glycol or oligoethylene glycol, (CI-C18)-alkyl phosphate diesters, -O-CH2-CH(OH)-O-(C 12-C I 8)-alkyl, and in particular conjugates with pyrene derivatives. The lipophilic moiety can be a lipophilic dye suitable for use in the invention include. but are not limited to, diphenylhexatriene, Nile Red, N-phenyl-I-naphthylamine, Prodan, Laurodan, Pyrene, Perylene, rhodamine, rhodamine B, tetramethylrhodamine, Texas Red, sulforhodamine, I ,1'-didodecyl-WO 99/20298 PG"T/US98/22227 3,3,3',3'tetramethylindocarbocyanine perchlorate, octadecyl rhodamine B and the BODIPY
dyes available from Molecular Probes Inc.
Other exemplary lipophilic moietites include aliphatic carbonyl radical groups include 1- or 2-adamantylacetyl, 3-methyladamant-1-ylacetyl, 3-methyl-3-bromo-1-adamantylacetyl, 1-decalinacetyl, camphoracetyl, camphaneacetyl, noradamantylacetyl, norbornaneacetyl, bicyclo[2.2.2.]-oct-5-eneacetyl, 1-methoxybicyclo(2.2.2.]-oct-5-ene-2-carbonyl, cis-5-norbornene-endo-2,3-dicarbonyl, 5-norbornen-2-ylacetyl, ( 1 R)-( - )-myrtentaneacetyl, 2-norbornaneacetyl, anti-3-oxo-tricyclo[2.2.1.0<2,6> ]-heptane-7-carbonyl, decanoyl, dodecanoyl, dodecenoyl, tetradecadienoyl, decynoyl or dodecynoyl.
The hedgehog polypeptide can be linked to the hydrophobic moiety in a number of ways including by chemical coupling means, or by genetic engineering.
There are a large number of chemical cross-linking agents that are known to those skilled in the art. For the present invention, the preferred cross-linking agents are heterobifunctional cross-linkers, which can be used to link the hedgehog polypeptide and hydrophobic moiety in a stepwise manner. Heterobifunctional cross-linkers provide the ability to design more specific coupling methods for conjugating to proteins, thereby reducing the occurrences of unwanted side reactions such as homo-protein polymers. A wide variety of heterobifunctional cross-linkers are known in the art. These include:
succinimidyl 4-(N-maleimidomethyl) cyclohexane- I-carboxylate (SMCC), m-Maleimidobenzoyl-N-hydroxysuccinimide ester (MBS); N-succinimidyl (4-iodoacetyl) aminobenzoate (SIAB), succinimidyl 4-(p-maleimidophenyl) butyrate (SMPB), I-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDC); 4-succinimidyloxycarbonyl- a-methyl-a-(2-pyridyldithio)-tolune (SMPT), N-succinimidyl 3-(2-pyridyldithio) propionate (SPDP), succinimidyl 6-[3-(2-pyridyldithio) propionate] hexanoate (LC-SPDP). Those cross-linking agents having N-hydroxysuccinimide moieties can be obtained as the N-hydroxysulfosuccinimide analogs, which generally have greater water solubility. In addition, those cross-linking agents having disulfide bridges within the linking chain can be synthesized instead as the alkyl derivatives so as to reduce the amount of linker cleavage in vivo.
In addition to the heterobifunctional cross-linkers, there exists a number of other cross-linking agents including homobifunctional and photoreactive cross-linkers.
Disuccinimidyl suberate (DSS), bismaleimidohexane (BMH) and dimethylpimelimidate~2 HCI (DMP) are examples of useful homobifunctional cross-linking agents, and bis-(13-(4-azidosalicylamido)ethyl]disulfide (BASED) and N-succinimidyl-6(4'-azido-2'-nitrophenyl-amino)hexanoate (SANPAH) are examples of useful photoreactive cross-linkers for use in this invention. For a recent review of protein coupling techniques, see Means et al. (1990) Bioconjugate Chemistry 1:2-12, incorporated by reference herein.
One particularly useful class of heterobifunctional cross-linkers, included above, contain the primary amine reactive group, N-hydroxysuccinimide (NHS), or its water soluble analog N-hydroxysulfosuccinimide (sulfo-NHS). Primary amines (lysine epsilon groups) at alkaline pH's are unprotonated and react by nucleophilic attack on NHS or sulfo-NHS esters.
This reaction results in the formation of an amide bond, and release of NHS or sulfo-NHS as a by-product.
Another reactive group useful as part of a heterobifunctional cross-linker is a thiol reactive group. Common thiol reactive groups include maleimides, halogens, and pyridyl disulfides. Maleimides react specifically with free sulfhydryls (cysteine residues) in minutes, under slightly acidic to neutral (pH 6.5-7.5) conditions. Halogens (iodoacetyl functions) react with -SH groups at physiological pH's. Both of these reactive groups result in the formation of stable thioether bonds.
The third component of the heterobifunctional cross-linker is the spacer arm or bridge.
The bridge is the structure that connects the two reactive ends. The most apparent attribute of the bridge is its effect on steric hindrance. In some instances, a longer bridge can more easily span the distance necessary to link two complex biomolecules. For instance, SMPB has a span of 14.5 angstroms.
Preparing protein-protein conjugates using heterobifunctional reagents is a two-step process involving the amine reaction and the sulfhydryl reaction. For the first step, the amine reaction, the protein chosen should contain a primary amine. This can be lysine epsilon amines or a primary alpha amine found at the N-terminus of most proteins. The protein should not contain free sulfliydryl groups. In cases where both proteins to be conjugated contain free sulflrydryl groups, one protein can be modified so that all sulfhydryls are blocked using for instance, N-ethylmaleimide (see Partis et al. (1983) J. Pro. Chem.
2:263, incorporated by reference herein). Ellman's Reagent can be used to calculate the quantity of sulfhydryls in a particular protein (see for example Ellman et al. ( 1958) Arch. Biochem.
Biophys. 74:443 and Riddles et al. ( 1979) Anal. Biochem. 94:75, incorporated by reference herein).
The reaction buffer should be free of extraneous amines and sulfhydryls. The pH of the reaction buffer should be 7.0-7.5. This pH range prevents maleimide groups from reacting with amines, preserving the maleimide group for the second reaction with sulfhydryls.
The NHS-ester containing cross-linkers have limited water solubility. They should be dissolved in a minimal amount of organic solvent (DMF or DMSO) before introducing the cross-linker into the reaction mixture. The cross-linker/solvent forms an emulsion which will allow the reaction to occur.
The sulfo-NHS ester analogs are more water soluble, and can be added directly to the reaction buffer. Buffers of high ionic strength should be avoided, as they have a tendency to "salt out" the sulfo-NHS esters. To avoid loss of reactivity due to hydrolysis, the cross-linker is added to the reaction mixture immediately after dissolving the protein solution.
The reactions can be more efficient in concentrated protein solutions. The more alkaline the pH of the reaction mixture, the faster the rate of reaction. The rate of hydrolysis of the NHS and sulfo-NHS esters will also increase with increasing pH. Higher temperatures will increase the reaction rates for both hydrolysis and acylation.
Once the reaction is completed, the first protein is now activated, with a sulfhydryl reactive moiety. The activated protein may be isolated from the reaction mixture by simple gel filtration or dialysis. To carry out the second step of the cross-linking, the sulfhydryl reaction, the lipophilic group chosen for reaction with maleimides, activated halogens, or pyridyl disulfides must contain a free sulfhydryl. Alternatively, a primary amine may be modified with to add a sulfhydryl In all cases, the buffer should be degassed to prevent oxidation of sulfhydryl groups.
EDTA may be added to chelate any oxidizing metals that may be present in the buffer. Buffers should be free of any sulfhydryl containing compounds.

Maleimides react specifically with -SH groups at slightly acidic to neutral pH
ranges (6.5-7.5). A neutral pH is sufficient for reactions involving halogens and pyridyl disulfides.
Under these conditions, maleimides generally react with -SH groups within a matter of minutes. Longer reaction times are required for halogens and pyridyl disulfides.
The first sulfl~ydryl reactive-protein prepared in the amine reaction step is mixed with the sulfhydryl-containing lipophilic group under the appropriate buffer conditions. The conjugates can be isolated from the reaction mixture by methods such as gel filtration or by dialysis.
Exemplary activated lipophilic moieties for conjugation include: N-(1-pyrene)maleimide; 2,5-dimethoxystilbene-4'-maleimide, eosin-5-maleimide;
fluorescein-~-maleimide; N-(4-(6-dimethylamino- 2-benzofuranyl)phenyl)maleimide;
benzophenone-4-maleimide; 4-dimethylaminophenylazophenyl- 4'-maleimide (DABMI), tetramethylrhodamine-5-maleimide, tetramethylrhodamine-6-maleimide, Rhodamine RedTM C2 maleimide, N-(5-aminopentyl)maleimide, trifluoroacetic acid salt, N-(2-aminoethyl)maleimide, trifluoroacetic 1 S acid salt, Oregon GreenTM 488 maleimide, N-(2-((2-({(4-azido- 2,3,5,6-tetrafluoro)benzoyl) amino)ethyl)dithio)ethyl)maleimide (TFPAM-SS 1 ), 2-( 1-{3-dimethylaminopropyl) -indol-3-yl)-3-(indol-3-yl) maleimide (bisindolylmaleimide; GF 109203X), BODIPY~ FL N-(2-aminoethyl)maleimide, N-(7-dimethylamino- 4-methylcoumarin-3-yl)maleimide (DACM), AlexaTM 488 CS maleimide, AlexaTM 594 CS maleimide, sodium saltN-(1-pyrene)maleimide, 2,5-dimethoxystilbene-4'-maleimide, eosin-~-maleimide, fluorescein-~-maleimide, N-(4-(6-dimethylamino- 2-benzofuranyl)phenyl)maleimide, benzophenone-4-maleimide, 4-dimethylaminophenylazophenyl- 4'-maleimide, 1-(2-maleimidylethyl)-4-(5- (4-methoxyphenyl)oxazol-2- yl)pyridinium methanesulfonate, tetramethylrhodamine-5-maleimide, tetramethylrhodamine-6-maleimide, Rhodamine RedTM C2 maleimide, N-(5-aminopentyl)maleimide, N-(2-aminoethyl)maleimide, N-(2-((2-(((4-azido- 2,3,5.6-tetrafluoro)benzoyl) amino)ethyl)dithio)ethyl)maleimide, 2-( 1-(3-dimethylaminopropyl) -indol-3-yl)-3-(indol-3-yl) maleimide, N-(7-dimethylamino- 4-methylcoumarin-3-yl)maleimide (DACM), 11H-Benzo[a]fluorene, Benzo[a]pyrene.

In one embodiment, the hedgehog polypeptide can be derivatived using pyrene.
maleimide, which can be purchased from Molecular Probes (Eugene, Oreg.), e.g., N-(I-pyrene)maleimide or I -pyrenemethyl iodoacetate (PMIA ester).
For those embodiments wherein the hydophobic moiety is a polypeptide, the modified hedgehog polypeptide of this invention can be constructed as a fusion protein, containing the hedgehog polypeptide and the hydrophobic moiety as one contiguous polypeptide chain.
In certain embodiments, the lipophilic moiety is an amphipathic polypeptide, such as magainin, cecropin, attacin, melittin, gramicidin S, alpha-toxin of Staph.
aureus, alamethicin or a synthetic amphipathic polypeptide. Fusogenic coat proteins from viral particles can also be a convenient source of amphipathic sequences for the subject hedgehog proteins Moreover, mutagenesis can be used to create modified hh polypeptides, e.g., for such purposes as enhancing therapeutic or prophylactic efficacy, or stability (e.g., ex vivo shelf life and resistance to proteolytic degradation in vivo). Such modified peptides can be produced, for instance, by amino acid substitution, deletion, or addition. Modified hedgehog polypeptides can also include those with altered post-translational processing relative to a naturally occurring hedgehog protein, e.g., altered glycosylation, cholesterolization, prenylation and the like.
In one embodiment, the hedgehog therapeutic is a polypeptide encodable by a nucleotide sequence that hybridizes under stringent conditions to a hedgehog coding sequence represented in one or more of SEQ ID NOs:I-7. Appropriate stringency conditions which promote DNA hybridization, for example, 6.0 x sodium chloride/sodium citrate (SSC) at about 45°C, followed by a wash of 2.0 x SSC at 50°C, are known to those skilled in the art or can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y.
(1989), 6.3.1-6.3.6. For example, the salt concentration in the wash step can be selected from a low stringency of about 2.0 x SSC at SO°C to a high stringency of about 0.2 x SSC at 50°C. In addition, the temperature in the wash step can be increased from low stringency conditions at room temperature, about 22°C, to high stringency conditions at about 65°C.
As described in the literature, genes for other hedgehog proteins, e.g., from other animals, can be obtained from mRNA or genomic DNA samples using techniques well known in the art. For example, a cDNA encoding a hedgehog protein can be obtained by isolating wo ~no29s pc~ncrs9snzn~

total mlRlVA from a cell, e.g. a mammalian cell, e.g. a human cell, including embryonic cells:
Double stranded cDNAs can then be prepared from the total mRNA, and subsequently inserted into a suitable plasmid or bacteriophage vector using any one of a number of known techniques. The gene encoding a hedgehog protein can also be cloned using established polymerase chain reaction techniques.
Preferred nucleic acids encode a hedgehog polypeptide comprising an amino acid sequence at least 60% homologous or identical, more preferably 70% homologous or identical, and most preferably 80% homologous or identical with an amino acid sequence selected from the group consisting of SEQ ID NOs:B-14. Nucleic acids which encode polypeptides at least about 90%, more preferably at least about 95%, and most preferably at least about 98-99%
homology or identity with an amino acid sequence represented in one of SEQ ID
NOs:B-14 are also within the scope of the invention.
In addition to native hedgehog proteins, hedgehog polypeptides preferred by the present invention are at least 60% homologous or identical, more preferably 70% homologous or identical and most preferably 80% homologous or identical with an amino acid sequence represented by any of SEQ ID NOs:B-14. Polypeptides which are at least 90%, more preferably at least 95%, and most preferably at least about 98-99% homologous or identical with a sequence selected from the group consisting of SEQ ID NOs:8-14 are also within the scope of the invention. The only prerequisite is that the hedgehog polypeptide is capable of modulating the growth of epithelial cells.
The term "recombinant protein" refers to a polypeptide of the present invention which is produced by recombinant DNA techniques, wherein generally, DNA encoding a hedgehog polypeptide is inserted into a suitable expression vector which is in turn used to transform a host cell to produce the heterologous protein. Moreover, the phrase "derived from", with respect to a recombinant hedgehog gene, is meant to include within the meaning of "recombinant protein" those proteins having an amino acid sequence of a native hedgehog protein, or an amino acid sequence similar thereto which is generated by mutations including substitutions and deletions (including truncation) of a naturally occurring form of the protein.
The method of the present invention can also be carried out using variant forms of the naturally occurnng hedgehog polypeptides, e.g., mutational variants.

As is known in the art, hedgehog polypeptides can be produced by standard biological-techniques or by chemical synthesis. For example, a host cell transfected with a nucleic acid vector directing expression of a nucleotide sequence encoding the subject polypeptides can be cultured under appropriate conditions to allow expression of the peptide to occur. The polypeptide hedgehog may be secreted and isolated from a mixture of cells and medium containing the recombinant hedgehog polypeptide. Alternatively, the peptide may be retained cytoplasmically by removing the signal peptide sequence from the recombinant hedgehog gene and the cells harvested, lysed and the protein isolated. A cell culture includes host cells, media and other byproducts. Suitable media for cell culture are well known in the art. The recombinant hedgehog polypeptide can be isolated from cell culture medium, host cells, or both using techniques known in the art for purifying proteins including ion-exchange chromatography, gel filtration chromatography, uitrafiltration, electrophoresis, and immunoaffinity purification with antibodies specific for such peptide. In a preferred embodiment, the recombinant hedgehog polypeptide is a fusion protein containing a domain which facilitates its purification, such as an hedgehoglGST fusion protein.
The host cell may be any prokaryotic or eukaryotic cell.
Recombinant hedgehog genes can be produced by ligating nucleic acid encoding an hedgehog protein, or a portion thereof, into a vector suiiable for expression in either prokaryotic cells, eukaryotic cells, or both. Expression vectors for production of recombinant forms of the subject hedgehog polypeptides include plasmids and other vectors.
For instance, suitable vectors for the expression of a hedgehog polypeptide include plasmids of the types:
pBR322-derived plasmids, pEMBL-derived plasmids, pEX-derived plasmids, pBTac-derived plasmids and pUC-derived plasmids for expression in prokaryotic cells, such as E. coli.
A number of vectors exist for the expression of recombinant proteins in yeast.
For instance, YEP24, YIPS, YEP51, YEP52, pYES2, and YRP17 are cloning and expression vehicles useful in the introduction of genetic constructs into S. cerevisiae (see, for example, Broach et al. (1983) in Experimental Manipulation of Gene Expression, ed. M.
Inouye Academic Press, p. 83, incorporated by reference herein). These vectors can replicate in E. coli due to the presence of the pBR322 ori, and in S. cerevisiae due to the replication determinant of the yeast 2 micron plasmid. In addition, drug resistance markers such as ampicillin can be used. In an illustrative embodiment, an hedgehog polypeptide is produced recombinantly utilizing an expression vector generated by sub-cloning the coding sequence of one of the hedgehog genes represented in SEQ ID NOs:I-7.
The preferred mammalian expression vectors contain both prokaryotic sequences, to facilitate the propagation of the vector in bacteria, and one or more eukaryotic transcription units that are expressed in eukaryotic cells. The pcDNAI/amp, pcDNAI/neo, pRc/CMV, pSV2gpt, pSV2neo, pSV2-dhfr, pTk2, pRSVneo, pMSG, pSVT7, pko-neo and pHyg derived vectors are examples of mammalian expression vectors suitable for transfection of eukaryotic cells. Some of these vectors are modified with sequences from bacterial plasmids, such as pBR322, to facilitate replication and drug resistance selection in both prokaryotic and eukaryotic cells. Alternatively, derivatives of viruses such as the bovine papillomavirus (BPV-1), or Epstein-Barr virus (pHEBo, pREP-derived and p205) can be used for transient expression of proteins in eukaryotic cells. The various methods employed in the preparation of the plasmids and transformation of host organisms are well known in the art.
For other suitable expression systems for both prokaryotic and eukaryotic cells, as well as general recombinant procedures, see Molecular Cloning A Laboratory Manual, .2nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1989) Chapters 16 and 17.
In some instances, it may be desirable to express the recombinant hedgehog polypeptide by the use of a baculovirus expression system. Examples of such baculovirus expression systems include pVL-derived vectors (such as pVL1392, pVL1393 and pVL941), pAcUW-derived vectors (such as pAcUW 1 ), and pBlueBac-derived vectors (such as the 13-gal containing pBlueBac III).
When it is desirable to express only a portion of an hedgehog protein, such as a form lacking a portion of the N-terminus, i.e. a truncation mutant which lacks the signal peptide, it may be necessary to add a start codon (ATG) to the oligonucleotide fragment containing the desired sequence to be expressed. It is well known in the art that a methionine at the N-terminal position can be enzymatically cleaved by the use of the enzyme methionine arninopeptidase (MAP). MAP has been cloned from E. coli (Ben-Bassat et al.
(1987) J. Bacteriol. 169:751-757) and Salmonella typhimurium and its in vitro activity has been wo ~nor~s rcrius9snz~~

demonstrated on recombinant proteins (Miller et al. (1987) PNAS 84:2718-1722).
Therefore;
removal of an N-terminal methionine, if desired, can be achieved either in vivo by expressing hedgehog-derived polypeptides in a host which produces MAP (e.g., E. coli or CM89 or S. cerevisiae), or in vitro by use of purified MAP (e.g., procedure of Miller et al., supra).
Alternatively, the coding sequences for the polypeptide can be incorporated as a part of a fusion gene including a nucleotide sequence encoding a different poiypeptide. It is widely appreciated that fusion proteins can also facilitate the expression of proteins, and accordingly, can be used in the expression of the hedgehog polypeptides of the present invention. For example, hedgehog polypeptides can be generated as glutathione-S-transferase (GST-fusion) proteins. Such GST-fusion proteins can enable easy purification of the hedgehog polypeptide, as for example by the use of glutathione-derivatized matrices (see, for example, Current Protocols in Molecular Biology, eds. Ausubel et al. (N.Y.: John Wiley & Sons, 1991)). In another embodiment, a fusion gene coding for a purification leader sequence, such as a poly-(His)/enterokinase cleavage site sequence, can be used to replace the signal sequence which naturally occurs at the N-terminus of the hedgehog protein (e.g.of the pro-form, in order to permit purification of the poly(His)-hedgehog protein by affinity chromatography using a Ni'-+
metal resin. The purification leader sequence can then be subsequently removed by treatment with enterokinase (e.g., see Hochuli et al. ( 1987) J. Chromatography 411:177;
and Janknecht et al. PNAS 88:8972).
Techniques for making fusion genes are known to those skilled in the art.
Essentia115~, the joining of various DNA fragments coding for different polypeptide sequences is performed in accordance with conventional techniques, employing blunt-ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation. In another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR
amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed to generate a chimeric gene sequence (see, for example, Current Protocols in Molecular Biology. eds. Ausubel et al. John Wiley & Sons: 1992).

Hedgehog polypeptides may also be chemically modified to create hedgehog derivatives by forming covalent or aggregate conjugates with other chemical moieties, such as glycosyl groups, cholesterol, isoprenoids, lipids, phosphate, acetyl groups and the like.
Covalent derivatives of hedgehog proteins can be prepared by linking the chemical moieties to functional groups on amino acid sidechains of the protein or at the N-terminus or at the C-terminus of the polypeptide.
For instance, hedgehog proteins can be generated to include a moiety, other than sequence naturally associated with the protein, that binds a component of the extracellular matrix and enhances localization of the analog to cell surfaces. For example, sequences derived from the fibronectin "type-III repeat", such as a tetrapeptide sequence R-G-D-S
(Pierschbacher et al. (1984) Nature 309:30-3; and Kornblihtt et al. (1985) EMBO 4:1755-9) can be added to the hedgehog polypeptide to support attachment of the chimeric molecule to a cell through binding ECM components (Ruoslahti et al. (1987) Science 238:491-497;
Pierschbacheret al. ( 1987) J. Biol. Chem. 262:17294-8.; Hynes ( 1987) Cell 48:549-54; and Hynes ( 1992) Cell 69:11-25).
In a preferred embodiment, the hedgehog polypeptide is isolated from, or is otherwise substantially free of, other cellular proteins, especially other extracellular or cell surface associated proteins which may normally be associated with the hedgehog polypeptide, unless provided in the form of fusion protein with the hedgehog polypeptide. The term "substantially free of other cellular or extracellular proteins" (also referred to herein as "contaminating proteins") or "substantially pure preparations" or ''purified preparations" are defined as encompassing preparations of hedgehog polypeptides having less than 20% (by dry weight) contaminating protein, and preferably having less than 5%
contaminating protein. By "purified", it is meant that the indicated molecule is present in the substantial absence of other biological macromolecules, such as other proteins. The term "purified" as used herein preferably means at least 80% by dry weight, more preferably in the range of 95-99% by weight, and most preferably at least 99.8% by weight, of biological macromolecules of the same type present (but water, buffers, and other small molecules, especially molecules having a molecular weight of less than 5000, can be present). The term "pure" as used herein preferably has the same numerical limits as "purified" immediately above.

As described above for recombinant polypeptides, isolated hedgehog polypeptides can.
include all or a portion of the amino acid sequences represented in any of SEQ
ID NOs:lO-18 or 20, or a homologous sequence thereto. Preferred fragments of the subject hedgehog proteins correspond to the N-terminal and C-terminal proteolytic fragments of the mature protein. Bioactive fragments of hedgehog polypeptides are described in great detail in PCT
publications WO 95/18856 and WO 96/17924.
With respect to bioctive fragments of hedgehog polypeptide, preferred hedgehog therapeutics include at least 50 (contiguous) amino acid residues of a hedgehog polypeptide, more preferably at least 100 (contiguous), and even more preferably at least 150 (contiguous) residues.
Another preferred hedgehog polypeptide which can be included in the hedgehog therapeutic is an N-terminal fragment of the mature protein having a molecular weight of approximately 19 kDa.
Preferred human hedgehog proteins include N-terminal fragments corresponding approximately to residues 24-197 of SEQ ID No. 15, 28-202 of SEQ ID No. 16, and 23-198 of SEQ ID No. 17. By "corresponding approximately" it is meant that the sequence of interest is at most 20 amino acid residues different in length to the reference sequence, though more preferably at most 5, 10 or 15 amino acid different in length.
As described above for recombinant polypeptides, isolated hedgehog polypeptides can include all or a portion of the amino acid sequences represented in SEQ ID
N0:8, SEQ ID
N0:9, SEQ ID NO:10, SEQ ID NO:11, SEQ ID N0:12, SEQ ID N0:13 or SEQ ID N0:14, or a homologous sequence thereto. Preferred fragments of the subject hedgehog proteins correspond to the N-terminal and C-terminal proteolytic fragments of the mature protein.
Bioactive fragments of hedgehog polypeptides are described in great detail in PCT
publications WO 95/18856 and WO 96/17924.
Still other preferred hedgehog polypeptides includes an amino acid sequence represented by the formula A-B wherein: (i) A represents all or the portion of the amino acid sequence designated by residues 1-168 of SEQ ID N0:21; and B represents at least one amino acid residue of the amino acid sequence designated by residues 169-221 of SEQ
ID N0:21; (ii) A represents all or the portion of the amino acid sequence designated by residues 24-193 of SEQ ID NO:15; and B represents at least one amino acid residue of the amino acid sequence-designated by residues 194-250 of SEQ ID NO:15; (iii) A represents all or the portion of the amino acid sequence designated by residues 25-193 of SEQ ID N0:13; and B
represents at least one amino acid residue of the amino acid sequence designated by residues 194-250 of SEQ ID N0:13; (iv) A represents all or the portion of the amino acid sequence designated by residues 23-193 of SEQ ID NO:I 1; and B represents at least one amino acid residue of the araino acid sequence designated by residues 194-250 of SEQ ID NO:11; (v) A
represents all or the portion of the amino acid sequence designated by residues 28-197 of SEQ ID
N0:12; and B represents at least one amino acid residue of the amino acid sequence designated by residues 198-250 of SEQ ID N0:12; (vi) A represents all or the portion of the amino acid sequence designated by residues 29-197 of SEQ ID N0:16; and B represents at least one amino acid residue of the amino acid sequence designated by residues 198-250 of SEQ ID
N0:16; or (vii) A represents all or the portion of the amino acid sequence designated by residues 23-193 of SEQ ID No. 17, and B represents at least one amino acid residue of the amino acid sequence designated by residues 194-250 of SEQ ID No. 17. In certain preferred embodiments, A and B
together represent a contiguous polypeptide sequence designated sequence, A
represents at least 25, 50, 75, 100, 125 or 150 (contiguous) amino acids of the designated sequence, and B
represents at least S, 10, or 20 (contiguous) amino acid residues of the amino acid sequence designated by corresponding entry in the sequence listing, and A and B
together preferably represent a contiguous sequence corresponding to the sequence listing entry.
Similar fragments from other hedgehog also contemplated, e.g., fragments which correspond to the preferred fragments from the sequence listing entries which are enumerated above. In preferred embodiments, the hedgehog polypeptide includes a C-terminal glycine (or other appropriate residue) which is derivatized with a cholesterol.
Isolated peptidyl portions of hedgehog proteins can be obtained by screening peptides recombinantly produced from the corresponding fragment of the nucleic acid encoding such peptides. In addition, fragments can be chemically synthesized using techniques known in the art such as conventional Merrifield solid phase f Moc or t-Boc chemistry. For example, a hedgehog polypeptide of the present invention may be arbitrarily divided into fragments of desired length with no overlap of the fragments, or preferably divided into overlapping fragments of a desired length. The fragments can be produced (recombinantly or by chemical WO 99n0298 PCT/US98/2Z227 synthesis) and tested to identify those peptidyl fragments which can function as either agonists-or antagonists of a wild-type (e.g., "authentic") hedgehog protein. For example, Roman et al.
(1994) Eur J Biochem 222:65-73 describe the use of competitive-binding assays using short, overlapping synthetic peptides from larger proteins to identify binding domains.
The recombinant hedgehog polypeptides of the present invention also include homologs of the authentic hedgehog proteins, such as versions of these protein which are resistant to proteolytic cleavage, as for example, due to mutations which alter potential cleavage sequences or which inactivate an enzymatic activity associated with the protein.
Hedgehog homologs of the present invention also include proteins which have been post-translationally modified in a manner different than the authentic protein.
Exemplary derivatives of hedgehog proteins include polypeptides which lack N-glycosylation sites (e.g. to produce an unglycosylated protein), which lack sites for cholesterolization, and/or which lack N-terminal and/or C-terminal sequences.
Modification of the structure of the subject hedgehog polypeptides can also be for such purposes as enhancing therapeutic or prophylactic efficacy, or stability (e.g., ex vivo shelf life and resistance to proteolytic degradation in vivo). Such modified peptides, when designed to retain at least one activity of the naturally-occurring form of the protein, are considered functional equivalents of the hedgehog polypeptides described in more detail herein. Such modified peptides can be produced, for instance, by amino acid substitution, deletion, or addition.
It is well known in the art that one could reasonably expect that certain isolated replacements of amino acids, e.g., replacement of an amino acid residue with another related amino acid (i.e. isosteric and/or isoelectric mutations), can be carried out without major effect on the biological activity of the resulting molecule. Conservative replacements are those that take place within a family of amino acids that are related in their side chains. Genetically encoded amino acids are can be divided into four families: (1) acidic =
aspartate, glutamate;
(2) basic = lysine, arginine, histidine; (3) nonpolar = alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan; and (4) uncharged polar =
glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine. Phenylalanine, tryptophan, and tyrosine are sometimes classified jointly as aromatic amino acids. In similar fashion, the amino acid repertoire can be grouped as ( 1 ) acidic = aspartate, glutamate; (2) basic =
lysine, arginine.
histidine, (3) aliphatic = glycine, alanine, valine, leucine, isoleucine, serine, threonine, with serine and threonine optionally be grouped separately as aliphatic-hydroxyl;
(4) aromatic =
phenylalanine, tyrosine, tryptophan; .(5) amide = asparagine, glutamine; and (6) sulfur -containing = cysteine and methionine. (see, for example, Biochemistry, 2nd ed., Ed. by L.
Stryer, WH Freeman and Co.: 1981 ). Whether a change in the amino acid sequence of a peptide results in a functional hedgehog homolog (e.g. functional in the sense that it acts to mimic or antagonize the wild-type form) can be readily determined by assessing the ability of the variant peptide to produce a response in cells in a fashion similar to the wild-type protein, or competitively inhibit such a response. Polypeptides in which more than one replacement has taken place can readily be tested in the same manner.
It is specifically contemplated that the methods of the present invention can be carried using homologs of naturally occurring hedgehog proteins. In one embodiment, the invention contemplates using hedgehog polypeptides generated by combinatorial mutagenesis. Such methods, as are known in the art, are convenient for generating both point and truncation mutants, and can be especially useful for identifying potential variant sequences (e.g.
homologs) that are functional in binding to a receptor for hedgehog proteins.
The purpose of screening such combinatorial libraries is to generate, for example, novel hedgehog homologs which can act as either agonists or antagonist. To illustrate, hedgehog homologs can be engineered by the present method to provide more efficient binding to a cognate receptor, such as patched, yet still retain at least a portion of an activity associated with hedgehog. Thus, combinatorially-derived homologs can be generated to have an increased potency relative to a naturally occurring form of the protein. Likewise, hedgehog homologs can be generated by the present combinatorial approach to act as antagonists, in that they are able to mimic, for example, binding to other extracellular matrix components {such as receptors), yet not induce any biological response, thereby inhibiting the action of authentic hedgehog or hedgehog agonists. Moreover, manipulation of certain domains of hedgehog by the present method can provide domains more suitable for use in fusion proteins, such as one that incorporates portions of other proteins which are derived from the extracellular matrix and/or which bind extracellular matrix components.

WO 99/Z029$ PCT/US9$/22227 To further illustrate the state of the art of combinatorial mutagenesis, it is noted that the .
review article of Gallop et al. (1994) J Med Chem 37:1233 describes the general state of the art of combinatorial libraries as of the earlier 1990's. In particular, Gallop et al state at page 1239 "[s]creeping the analog libraries aids in determining the minimum size of the active sequence and in identifying those residues critical for binding and intolerant of substitution". In addition, the Ladner et al. PCT publication W090/02809, the Goeddel et al.
U.S. Patent 5,223,408, and the Markland et al. PCT publication W092/15679 illustrate specific techniques which one skilled in the art could utilize to generate libraries of hedgehog variants which can be rapidly screened to identify variants/fragments which retained a particular activity of the hedgehog polypeptides. These techniques are exemplary of the art and demonstrate that large libraries of related variapts/truncants can be generated and assayed to isolate particular variants without undue experimentation. Gustin et al. (1993) Virology 193:653, and Bass et al. (1990) Proteins: Structure, Function and Genetics 8:309-314 also describe other exemplary techniques from the art which can be adapted as means for generating mutagenic variants of hedgehog polypeptides.
Indeed, it is plain from the combinatorial mutagenesis art that large scale mutagenesis of hedgehog proteins, without any preconceived ideas of which residues were critical to the biological function, and generate wide arrays of variants having equivalent biological activity.
Indeed, it is the ability of combinatorial techniques to screen billions of different variants by high throughout analysis that removes any requirement of a priori understanding or knowledge of critical residues.
To illsutrate, the amino acid sequences for a population of hedgehog homologs or other related proteins are aligned, preferably to promote the highest homology possible. Such a population of variants can include, for example, hedgehog homologs from one or more species. Amino acids which appear at each position of the aligned sequences are selected to create a degenerate set of combinatorial sequences. In a preferred embodiment, the variegated library of hedgehog variants is generated by combinatorial mutagenesis at the nucleic acid level, and is encoded by a variegated gene library. For instance, a mixture of synthetic oligonucleotides can be enzymatically ligated into gene sequences such that the degenerate set of potential hedgehog sequences are expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g. for phage display) containing the set of hedgehog sequences.
therein.
As illustrated in PCT publication WO 95/18856, to analyze the sequences of a population of variants, the amino acid sequences of interest can be aligned relative to sequence homology. The presence or absence of amino acids from an aligned sequence of a particular variant is relative to a chosen consensus length of a reference sequence, which can be real or artificial.
In an illustrative embodiment, alignment of exons 1, 2 and a portion of exon 3 encoded sequences (e.g. the N-terminal approximately 221 residues of the mature protein) of each of the Shh clones produces a degenerate set of Shh polypeptides represented by the general formula:
C-G-P-G-R-G-X(1)-G-X(2)-R-R-H-P-K-K-L-T-P-L-A-Y-K-Q-F-I-P-N-V-A-E-K-T-L-G-A-S-G-R-Y-E-G-K-I-X(3)-R-N-S-E-R-F-K-E-L-T-P-N-Y-N-P-D-I-I-F-K-D-E-E-N-T-G-A-D-R-L-M-T-Q-R-C-K-D-K-L-N-X(4)-L-A-I-S-V-M-N-X(5)-W-P-G-V-X(6)-L-R-V-T-E-G-W-D-E-D-G-H-H-X(7)-E-E-S-L-H-Y-E-G-R-A-V-D-I-T-T-S-D-R-D-X(8)-S-K-Y-G-X(9)-L-X{10)-R-L-A-V-E-A-G-F-D-W-V-Y-Y-E-S-K-A-H-I-H-C-S-V-K-A-E-N-S-V-A-A-K-S-G-G-C-F-P-G-S-A-X{ 11 )-V-X( 12)-L-X( 13 )-X( 14)-G-G-X( 15)-K-X-( 16)-V-K-D-L-X( 17)-P-G-D-X( 18)-V-L-A-A-D-X(19)-X(20)-G-X(21 )-L-X(22)-X(23)-S-D-F-X(24)-X(25)-F-X(26)-D-R (SEQ ID NO: 21 wherein each of the degenerate positions "X" can be an amino acid which occurs in that position in one of the human, mouse, chicken or zebrafish Shh clones, or, to expand the library, each X can also be selected from amongst amino acid residue which would be conservative substitutions for the amino acids which appear naturally in each of those positions. For instance, Xaa( 1 ) represents Gly, Ala, Val, Leu, Ile, Phe, Tyr or Trp ; Xaa(2) represents Arg, His or Lys; Xaa(3) represents Gly, Ala, Val, Leu, Ile, Ser or Thr; Xaa(4) represents Gly, Ala, Val, Leu, Ile, Ser or Thr; Xaa(5) represents Lys, Arg, His, Asn or Gln;
Xaa(6) represents Lys, Arg or His; Xaa(7) represents Ser, Thr, Tyr, Trp or Phe; Xaa(8) represents Lys, Arg or His; Xaa(9) represents Met, Cys, Ser or Thr; Xaa(10) represents Gly, Ala, Val, Leu, Ile, Ser or Thr; Xaa{11) represents Leu, Val, Met, Thr or Ser;
Xaa(12) represents His, Phe, Tyr, Ser, Thr, Met or Cys; Xaa(13) represents Gln, Asn, Glu, or Asp;

WO 99/20298 PCT/ItS98I22Z27 Xaa{14) represents His, Phe, Tyr, Thr, Gln, Asn, Glu or Asp; Xaa(15) represents Gln, Asn, .
Glu, Asp, Thr, Ser, Met or Cys; Xaa(16) represents Ala, Gly, Cys, Leu, Val or Met; Xaa(17) represents Arg, Lys, Met, Ile, Asn, Asp, Glu, Gln, Ser, Thr or Cys; Xaa( 18) represents Arg, Lys, Met. or Ile; Xaa( 19) represents Ala, Gly, Cys, Asp, Glu, Gln, Asn, Ser, Thr or Met;
Xaa(20) represents Ala, Gly, Cys, Asp, Asn, Glu or GIn; Xaa(21 ) represents Arg, Lys, Met, Ile, Asn, Asp, Glu or Gln; Xaa(22) represent Leu, Val, Met or Ile; Xaa(23) represents Phe, Tyr, Thr, His or Trp; Xaa(24) represents Ile, Val, Leu or Met; .Xaa(25) represents Met, Cys, Ile, Leu, Val, Thr or Ser; Xaa(26) represents Leu, Val, Met, Thr or Ser. In an even more expansive library, each X can be selected from any amino acid.
In similar fashion, alignment of each of the human, mouse, chicken and zebrafish hedgehog clones, can provide a degenerate polypeptide sequence represented by the general formula:
C-G-P-G-R-G-X( 1 )-X(2)-X(3)-R-R-X(4)-X(5)-X(6)-P-K-X(7)-L-X(8)-P-L-X(9)-Y-K-Q-F-X( 10)-P-X( 11 )-X( 12)-X( 13 )-E-X( 14)-T-L-G-A-S-G-X{ 15)-X( 16)-E-G-X{ 17)-X( 18)-X( 19)-R-X(20)-S-E-R-F-X(2 I )-X(22)-L-T-P-N-Y-N-P-D-I-I-F-K-D-E-E-N-X(23)-G-A-D-R-L-M-T-X(24)-R-C-K-X(25)-X(26)-X(27)-N-X(28)-L-A-I-S-V-M-N-X(29)-W-P-G-V-X(30)-L-R-V-T-E-G-X(31 )-D-E-D-G-H-H-X(32)-X(33)-X(34)-S-L-H-Y-E-G-R-A-X(35)-D-I-T-T-S-D-R-.
D-X(36)-X(37)-K-Y-G-X(38)-L-X(39)-R-L-A-V-E-A-G-F-D-W-V-Y-Y-E-S-X(40)-X(41)-H-X(42)-H-X(43)-S-V-K-X{44)-X(45) (SEQ IDNo:22) wherein, as above, each of the degenerate positions "X" can be an amino acid which occurs in a corresponding position in one of the wild-type clones, and may also include amino acid residue which would be conservative substitutions, or each X can be any amino acid residue.
In an exemplary embodiment, Xaa( 1 ) represents Gly, Ala, Val, Leu, Ile, Pro, Phe or Tyr;
Xaa(2) represents Gly, Ala, Val, Leu or Ile; Xaa(3) represents Gly, Ala, Val, Leu, Ile, Lys, His or Arg; Xaa(4) represents Lys, Arg or His; Xaa(5) represents Phe, Trp, Tyr or an amino acid gap; Xaa(6) represents Gly, Ala, Val, Leu, Ile or an amino acid gap; Xaa(7) represents Asn, Gln, His, Arg or Lys; Xaa(8) represents Gly, Ala, Val, Leu, Ile, Ser or Thr;
Xaa{9) represents Gly, Ala, Val, Leu, Ile, Ser or Thr; Xaa(10) represents Gly, Ala, Val, Leu, Ile, Ser or Thr;
Xaa(11) represents Ser, Thr, Gln or Asn; Xaa(12) represents Met, Cys, Gly, Ala, Val, Leu, Ile, Ser or Thr; Xaa(13) represents Gly, Ala, Val, Leu, Ile or Pro; Xaa(14) represents Arg, His or Lys; Xaa(15) represents Gly, Ala, Val, Leu, Ile, Pro, Arg, His or Lys; Xaa(16) represents Gly, Ala, Val, Leu, Ile, Phe or Tyr; Xaa(17) represents Arg, His or Lys; Xaa(18) represents Gly, .
Ala, Val, Leu, Ile, Ser or Thr; Xaa(19) represents Thr or Ser; Xaa(20) represents Gly, Ala, Val, Leu, Ile, Asn or Gln; Xaa(21 ) represents Arg, His or Lys; Xaa(22) represents Asp or Glu;
Xaa(23) represents Ser or Thr; Xaa(24) represents Glu, Asp, Gln or Asn;
Xaa(25) represents Glu or Asp; Xaa(26) represents Arg, His or Lys; Xaa(27) represents Gly, Ala, Val, Leu or Ile;
Xaa(28) represents Gly, Ala, Val, Leu, Ile, Thr or Ser; Xaa(29) represents Met, Cys, Gln, Asn, Arg, Lys or His; Xaa(30) represents Arg, His or Lys; Xaa(31 ) represents Trp, Phe, Tyr, Arg, His or Lys; Xaa(32) represents Gly, Ala, Val, Leu, Ile, Ser, Thr, Tyr or Phe;
Xaa(33) represents Gln, Asn, Asp or Glu; Xaa(34) represents Asp or Glu; Xaa(35) represents Gly, Ala, Val, Leu, or Ile; Xaa(36) represents Arg, His or Lys; Xaa(37) represents Asn, Gln, Thr or Ser; Xaa(38) represents Gly, Ala, Val, Leu, Ile, Ser, Thr, Met or Cys; Xaa(39) represents Gly, Ala, Val, Leu, Ile, Thr or Ser; Xaa(40) represents Arg, His or Lys; Xaa(41 ) represents Asn, Gln, Gly, Ala, Val, Leu or Ile; Xaa(42) represents Gly, Ala, Val, Leu or Ile; Xaa(43) represents Gly, Ala, Val, Leu, Ile, Ser, Thr or Cys; Xaa(44) represents Gly, Ala, Val, Leu, Ile, Thr or Ser; and Xaa(45) represents Asp or Glu.
There are many ways by which the library of potential hedgehog homologs can be generated from a degenerate oligonucleotide sequence. Chemical synthesis of a degenerate gene sequence can be carried out in an automatic DNA synthesizer, and the synthetic genes then ligated into an appropriate expression vector. The purpose of a degenerate set of genes is to provide, in one mixture, all of the sequences encoding the desired set of potential hedgehog sequences. The synthesis of degenerate oligonucleotides is well known in the art (see for example, Narang, SA (1983) Tetrahedron 39:3; Itakura et al. (1981) Recombinant DNA, Proc 3rd Cleveland Sympos. Macromolecules, ed. AG Walton, Amsterdam: Elsevier pp273-289;
Itakura et al. ( 1984) Annu. Rev. Biochem. 53:323; Itakura et al. ( 1984) Science 198:1056; Ike et al. (1983) Nucleic Acid Res. 11:477. Such techniques have been employed in the directed evolution of other proteins (see, for example, Scott et al. (1990) Science 249:386-390; Roberts et al. ( 1992) PNAS 89:2429-2433; Devlin et al. { 1990) Science 249: 404-406;
Cwirla et al.
(1990) PNAS 87: 6378-.6382; as well as U.S. Patents Nos. 5,223,409, 5,198,346, and 5,096,815).

A wide range of techniques are known in the art for screening gene products of combinatorial libraries made by point mutations, and for screening cDNA
libraries for gene products having a certain property. Such techniques will be generally adaptable for rapid screening of the gene libraries generated by the combinatorial mutagenesis of hedgehog homologs. The most widely used techniques for screening large gene libraries typically comprises cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates relatively easy isolation of the vector encoding the gene whose product was detected. Each of the illustrative assays described below are amenable to high through-put analysis as necessary to screen large numbers of degenerate hedgehog sequences created by combinatorial mutagenesis techniques.
In one embodiment, the combinatorial library is designed to be secreted (e.g.
the polypeptides of the library all include a signal sequence but no transmembrane or cytoplasmic domains), and is used to transfect a eukaryotic cell that can be co-cultured with epithelial stem cells. A functional hedgehog protein secreted by the cells expressing the combinatorial library will diffuse to neighboring epithelial cells and induce a particular biological. response, such as proliferation. The pattern of detection of proliferation will resemble a gradient function, and will allow the isolation (generally after several repetitive rounds of selection) of cells producing hedgehog homologs active as proliferative agents with respect to epithelial cells.
Likewise, hedgehog antagonists can be selected in similar fashion by the ability of the cell producing a functional antagonist to protect neighboring cells (e.g., to inhibit proliferation) from the effect of wild-type hedgehog added to the culture media.
To illustrate, target epithelial cells are cultured in 24-well microtitre plates. Other eukaryotic cells are transfected with the combinatorial hedgehog gene library and cultured in cell culture inserts (e.g. Collaborative Biomedical Products, Catalog #40446) that are able to fit into the wells of the microtitre plate. The cell culture inserts are placed in the wells such that recombinant hedgehog homologs secreted by the cells in the insert can diffuse through the porous bottom of the insert and contact the target cells in the microtitre plate wells. After a period of time sufficient for functional forms of a hedgehog protein to produce a measurable response in the target cells, such as proliferation, the inserts are removed and the effect of the variant hedgehog proteins on the target cells determined. Cells from the inserts corresponding to wells which score positive for activity can be split and re-cultured on several inserts, the process being repeated until the active clones are identified.
In yet another screening assay, the candidate hedgehog gene products are displayed on the surface of a cell or viral particle, and the ability of particular cells or viral particles to associate with a hedgehog-binding moiety (such as the patched protein or other hedgehog receptor) via this gene product is detected in a "panning assay". Such panning steps can be carried out on cells cultured from embryos. For instance, the gene library can be cloned into the gene for a surface membrane protein of a bacterial cell, and the resulting fusion protein detected by panning (Ladner et al., WO 88/06630; Fuchs et al. (1991) BiolTechnolo~r 9:1370-1371; and Goward et al. (1992) TIBS 18:136-140). In a similar fashion, fluorescently labeled molecules which bind hedgehog can be used to score for potentially functional hedgehog homologs. Cells can be visually inspected and separated under a fluorescence microscope, or, where the morphology of the cell permits, separated by a fluorescence-activated cell sorter.
In an alternate embodiment, the gene library is expressed as a fusion protein on the surface of a viral particle. For instance, in the filamentous phage system, foreign peptide sequences can be expressed on the surface of infectious phage, thereby conferring two significant benefits. First, since these phage can be applied to affinity matrices at very high concentrations, large number of phage can be screened at one time. Second, since each infectious phage displays the combinatorial gene product on its surface, if a particular phage is recovered from an affinity matrix in low yield, the phage can be amplified by another round of infection. The group of almost identical E.coli filamentous phages M13, fd, and fl are most often used in phage display libraries, as either of the phage gIII or gVIII
coat proteins can be used to generate fusion proteins without disrupting the ultimate packaging of the viral particle (Ladner et al. PCT publication WO 90/02909; Garrard et al., PCT publication WO
92/09690;
Marks et al. (1992) J. Biol. Chem. 267:16007-16010; Griffths et al. (1993) EMBO J 12:725-734; Clackson et al. (199I) Nature 352:624-628; and Barbas et al. (1992) PNAS
89:4457-4461 ).
In an illustrative embodiment, the recombinant phage antibody system (RPAS, Pharamacia Catalog number 27-9400-O1) can be easily modified for use in expressing and screening hedgehog combinatorial libraries. For instance, the pCANTAB 5 phagemid of the RPAS kit contains the gene which encodes the phage gIII coat protein. The hedgehog combinatorial gene library can be cloned into the phagemid adjacent to the gIII signal sequence such that it will be expressed as a gIII fusion protein. After ligation, the phagemid is used to transform competent E. coli TG1 cells. Transformed cells are subsequently infected with M13K07 helper phage to rescue the phagemid and its candidate hedgehog gene insert. The resulting recombinant phage contain phagemid DNA encoding a specific candidate hedgehog, and display one or more copies of the corresponding fusion coat protein. The phage-displayed candidate hedgehog proteins which are capable of binding an hedgehog receptor are selected or enriched by panning. For instance, the phage library can be applied to cells which express the patched protein and unbound phage washed away ftom the cells. The bound phage is then isolated, and if the recombinant phage express at least one copy of the wild type gIII coat protein, they will retain their ability to infect E. coli. Thus, successive rounds of reinfection of E. coli, and panning will greatly enrich for hedgehog homologs, which can then be screened for further biological activities in order to differentiate agonists and antagonists.
Combinatorial mutagenesis has a potential to generate very large libraries of mutant proteins, e.g., in the order of 1026 molecules. Combinatorial libraries of this size may be technically challenging to screen even with high throughput screening assays such as phage display. To overcome this problem, a new technique has been developed recently, recursive ensemble mutagenesis (REM), which allows one to avoid the very high proportion of non-functional proteins in a random library and simply enhances the frequency of functional proteins, thus decreasing the complexity required to achieve a useful sampling of sequence space. REM is an algorithm which enhances the frequency of functional mutants in a library when an appropriate selection or screening method is employed (Arkin and Yourvan, 1992, PNAS USA 89:7811-7815; Yourvan et al., 1992, Parallel Problem Solving from Nature, 2., In Maenner and Manderick, eds., Elsevir Publishing Co., Amsterdam, pp. 401-410;
Delgrave et al., 1993, Protein Engineering 6(3):327-331 ).
The invention also provides for reduction of the hedgehog protein to generate mimetics, e.g. peptide or non-peptide agents, which are able to disrupt binding of a hedgehog polypeptide of the present invention with an hedgehog receptor. Thus, such mutagenic techniques as described above are also useful to map the determinants of the hedgehog.
proteins which participate in protein-protein interactions involved in, for example, binding of the subject hedgehog polypeptide to other extracellular matrix components. To illustrate, the critical residues of a subject hedgehog polypeptide which are involved in molecular recognition of an hedgehog receptor such as patched can be determined and used to generate hedgehog-derived peptidomimetics which competitively inhibit binding of the authentic hedgehog protein with that moiety. By employing, for example, scanning mutagenesis to map the amino acid residues of each of the subject hedgehog proteins which are involved in binding other extracellular proteins, peptidomimetic compounds can be generated which mimic those residues of the hedgehog protein which facilitate the interaction. Such mimetics may then be used to interfere with the normal function of a hedgehog protein. For instance, non-hydrolyzable peptide analogs of such residues can be generated using benzodiazepine (e.g., see Freidinger et al. in Peptides: Chemistry and Biology, G.R. Marshall ed., ESCOM
Publisher:
Leiden, Netherlands, 1988), azepine (e.g., see Huffman et al. in Peptides:
Chemistry and Biology, G.R. Marshall ed., ESCOM Publisher: Leiden, Netherlands, 1988), substituted gams lactam rings {Garvey et al. in Peptides: Chemistry and Biology, G.R. Marshall ed., ESCOM
Publisher: Leiden, Netherlands, 1988), keto-methylene pseudopeptides (Ewenson et al. ( 1986) J Med Chem 29:295; and Ewenson et al. in Peptides: Structure and Function (Proceedings of the 9th American Peptide Symposium) Pierce Chemical Co. Rockland, IL, 1985), (3-turn dipeptide cores (Nagai et al. ( 1985) Tetrahedron Lett 26:647; and Sato et al.
( 1986) J Chem Soc Perkin Trans 1:1231), and ~i-aminoalcohols (Gordon et al. (1985) Biochem Biophys Res Communl26:419; and Dann et al. (1986) Biochem Biophys Res Commun 134:71).
Recombinantly produced forms of the hedgehog proteins can be produced using, e.g, expression vectors containing a nucleic acid encoding a hedgehog polypeptide, operably linked to at least one transcriptional regulatory sequence. Operably linked is intended to mean that the nucleotide sequence is linked to a regulatory sequence in a manner which allows expression of the nucleotide sequence. Regulatory sequences are art-recognized and are selected to direct expression of a hedgehog polypeptide. Accordingly, the term transcriptional regulatory sequence includes promoters, enhancers and other expression control elements.
Such regulatory sequences are described in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA ( 1990). For instance, any of a wide wo 9snoz9s rc~rrnrs9snzzz~

variety of expression control sequences, sequences that control the expression of a DNA.
sequence when operatively linked to it, may be used in these vectors to express DNA
sequences encoding hedgehog polypeptide. Such useful expression control sequences, include, for example, a viral LTR, such as the LTR of the Moloney marine leukemia virus, the early and late promoters of SV40, adenovirus or cytomegalovirus immediate early promoter, the lac system, the trp system, the TAC or TRC system, T7 promoter whose expression is directed by T7 RNA polymerise, the major operator and promoter regions of phage ~, , the control regions for fd coat protein, the promoter for 3-phosphoglycerate kinase or other glycolytic enzymes, the promoters of acid phosphatase, e.g., PhoS, the promoters of the yeast I0 a-mating factors, the polyhedron promoter of the baculovirus system and other sequences known to control the expression of genes of prokaryotic or eukaryotic cells or their viruses, and various combinations thereof. It should be understood that the design of the expression vector may depend on such factors as the choice of the host cell to be transformed and/or the type of protein desired to be expressed. Moreover, the vector's copy number, the ability to control that copy number and the expression of any other proteins encoded by the vector, such as antibiotic markers, should also be considered.
In addition to providing a ready source of hedgehog polypeptides for purification, the gene constructs of the present invention can also be used as a part of a gene therapy protocol to deliver nucleic acids encoding either an agonistic or antagonistic form of a hedgehog polypeptide. Thus, another aspect of the invention features expression vectors for in vivo transfection of a hedgehog polypeptide in particular cell types so as cause ectopic expression of a hedgehog polypeptide in an epithelial tissue.
Formulations of such expression constructs may be administered in any biologically effective carrier, e.g. any formulation or composition capable of effectively delivering the recombinant gene to cells in vivo. Approaches include insertion of the hedgehog coding sequence in viral vectors including recombinant retroviruses, adenovirus, adeno-associated virus, and herpes simplex virus-1, or recombinant bacterial or eukaryotic plasmids. Viral vectors transfect cells directly; plasmid DNA can be delivered with the help of, for example, cationic liposomes (lipofectin) or derivatized (e.g. antibody conjugated), polylysine conjugates, gramicidin S, artificial viral envelopes or other such intracellular carriers, as well as direct injection of the gene construct or CaP04 precipitation carried out in vivo. It will be-appreciated that because transduction of appropriate target cells represents the critical first step in gene therapy, choice of the particular gene delivery system will depend on such factors as the phenotype of the intended target and the route of administration, e.g.
locally or systemically. Furthermore, it will be recognized that the particular gene construct provided for in vivo transduction of hedgehog expression are also useful for in vitro transduction of cells, such as for use in the ex vivo tissue culture systems described below.
A preferred approach for in vivo introduction of nucleic acid into a cell is by use of a viral vector containing nucleic acid, e.g. a cDNA, encoding the particular form of the hedgehog polypeptide desired. Infection of cells with a viral vector has the advantage that a large proportion of the targeted cells can receive the nucleic acid.
Additionally, molecules encoded within the viral vector, e.g., by a cDNA contained in the viral vector, are expressed efficiently in cells which have taken up viral vector nucleic acid.
Retrovirus vectors and adeno-associated virus vectors are generally understood to be the recombinant gene delivery system of choice for the transfer of exogenous genes in vivo, particularly into humans. These vectors provide efficient delivery of genes into cells, and the transferred nucleic acids are stably integrated into the chromosomal DNA of the host. A major prerequisite for the use of retroviruses is to ensure the safety of their use, particularly with regard to the possibility of the spread of wild-type virus in the cell population. The development of specialized cell lines (termed "packaging cells") which produce only replication-defective retroviruses has increased the utility of retroviruses for gene therapy, and defective retroviruses are well characterized for use in gene transfer for gene therapy purposes (for a review see Miller, A.D. ( 1990) Blood 76:271 ). Thus, recombinant retrovirus can be constructed in which part of the retroviral coding sequence (gag, pol, env) has been replaced by nucleic acid encoding a hedgehog polypeptide and renders the retrovirus replication defective. The replication defective retrovirus is then packaged into virions which can be used to infect a target cell through the use of a helper virus by standard techniques. Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current Protocols in Molecular Bioloav, Ausubel, F.M. et al.
(eds.) Greene Publishing Associates, (1989), Sections 9.10-9.14 and other standard laboratory manuals.

WO 99/20298 pCT/US98/22227 Examples of suitable retroviruses include pLJ, pZIP, pWE and pEM which are well known to.
those skilled in the art. Examples of suitable packaging virus lines for preparing both ecotropic and amphotropic retroviral systems include Crip, Cre, 2 and Am.
Retroviruses have been used to introduce a variety of genes into many different cell types, including epithelial S cells, in vitro and/or in vivo (see for example Eglitis, et al. (1985) Science 230:1395-1398;
Danos and Mulligan (1988) Proc. Natl. Acad. Sci. USA 85:6460-6464; Wilson et al. (1988) Proc. Natl. Acad. Sci. USA 85:3014-3018; Armentano et al. (1990) Proc. Natl.
Acad. Sci. USA
87:6141-6145; Huber et al. (1991) Proc. Natl. Acad. Sci. USA 88:8039-8043;
Ferry et al.
(1991) Proc. Natl. Acad. Sci. USA 88:8377-8381; Chowdhury et al. {1991) Science 254:1802-1805; van Beusechem et al. (1992) Proc. Natl. Acad. Sci. USA 89:7640-7644; Kay et al.
( 1992) Human Gene Therapy 3:641-647; Dai et al. ( 1992) Proc. Natl. Acad.
Sci. USA
89:10892-10895; Hwu et al. (1993) J. Immunol. 150:4104-4115; U.S. Patent No.
4,868,116;
U.S. Patent No. 4,980,286; PCT Application WO 89/07136; PCT Application WO
89/02468;
PCT Application WO 89/05345; and PCT Application WO 92/07573).
Furthermore, it has been shown that it is possible to limit the infection spectrum of retroviruses and consequently of retroviral-based vectors, by modifying the viral packaging proteins on the surface of the viral particle (see, for example PCT
publications W093/25234 and W094/06920). For instance, strategies for the modification of the infection spectrum of retroviral vectors include: coupling antibodies specific for cell surface antigens to the viral env 20. protein (Roux et al. ( 1989) PNAS 86:9079-9083; Julan et al. ( 1992} J.
Gen Yirol 73:3251-3255; and Goud et al. (1983) Virology 163:251-254); or coupling cell surface receptor ligands to the viral env proteins (Veda et al. (1991 ) J Biol Chem 266:14143-14146).
Coupling can be in the form of the chemical cross-linking with a protein or other variety (e.g. lactose to convert the env protein to an asialoglycoprotein), as well as by generating fusion proteins (e.g. single-chain antibody/env fusion proteins). This technique, while useful to limit or otherwise direct the infection to certain tissue types, can also be used to convert an ecotropic vector in to an amphotropic vector.
Moreover, use of retroviral gene delivery can be further enhanced by the use of tissue or cell-specific transcriptional regulatory sequences which control expression of the hedgehog gene of the retroviral vector.

wo ~no29s rcricrs9snz~~
-s8-Another viral gene delivery system useful in the present method utilizes adenovirus- .
derived vectors. The genome of an adenovirus can be manipulated such that it encodes and expresses a gene product of interest but is inactivated in terms of its ability to replicate in a normal lytic viral life cycle. See for example Berkner et al. (1988) BioTechnigues 6:616;
s Rosenfeld et al. (1991) Science 2s2:431-434; and Rosenfeld et al. (1992) Cell 68:143-lss.
Suitable adenoviral vectors derived from the adenovirus strain Ad type s d1324 or other strains of adenovirus (e.g., Ad2, Ad3, Ad7 etc.) are well known to those skilled in the art.
Recombinant adenoviruses can be advantageous in certain circumstances in that they can be used to infect a wide variety of cell types, including epithelial cells (Rosenfeld et al. (1992) cited supra). Furthermore, the virus particle is relatively stable and amenable to purification and concentration, and as above, can be modified so as to affect the spectrum of infectivity.
Additionally, introduced adenoviral DNA (and foreign DNA contained therein) is not integrated into the genome of a host cell but remains episomal, thereby avoiding potential problems that can occur as a result of insertional mutagenesis in situations where introduced 1 s DNA becomes integrated into the host genome (e.g., retroviral DNA).
Moreover, the carrying capacity of the adenoviral genome for foreign DNA is large (up to 8 kilobases) relative to other gene delivery vectors (Berkner et al. cited supra; Haj-Ahmand and Graham ( 1986) J.
Virol. s7:267). Most replication-defective adenoviral vectors currently in use and therefore favored by the present invention are deleted for all or parts of the viral E1 and E3 genes but retain as much as 80% of the adenoviral genetic material (see, e.g., Jones et al. (1979) Cell 16:683; Berkner et al., supra; and Graham et al. in Methods in Molecular Biology, E.J.
Murray, Ed. (Humana, Clifton, NJ, 1991) vol. 7. pp. 109-127). Expression of the inserted hedgehog gene can be under control of, for example, the E 1 A promoter, the major late promoter (MLP) and associated leader sequences, the E3 promoter, or exogenously added 2s promoter sequences.
In addition to viral transfer methods, such as those illustrated above, non-viral methods can also be employed to cause expression of a hedgehog polypeptide in the tissue of an animal.
Most nonviral methods of gene transfer rely on normal mechanisms used by mammalian cells for the uptake and intracellular transport of macromolecules. In preferred embodiments, non-viral gene delivery systems of the present invention rely on endocytic pathways for the uptake of the hedgehog polypeptide gene by the targeted cell. Exemplary gene delivery systems of this type include liposomal derived systems, poly-lysine conjugates, and artificial viral-envelopes.
In clinical settings, the gene delivery systems for the therapeutic hedgehog gene can be introduced into a patient by any of a number of methods, each of which is familiar in the art.
For instance, a pharmaceutical preparation of the gene delivery system can be introduced systemically, e.g. by intravenous injection, and specific transduction of the protein in the target cells occurs predominantly from specificity of transfection provided by the gene delivery vehicle, cell-type or tissue-type expression due to the transcriptional regulatory sequences controlling expression of the receptor gene, or a combination thereof. In other embodiments, initial delivery of the recombinant gene is more limited with introduction into the animal being quite localized. For example, the gene delivery vehicle can be introduced by catheter (see U.S.
Patent 5,328,470) or by stereotactic injection (e.g. Chen et al. (1994) PNAS
91: 3054-3057). A
hedgehog expression construct can be delivered in a gene therapy construct to dermal cells by, e.g., electroporation using techniques described, for example, by Dev et al.
(( 1994) Cancer Treat Rev 20:105-115).
The pharmaceutical preparation of the gene therapy construct can consist essentially of the gene delivery system in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery system can be produced intact from recombinant cells, e.g. retroviral vectors, the pharmaceutical preparation can comprise one or more cells which produce the gene delivery system.
In yet another embodiment, the hedgehog or ptc therapeutic can be a "gene activation"
construct which, by homologous recombination with a genomic DNA, alters the transcriptional regulatory sequences of an endogenous gene. For instance, the gene activation construct can replace the endogenous promoter of a hedgehog gene with a heterologous promoter, e.g., one which causes consitutive expression of the hedgehog gene or which causes inducible expression of the gene under conditions different from the normal expression pattern of the gene. Other genes in the patched signaling pathway can be similarly targeted.
A vareity of different formats for the gene activation constructs are available. See, for example, the Transkaryotic Therapies, Inc PCT publications W093/09222, W095/31560, W096/29411, -W095/31560 and W094/12650.
In preferred embodiments, the nucleotide sequence used as the gene activation construct can be comprised of ( 1 ) DNA from some portion of the endogenous hedgehog gene (exon sequence, intron sequence, promoter sequences, etc.) which direct recombination and (2) heterologous transcriptional regulatory sequences) which is to be operably linked to the coding sequence for the genomic hedgehog gene upon recombination of the gene activation conshvct. For use in generating cultures of hedgehog producing cells, the construct may further include a reporter gene to detect the presence of the knockout construct in the cell.
The gene activation construct is inserted into a cell, and integrates with the genomic DNA of the cell in such a position so as to provide the heterologous regulatory sequences in operative association with the native hedgehog gene. Such insertion occurs by homologous recombination, i.e., recombination regions of the activation construct that are homologous to the endogenous hedgehog gene sequence hybridize to the genomic DNA and recombine with the genomic sequences so that the construct is incorporated into the corresponding position of the genomic DNA.
The terms "recombination region" or "targeting sequence" refer to a segment (i.e., a portion) of a gene activation construct having a sequence that is substantially identical to or substantially complementary to a genomic gene sequence, e.g., including 5' flanking sequences of the genomic gene, and can facilitate homologous recombination between the genomic sequence and the targeting transgene construct.
As used herein, the term "replacement region" refers to a portion of a activation construct which becomes integrated into an endogenous chromosomal location following homologous recombination between a recombination region and a genomic sequence.
The heterologous regulatory sequences, e.g., which are provided in the replacement region, can include one or more of a variety elements, including: promoters (such as constitutive or inducible promoters), enhancers, negative regualtory elements, locus control regions, transcription factor binding sites, or combinations thereof.
Promoters/enhancers which may be used to control the expression of the targeted gene in vivo include, but are not limited to, the cytomegalovirus (CMV) promoter/enhancer (Karasuyama et al., 1989, J. Exp.

WO 99n0298 PGT/US98I22227 Med., 169:13), the human (3-actin promoter (Gunning et al. (1987) PNAS 84:4831-4835), the glucocorticoid-inducible promoter present in the mouse mammary tumor virus long terminal repeat (MMTV LTR) (Klessig et al. (1984) Mol. Cell Biol. 4:1354-1362), the long terminal repeat sequences of Moloney marine leukemia virus (MuLV LTR) (Weiss et al.
(1985) RNA
Tumor Viruses, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York);
the SV40 early or late region promoter (Bernoist et al. (1981) Nature 290:304-310;
Templeton et al.
(1984) Mol. Cell Biol., 4:817; and Sprague et al. (1983) J. Virol., 45:773), the promoter contained in the 3' long terminal repeat of Rous sarcoma virus (RSV) (Yamamoto et al., 1980, Cell, 22:787-797), the herpes simplex virus {HSV) thymidine kinase promoter/enhancer (Wagner et al. ( 1981 ) PNAS 82:3567-71 ), and the herpes simplex virus LAT
promoter (Wolfe et al. {1992) Nature Genetics, 1:379-384).
In an exemplary embodiment, portions of the 5' flanking region of the human Shh gene are amplified using primers which add restriction sites, to generate the following fragments 5'-gcgcgcttcgaaGCGAGGCAGCCAGCGAGGGAGAGAGCGAGCGGGCGAGCCGGAGC-GAGGAAatcgatgcgcgc (primer 1) 5'-gcgcgcagatctGGGAAAGCGCAAGAGAGAGCGCACACGCACACACCCGCCGCGCG-CACTCGggatccgcgcgc (primer 2) As illustrated, primer 1 includes a 5' non-coding region of the human Shh gene and is flanked by an AsuII and CIaI restriction sites. Primer 2 includes a portion of the 5' non-coding region immediately 3' to that present in primer 1. The hedgehog gene sequence is flanked by XhoII
and BamHI restriction sites. The purified amplimers are cut with each of the enzymes as appropriate.
The vector pCDNAI.I (Invitrogen) includes a CMV promoter. The plasmid is cut with with AsuII, which cleaves just 3' to the CMV promoter sequence. The AsuII/CIaI
fragment of primer 1 is ligated to the AsuII cleavage site of the pcDNA vector. The CIaI/AsuII ligation destroys the AsuII site at the 3' end of a properly inserted primer 1.

The vector is then cut with BamHI, and an XhoIIBamHI fragment of primer 2 is -ligated to the BamHI cleavage site. As above, the BamHI/XhoII ligation destroys .the BamHI
site at the 5' end of a properly inserted primer 2.
Individual colonies are selected, cut with AsuII and BamHI, and the size of the AsuII/BamHI fragment determined. Colonies in which both the primer 1 and primer 2 sequences are correctly inserted are further amplified, an cut with AsuII and BamHI to produce the gene activation construct cgaagcgaggcagccagcgagggagagagcgagcgggcgagccggagcgaggaaATCGAAGGT
TCGAATCCTTCCCCCACCACCATCACTTTCAAAAGTCCGAAAGAATCTGCTCCCTGCTTGTGT
GTTGGAGGTCGCTGAGTAGTGCGCGAGTAAAATTTAAGCTACAACAAGGCAAGGCTTGACCGA
CAATTGCATGAAGAATCTGCTTAGGGTTAGGCGTTTTGCGCTGCTTCGCGATGTACGGGCCAG
ATATACGCGTTGACATTGATTATTGACTAGTTATTAATAGTAATCAATTACGGGGTCATTAGT
TCATAGCCCATATATGGAGTTCCGCGTTACATAACTTACGGTAAATGGCCCGCCTGGCTGACC
GCCCAACGACCCCCGCCCATTGACGTCAATAATGACGTATGTTCCCATAGTAACGCCAATAGG
GACTTTCCATTGACGTCAATGGGTGGACTATTTACGGTAAACTGCCCACTTGGCAGTACATCA
AGTGTATCATATGCCAAGTACGCCCCCTATTGACGTCAATGACGGTAAATGGCCCGCCTGGCA
TTATGCCCAGTACATGACCTTATGGGACTTTCCTACTTGGCAGTACATCTACGTATTAGTCAT
CGCTATTACCATGGTGATGCGGTTTTGGCAGTACATCAATGGGCGTGGATAGCGGTTTGACTC
ACGGGGATTTCCAAGTCTCCACCCCATTGACGTCAATGGGAGTTTGTTTTGGCACCAAAATCA
ACGGGACTTTCCAAAATGTCGTAACAACTCCGCCCCATTGACGCAAATGGGCGGTAGGCGTGT
ACGGTGGGAGGTCTATATAAGCAGAGCTCTCTGGCTAACTAGAGAACCCACTGCTTACTGGCT
TATCGAAATTAATACGACTCACTATAGGGAGACCCAAGCTTGGTACCGAGCTCGGATCgatct gggaaagcgcaagagagagcgcacacgcacacacccgccgcgcgcactcgg In this construct, the flanking primer 1 and primer 2 sequences provide the recombination region which permits the insertion of the CMV promoter in front of the coding sequence for the human Shh gene. Other heterologous promoters (or other transcriptional regulatory sequences) can be inserted in a genomic hedgehog gene by a similar method.
In still other embodiments, the replacement region merely deletes a negative transcriptional control element of the native gene, e.g., to activate expression, or ablates a positive control element, e.g., to inhibit expression of the targeted gene.

V. Exemplary ptc therapeutic compounds.
In another embodiment, the subject method is carried out using a ptc therapeutic composition. Such compositions can be generated with, for example, compounds which bind to patched and alter its signal transduction activity, compounds which alter the binding and/or enzymatic activity of a protein (e.g., intracellular) involved in patched signal pathway, and compounds which alter the level of expression of a hedgehog protein, a patched protein or a protein involved in the intracellular signal transduction pathway of patched.
The availability of purified and recombinant hedgehog polypeptides facilitates the generation of assay systems which can be used to screen for drugs, such as small organic molecules, which are either agonists or antagonists of the normal cellular function of a hedgehog and/or patched protein, particularly their role in the pathogenesis of epithelial cell proliferation andlor differentiation. In one embodiment, the assay evaluates the ability of a compound to modulate binding between a hedgehog polypeptide and a hedgehog receptor such as patched. In other embodiments, the assay merely scores for the ability of a test compound to alter the signal transduction acitity of the patched protein. In this manner, a variety of hedgehog and/or ptc therapeutics, both proliferative and anti-proliferative in activity, can be identified. A variety of assay formats will suffice and, in light of the present disclosure, will be comprehended by skilled artisan.
In many drug screening programs which test libraries of compounds and natural extracts, high throughput assays are desirable in order to maximize the number of compounds surveyed in a given period of time. Assays which are performed in cell-free systems, such as may be derived with purified or semi-purified proteins, are often preferred as "primary"
screens in that they can be generated to permit rapid development and relatively easy detection of an alteration in a molecular target which is mediated by a test compound.
Moreover, the effects of cellular toxicity and/or bioavailability of the test compound can be generally ignored in the in vitro system, the assay instead being focused primarily on the effect of the drug on the molecular target as may be manifest in an alteration of binding affinity with receptor proteins.
Acordingly, in an exemplary screening assay for ptc therapeutics, the compound of interest is contacted with a mixture including a hedgehog receptor protein (e.g., a cell expressing the patched receptor) and a hedgehog protein under conditions in which it is ordinarily capable of binding the hedgehog protein. To the mixture is then added a composition containing a test compound. Detection and quantification of receptorlhedgehog complexes provides . a means for determining the test compound's efficacy at inhibiting (or potentiating) complex formation between the receptor protein and the hedgehog polypeptide.
The efficacy of the compound can be assessed by generating dose response curves from data obtained using various concentrations of the test compound. Moreover, a control assay can also be performed to provide a baseline for comparison. In the control assay, isolated and purified hedgehog polypeptide is added to the receptor protein, and the formation of receptorlhedgehog complex is quantitated in the absence of the test compound.
In other embodiments, a ptc therapeutic of the present invention is one which disrupts the association of patched with smoothened.
Agonist and antagonists of epithelial cell growth can be distinguished, and the efficacy of the compound can be assessed, by subsequent testing with epithelial cells, e.g., in culture.
In an illustrative embodiment, the polypeptide utilized as a hedgehog receptor can be generated from the patched protein. Accordingly, an exemplary screening assay includes all or a suitable portion of the patched protein which can be obtained from, for example, the human patched gene (GenBank U43148) or other vertebrate sources (see GenBank Accession numbers U40074 for chicken patched and U46155 for mouse patched), as well as from drosophila (GenBank Accession number M28999) or other invertebrate sources.
The patched protein can be provided in the screening assay as a whole protein (preferably expressed on the surface of a cell), or alternatively as a fragment of the full length protein which binds to hedgehog polypeptides, e.g., as one or both of the substantial extracellular domains (e.g.
corresponding to residues Asn120-Ser438 and/or Arg770-Trp1027 of the human patched protein - which are also potential antagonists of hedgehog-dependent signal transduction). For instance, the patched protein can be provided in soluble form, as for example a preparation of one of the extracellular domains, or a preparation of both of the extracellular domains which are covalently connected by an unstructured linker (see, for example, Huston et al. ( 1988) PNAS 85:4879; and U.S. Patent No. 5,091,513). In other embodiments, the protein can be provided as part of a liposomal preparation or expressed on the surface of a cell. The patched WO 99tZ0298 PCT/US98/22227 protein can derived from a recombinant gene, e.g., being ectopically expressed in a -heterologous cell. For instance, the protein can be expressed on oocytes, mammalian cells (e.g., COS, CHO, 3T3 or the like), or yeast cell by standard recombinant DNA
techniques.
These recombinant cells can be used for receptor binding, signal transduction or gene expression assays. Mango et al. (1996) Development 122:1225-1233 illustrates a binding assay of human hedgehog to chick patched protein ectopically expressed in Xenopus laevis oocytes.
The assay system of Marigo et al. can be adapted to the present drug screening assays. As illustrated in that reference, Shh binds to the patched protein in a selective, saturable, dose-dependent manner, thus demonstrating that patched is a receptor for Shh.
Complex formation between the hedgehog polypeptide and a hedgehog receptor may be detected by a variety of techniques. For instance, modulation of the formation of complexes can be quantitated using, for example, detestably labelled proteins such as radiolabelled, fluorescently labelled, or enzymatically labelled hedgehog polypeptides, by immunoassay, or by chromatographic detection.
Typically, for cell-free assays, it will be .desirable to immobilize either the hedgehog receptor or the hedgehog polypeptide to facilitate separation of receptorlhedgehog complexes from uncomplexed forms of one of the proteins, as well as to accommodate automation of the assay. In one embodiment, a fusion protein can be provided which adds a domain that allows the protein to be bound to a matrix. For example, glutathione-S-transferase/receptor (GST/receptor) fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, MO) or glutathione derivatized microtitre plates, which are then combined with the hedgehog polypeptide, e.g. an 35S-labeled hedgehog polypeptide, and the test compound and incubated under conditions conducive to complex formation, e.g. at physiological conditions for salt and pH, though slightly more stringent conditions may be desired. Following incubation, tlae beads are washed to remove any unbound hedgehog polypeptide, and the matrix bead-bound radiolabel determined directly (e.g.
beads placed in scintillant), or in the supernatant after the receptor/hedgehog complexes are dissociated.
Alternatively, the complexes can be dissociated from the bead, separated by SDS-PAGE gel, and the level of hedgehog polypeptide found in the bead fraction quantitated from the gel using standard electrophoretic techniques.

Other techniques for immobilizing proteins on matrices are also available for use in the.
subject assay. For instance, soluble portions of the hedgehog receptor protein can be immobilized utilizing conjugation of biotin and streptavidin. For instance, biotinylated receptor molecules can be prepared from biotin-NHS (N-hydroxy-succinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, IL), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
Alternatively, antibodies reactive with the hedgehog receptor but which do not interfere with hedgehog binding can be derivatized to the wells of the plate, and the receptor trapped in the wells by antibody conjugation. As above, preparations of a hedgehog polypeptide and a test compound are incubated in the receptor-presenting wells of the plate, and the amount of receptorlhedgehog complex trapped in the well can be quantitated. Exemplary methods for detecting such complexes, in addition to those described above for the GST-immobilized complexes, include immunodetection of complexes using antibodies reactive with the hedgehog polypeptide, or which are reactive with the receptor protein and compete for binding with the hedgehog polypeptide; as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with the hedgehog polypeptide. In the instance of the latter, the enzyme can be chemically conjugated or provided as a fusion protein with the hedgehog polypeptide. To illustrate, the hedgehog polypeptide can be chemically cross-linked or genetically fused with alkaline phosphatase, and the amount of hedgehog polypeptide trapped in the complex can be assessed with a chromogenic substrate of the enzyme, e.g.
paranitrophenylphosphate. Likewise, a fusion protein comprising the hedgehog polypeptide and glutathione-S-transferase can be provided, and complex formation quantitated by detecting the GST activity using 1-chloro-2,4-dinitrobenzene (Habig et al (1974) J Biol Chem 249:7130).
For processes which rely on immunodetection for quantitating one of the proteins trapped in the complex, antibodies against the protein, such as the anti-hedgehog antibodies described herein, can be used. Alternatively, the protein to be detected in the complex can be "epitope tagged" in the form of a fusion protein which includes, in addition to the hedgehog polypeptide or hedgehog receptor sequence, a second polypeptide for which antibodies are readily available (e.g. from commercial sources). For instance, the GST fusion proteins described above can also be used for quantification of binding using antibodies against the WO 99/20198 . PCT/US98/22227 GST moiety. Other useful epitope tags include myc-epitopes (e.g., see Ellison et al. ( 1991 ) J
Biol Chem 266:21150-21157) which includes a 10-residue sequence from c-myc, as well as the pFLAG system (International Biotechnologies, Inc.) or the pEZZ-protein A
system (Pharamacia, NJ).
Where the desired portion of the hedgehog receptor (or other hedgehog binding molecule) cannot be provided in soluble form, liposomal vesicles can be used to provide manipulatable and isolatable sources of the receptor. For example, both authentic and recombinant forms of the patched protein can be reconstituted in artificial lipid vesicles (e.g.
phosphatidylcholine liposomes) or in cell membrane-derived vesicles (see, for example, Bear et al. (1992) Cell 68:809-818; Newton et al. (1983) Biochemistry 22:6110-6117;
and Reber et al. (1987) JBiol Chem 262:11369-11374).
In addition to cell-free assays, such as described above, the readily available source of hedgehog proteins provided by the art also facilitates the generation of cell-based assays for identifying small molecule agonists/antagonists and the like. Analogous to the cell-based assays described above for screening combinatorial libraries, cells which are sensitive to hedgehog induction, e.g. patched expressing cells or other epithelially-derived cells sensitive to hedgehog induction, can be contacted with a hedgehog protein and a test agent of interest, with the assay scoring for anything from simple binding to the cell to modulation in hedgehog inductive responses by the target cell in the presence and absence of the test agent. As with the cell-free assays, agents which produce a statistically significant change in hedgehog activities (either inhibition or potentiation) can be identified.
In other emdodiments, the cell-based assay scores for agents which disrupt association of patched and smoothened proteins, e.g., in the cell surface membrane or liposomal preparation.
In addition to characterizing cells that naturally express the patched protein, cells which have been genetically engineered to ectopically express patched can be utilized for drug screening assays. As an example, cells which either express low levels or lack expression of the patched protein, e.g. Xenopus laevis oocytes, COS cells or yeast cells, can be genetically modified using standard techniques to ectopically express the patched protein.
(see Marigo et al., supra).

WO 99n0298 PCf/US98/222Z7 The resulting recombinant cells, e.g., which express a functional patched receptor, can -be utilized in receptor binding assays to identify agonist or anatagonsts of hedgehog binding.
Binding assays can be performed using whole cells. Furthermore, the recombinant cells of the present invention can be engineered to include other heterolgous genes encoding proteins involved in hedgehog-dependent siganl pathways. For example, the gene products of one or more of smoothened, costal-2 and/or fused can be co-expressed with patched in the reagent cell, with assays being sensitive to the functional reconstituion of the hedgehog signal transduction cascade.
Alternatively, liposomal preparations using reconstituted patched protein can be utilized. Patched protein purified from detergent extracts from both authentic and recombinant origins can be reconstituted in in artificial lipid vesicles (e.g.
phosphatidylcholine liposomes) or in cell membrane-derived vesicles (see, for example, Bear et al.
(1992) Cell 68:809-818; Newton et al. (1983) Biochemistry 22:6110-6117; and Reber et al.
(1987) J Biol Chem 262:11369-11374). The lamellar structure and size of the resulting liposomes can be characterized using electron microscopy. External orientation of the patched protein in the reconstituted membranes can be demonstrated, for example, by immunoelectron microscopy.
The hedgehog protein binding activity of liposomes containing patched and liposomes without the protein in the presence of candidate agents can be compared in order to identify potential modulators of the hedgehog patched interaction.
The hedgehog protein used in these cell-based assays can be provided as a purified source (natural or recombinant in origin), or in the form of cells/tissue which express the protein and which are co-cultured with the target cells. As in the cell-free assays, where simple binding (rather than induction) is the hedgehog activity scored for in the assay, the protein can be labelled by any of the above-mentioned techniques, e.g., fluorescently, enzymatically or radioactively, or detected by immunoassay.
In addition to binding studies, functional assays can be used to identified modulators, i.e., agonists or antagonists, of hedgehog or patched activities. By detecting changes in intracellular signals, such as alterations in second messengers or gene expression, in patched expressing cells contacted with a test agent, candidate agonists and antagonists to patched signaling can be identified.

WO 99/20298 PCT/US98lZ2227 A number of gene products have been implicated in patched mediated signal .
transduction, including patched, the transcription factor cubitus interruptus (ci), the serine/threonine kinase fused (fu) and the gene products of costal-2, smoothened and suppressor of fused.
The interaction of a hedgehog protein with patched sets in motion a cascade involving the activation and inhibition of downstream effectors, the ultimate consequence of which is, in some instances, a detectable change in the transcription or translation of a gene. Potential transcriptional targets of patched signaling are the patched gene itself (Hidalgo and Ingham, 1990 Development 110, 291-301; Marigo et al., 1996 ) and the vertebrate homologs of the drosophila cubitus interruptus gene, the GLI genes (Hui et al. (1994) Dev Biol 162:402-413).
Patched gene expression has been shown to be induced in cells of the limb bud and the neural plate that are responsive to Shh. (Marigo et al. ( 1996) PNAS, in press;
Marigo et al. ( 1996}
Development 122:1225-1233). The GLI genes encode putative transcription factors having zinc finger DNA binding domains (Urenic et al. (1990) Genes & Dev 4:1053-1067;
Kinzler et al. (1990) Mol Cell Biol 10:634-642). Transcription of the GLI gene has been reported to be upregulated in response to hedgehog in limb buds, while transcription of the GLl3 gene is downregulated in response to hedgehog induction (Marigo et al. ( 1996) Development 122:1225-1233). By selecting transcriptional regulatory sequences from such target genes, e.g.
from patched or GLI genes, that are responsible for the up- or down regulation of these genes im response to patched signalling, and operatively linking such promoters to a reporter gene, one can derive a transcription based assay which is sensitive to the ability of a specific test compound to modify patched signalling pathways. Expression of the reporter gene, thus, provides a valuable screening tool for the development of compounds that act as agonists or antagonists of ptc induction of differentiation/quiescence.
Reporter gene based assays of this invention measure the end stage of the above described cascade of events, e.g., transcriptional modulation. Accordingly, in practicing one embodiment of the assay, a reporter gene construct is inserted into the reagent cell in order to generate a detection signal dependent on ptc signaling. To identify potential regulatory elements responsive to ptc signaling present in the transcriptional regulatory sequence of a target gene, nested deletions of genomic clones of the target gene can be constructed using standard techniques. See, for example, Current Protocols in Molecular Bioloev, Ausubel, -F.M. et al. (eds.) Greene Publishing Associates, (1989); U.S. Patent 5,266,488; Sato et al.
(1995) JBiol Chem 270:10314-10322; and Kube et al. (1995) Cytokine 7:1-7. A
nested set of DNA fragments from the gene's 5'-flanking region are placed upstream of a reporter gene, such as the luciferase gene, and assayed for their ability to direct reporter gene expression in patched expressing cells. Host cells transiently transfected with reporter gene constructs can be scored for the induction of expression of the reporter gene in the presence and absence of hedgehog to determine regulatory sequences which are responsice to patched dependent signalling.
In practicing one embodiment of the assay, a reporter gene construct is inserted into the reagent cell in order to generate a detection signal dependent on second messengers generated by induction with hedgehog protein. Typically, the reporter gene construct will include a reporter gene in operative linkage with one or more transcriptional regulatory elements responsive to the hedgehog activity, with the level of expression of the reporter gene providing the hedgehog-dependent detection signal. The amount of transcription from the reporter gene may be measured using any method known to those of skill in the art to be suitable. For example, mRNA expression from the reporter gene may be detected using RNAse protection or RNA-based PCR, or the protein product of the reporter gene may be identified by a characteristic stain or an intrinsic activity. The amount of expression from the reporter gene is then compared to the amount of expression in either the same cell in the absence of the test compound (or hedgehog) or it may be compared with the amount of transcription in a substantially identical cell that lacks the target receptor protein. Any statistically or otherwise significant difference in the amount of transcription indicates that the test compound has in some manner altered the signal transduction of the patched protein, e.g., the test compound is a potential ptc therapeutic.
As described in further detail below, in preferred embodiments the gene product of the reporter is detected by an intrinsic activity associated with that product.
For instance, the reporter gene may encode a gene product that, by enzymatic activity, gives rise to a detection signal based on color, fluorescence, or luminescence. In other preferred embodiments, the reporter or marker gene provides a selective growth advantage, e.g., the reporter gene may wo ~noZ9s rcrms9anz2Z~

enhance cell viability, relieve a cell nutritional requirement, and/or provide resistance to a -drug.
Preferred reporter genes are those that are readily detectable. The reporter gene may also be included in the construct in the form of a fusion gene with a gene that includes desired transcriptional regulatory sequences or exhibits other desirable properties.
Examples of reporter genes include, but are not limited to CAT (chloramphenicol acetyl transferase) (Alton and Vapnek {1979), Nature 282: 864-869) luciferase, and other enzyme detection systems, such as beta-galactosidase; firefly luciferase (deWet et al. (1987), Mol.
Cell. Biol. 7:725-737);
bacterial luciferase (Engebrecht and Silverman (1984), PNAS 1: 4154-4158;
Baldwin et al.
(1984), Biochemistry 23: 3663-3667); alkaline phosphatase (Toh et al. {1989) Eur. J.
Biochem. 182: 231-238, Hall et al. (1983) J. Mol. Appl. Gen. 2: 101), human placental secreted alkaline phosphatase (Cullen and Malim (1992) Methods in Enzymol.
216:362-368).
Transcriptional control elements which may be included in a reporter gene construct include, but are not limited to, promoters, enhancers, and repressor and activator binding sites.
Suitable transcriptional regulatory elements may be derived from the transcriptional regulatory regions of genes whose expression is induced after modulation of a patched signal transduction pathway. The characteristics of preferred genes from which the transcriptional control elements are derived include, but are not limited to, low or undetectable expression in quiescent cells, rapid induction at the transcriptional level within minutes of extracellular simulation, induction that is transient and independent of new protein synthesis, subsequent shut-off of tianscription requires new protein synthesis, and mRNAs transcribed from these genes have a short half life. It is not necessary for all of these properties to be present.
In yet other embodiments, second messenger generation can be measured directly in the detection step, such as mobilization of intracellular calcium, phospholipid metabolism or adenylate cyclase activity are quantitated, for instance, the products of phospholipid hydrolysis IP3, DAG or cAMP could be measured For example, recent studies have implicated protein kinase A (PKA) as a possible component of hedgehoglpatched signaling (Hammerschmidt et al. ( 1996) Genes & Dev 10:647). High PKA activity has been shown to antagonize hedgehog signaling in these systems. Although it is unclear whether PKA acts directly downstream or in parallel with hedgehog signaling, it is possible that hedgehog signalling occurs via inhibition
9$ PCT/IJS98IZ2227 of PKA activity. Thus, detection of PKA activity provides a potential readout for the instant -assays.
In a preferred embodiment, the ptc therapeutic is a PKA inhibitor. A variety of PKA
inhibitors are known in the art, including both peptidyl and organic compounds. For instance, the ptc therapeutic can be a 5-isoquinolinesulfonamide, such as represented in the general formula:
R2~N~R1 I
O=S=O
N~

wherein, R1 and R2 each can independently represent hydrogen, and as valence and stability permit a lower alkyl, a lower alkenyl, a lower alkynyl, a carbonyl (such as a carboxyl, an ester, a formate, or a ketone), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an amino, an acylarnino, an amido, a cyano, a nitro, an azido, a sulfate, a sulfonate, a sulfonamido, -(CH2)m R8, -(CH2)m-OH, -(CH2)m O-lower alkyl, -(CH2)m-O-lower alkenyl, -(CH2)n-O-(CH2)m R8, -(CH2)m SH, -(CH2)m-S-lower alkyl, -(CH2)m S-lower alkenyl, -(CH2)n S-(CH2)m-R8, or R~ and R2 taken together with N form a heterocycle (substituted or unsubstituted);
R3 is absent or represents one or more substitutions to the isoquinoline ring such as a lower alkyl, a lower alkenyl, a lower alkynyl, a carbonyl (such as a carboxyl, an ester, a formate, or a ketone), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an amino, an acylamino, an amido, a cyano, a nitro, an azido, a sulfate, a sulfonate, a sulfonamido, -(CH2)m-Rg, -(CH2)m OH, -(CH2)m O-lower alkyl, -(CH2)m O-lower alkenyl, -(CH2)"O-(CH2)m-Rg, -(CH2)m SH, -(CH2)m-S-lower alkyl, -(CH2)m S-lower alkenyl, -(CH2)ri S-(CH2)m-R8~

WO 99!20298 PCT/US98/22227 Rg represents a substituted or unsubstituted aryl, aralkyl, cycloalkyl, cycloalkenyl, or-heterocycle; and n and m are independently for each occurrence zero or an integer in the range of 1 to 6.
In a preferred embodiment, the PKA inhibitor is N-[2-((p-bromocinnamyl)amino)ethyl]-5-isoquinolinesulfonamide (H-89; Calbiochem Cat. No. 371963), e.g., having the formula:
N / \
NH H I
Br O=S=O
I
N~
In another embodiment, the PKA inhibitor is 1-(5-isoquinolinesulfonyl)-2-methylpiperazine (H-7; Calbiochem Cat. No. 371955), e.g., having the formula:
N
N
I
O=S=O
I
N~
In still other embodiments, the PKA inhibitor is KT5720 (Calbiochem Cat. No.
42031 ~), having the structure WO 99!20298 PGT/US98/22227 OH
CH3(CH2)4CH200C~"~
",,.. O
A variety of nucleoside analogs are also useful as PKA inhibitors. For example, the subject method can be carried out cyclic AMP analogs which inhibit the kinase activity of PICA, as for example, 8-bromo-cAMP or dibutyryl-cAMP
NH2 NHCO(CH2)2CH3 N \ N N \ N
i ~Br ~ i 0 N N O\ N N
O- \ O O O-P\ O O
O, O
OH OCO(CH2)2CH3 Exemplary peptidyl inhibitors of PICA activity include the PKA Heat Stable Inhibitor (isoform a; see, for example, Calbiochem Cat. No. 539488, and Wen et al.
(1995) JBiol Chem 270:2041 ).
Certain hedehog receptors may stimulate the activity of phospholipases.
Inositol lipids can be extracted and analyzed using standard lipid extraction techniques.
Water soluble derivatives of all three inositol lipids (IPA, IP2, IP3) can also be quantitated using radiolabelling techniques or HPLC.
The mobilization of intracellular calcium or the influx of calcium from outside the cell may be a response to hedgehog stimulation or lack there of. Calcium flux in the reagent cell can be measured using standard techniques. The choice of the appropriate calcium indicator, fluorescent, bioluminescent, metallochromic, or Ca++-sensitive microelectrodes depends on rv H

WO 99/0298 PGT/US98f12227 the cell type and the magnitude and time constant of the event under study (Borle (1990) Environ Health Perspect 84:45-56). As an exemplary method of Cap-'+' detection, cells could be loaded with the Ca'+"~'sensitive fluorescent dye furs-2 or indo-1, using standard methods, and any change in Cap measured using a f~orometer.
In certain embodiments of the assay, it may be desirable to screen for changes in cellular phosphorylation. As an example, the drosophila gene fused (fu) which encodes a serine/threonine kinase has been identified as a potential downstream target in hedgehog signaling. (Preat et al., 1990 Nature 347, 87-89; Therond et al. 1993, Mech.
Dev. 44. 65-80).
The ability of compounds to modulate serine/threonine kinase activation could be screened using colony immunoblotting (Lyons and Nelson ( 1984) Proc. Natl. Acad. Sci.
USA 81:7426-7430) using antibodies against phosphorylated serine or threonine residues.
Reagents for performing such assays are commercially available, for example, phosphoserine and phosphothreonine specific antibodies which measure increases in phosphorylation of those residues can be purchased from comercial sources.
IS In yet another embodiment, the ptc therapeutic is an antisense molecule which inhibits expression of a protein involved in a patched mediated signal transduction pathway. To illustrate, by inhibiting the expression of a protein which are involved in patched signals, such as fused, costal-2, smoothened and/or Gli genes, the ability of the patched signal pathways) to inhibit proliferation of a cell can be altered, e.g., potentiated or repressed.
As used herein, "antisense" therapy refers to administration or in situ generation of oligonucleotide probes or their derivatives which specifically hybridize (e.g.
bind) under cellular conditions with cellular mRNA and/or genomic DNA encoding a hedgehog protein, patched, or a protein involved in patched-mediated signal transduction. The hybridization should inhibit expression of that protein, e.g. by inhibiting transcription and/or translation.
The binding may be by conventional base pair complementarity, or, for example, in the case of binding to DNA duplexes, through specific interactions in the major groove of the double helix. In general, "antisense" therapy refers to the range of techniques generally employed in the art, and includes any therapy which relies on specific binding to oligonucleotide sequences.

An antisense construct of the present invention can be delivered, for example, as an expression plasmid which, when transcribed in the cell, produces RNA which is complementary to at least a unique portion of the target cellular mRNA.
Alternatively, the antisense construct .is an oligonucleotide probe which is generated ex vivo and which, when introduced into the cell causes inhibition of expression by hybridizing with the mRNA and/or genomic sequences of a target gene. Such oligonucleotide probes are preferably modified oligonucleotide which are resistant to endogenous nucleases, e.g. exonucleases and/or endonucleases, and is therefore stable in vivo. Exemplary nucleic acid molecules for use as antisense oligonucleotides are phosphoramidate, phosphothioate and methylphosphonate analogs of DNA (see also U.S. Patents 5,176,996; 5,264,564; and 5,256,775).
Additionally, general approaches to constructing oligomers useful in antisense therapy have been reviewed, for example, by Van der Krol et al. (1988) Biotechniques 6:958-976; and Stein et al. (1988) Cancer Res 48:2659-2668.
Several considerations should be taken into account when constructing antisense oligonucleotides for the use in the methods of the invention: ( 1 ) oligos should have a GC
content of 50% or more; (2) avoid sequences with stretches of 3 or more G's;
and (3) oligonucleotides should not be longer than 25-26 mers. When testing an antisense oligonucleotide, a mismatched control can be constructed. The controls can be generated by reversing the sequence order of the corresponding antisense oligonucleotide in order to conserve the same ratio of bases.
In an illustrative embodiment, the ptc therapeutic can be an antisense construct for inhibiting the expression of patched, e.g., to mimic the inhibition of patched by hedgehog.
Exemplary antisense constructs include:
5'-GTCCTGGCGCCGCCGCCGCCGTCGCC
5'-TTCCGATGACCGGCCTTTCGCGGTGA
5'-GTGCACGGAAAGGTGCAGGCCACACT

_77_ YI. Exemplary pharmaceutical preparations of hedgehog and ptc therapeutics The source of the hedgehog and ptc therapeutics to be formulated will depend on the particular form of the agent. Small organic molecules and peptidyl fragments can be chemically synthesized and provided in a- pure form suitable for pharmaceutical/cosmetic usage. Products of natural extracts can be purified according to techniques known in the art.
For example, the Cox et al. U.S. Patent 5,286,654 describes a method for purifying naturally occurring forms of a secreted protein and can be adapted for purification of hedgehog polypeptides. Recombinant sources of hedgehog polypeptides are also available.
For example, the gene encoding hedgehog polypeptides, are known, inter alia, from PCT
publications WO 95/18856 and WO 96/17924.
Those of skill in treating epithelial tissues can determine the effective amount of an hedgehog or ptc therapeutic to be formulated in a pharmaceutical or cosmetic preparation.
The hedgehog or ptc therapeutic formulations used in the method of the invention are most preferably applied in the form of appropriate compositions. As appropriate compositions there may be cited all compositions usually employed for systemically or topically administering drugs. The pharmaceutically acceptable carrier should be substantially inert. so as not to act with the active component. Suitable inert carriers include water, alcohol polyethylene glycol, mineral oil or petroleum gel, propylene glycol and the like.
To prepare the pharmaceutical compositions of this invention, an effective amount of the particular hedgehog or ptc therapeutic as the active ingredient is combined in intimate admixture with a pharmaceutically acceptable Garner, which carrier may take a wide variety of forms depending on the form of preparation desired for administration. These pharmaceutical compositions are desirable in unitary dosage form suitable, particularly, for administration orally, rectally, percutaneously, or by parenteral injection. For example, in preparing the compositions in oral dosage form, any of the usual pharmaceutical media may be employed such as, for example, water, glycols, oils, alcohols and the like in the case of oral liquid preparations such as suspensions, syrups, elixirs and solutions; or solid can-iers such as starches, sugars, kaolin, lubricants, binders. disintegrating agents and the like in the case of powders, pills, capsules, and tablets. Because of their ease in administration, tablets and capsules represents the most advantageous oral dosage unit form, in which case solid WO 99/20298 PCT/US98/2222'7 _78_ pharmaceutical carriers are obviously employed. For parenteral compositions, the carrier will.
usually comprise sterile water, at least in large part, though other ingredients, for example, to aid solubility, may be included. Injectable solutions, for example, may be prepared in which the carrier comprises saline solution, glucose solution or a mixture of saline and glucose solution. Injectable suspensions may also be prepared in which case appropriate liquid carriers, suspending agents and the like may be employed. Also included are solid form preparations which are intended to be converted, shortly before use, to liquid form preparations. In the compositons suitable for percutaneous administration, the carrier optionally comprises a penetration enhancing agent and/or a suitable wetting agent, optionally combined with suitable additives of any nature in minor proportions, which additives do not introduce a significant deleterious effect on the skin.
In addition to the direct topical application of the preparations they can be topically administered by other methods, for example, encapsulated in a temperature and/or pressure sensitive matrix or in film or solid Garner which is soluble in body fluids and the like for subsequent release, preferably sustained-release of the active component.
As appropriate compositions for topical application there may be cited all compositions usually employed for topically administering therapeuitcs, e.g., creams, gellies, dressings, shampoos, tinctures, pastes, ointments, salves, powders, liquid or semiliquid formulation and the like. Application of said compositions may be by aerosol e.g. with a propellent such as nitrogen carbon dioxide, a freon, or without a propellent such as a pump spray, drops, lotions, or a semisolid such as a thickened composition which can be applied by a swab.
In particular compositions, semisolid compositions such as salves, creams, pastes, gellies, ointments and the like will conveniently be used.
It is especially advantageous to formulate the subject compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used in the specification and claims herein refers to physically discreate units suitable as unitary dosages, each unit containing a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical Garner. Examples of such dosage unit forms are tablets (including scored or coated tablets), capsules, pills, powders packets, wafers, injectable solutions or suspensions, teaspoonfuls, tablespoonfuls and the like,-and segregated multiples thereof.
The pharmaceutical preparations of the present invention can be used, as stated above, for the many applications whcih can be considered cosmetic uses. Cosmetic compositions known in the art, preferably hypoallergic and pH controlled are especially preferred, and include toilet waters, packs, lotions, skin milks or milky lotions. The preparations contain, besides the hedgehog or ptc therapeutic, components usually employed in such preparations.
Examples of such components are oils, fats, waxes, surfactants, humectants, thickening agents, antioxidants, viscosity stabilizers, chelating agents, buffers, preservatives, perfumes, dyestuffs, lower alkanols, and the like. If desired, further ingredients may be incorporated in the compositions, e.g. antiinflammatory agents, antibacterials, antifungals, disinfectants, vitamins, sunscreens, antibiotics, or other anti-acne agents.
Examples of oils comprise fats and oils such as olive oil and hydrogenated oils; waxes such as beeswax and lanolin; hydrocarbons such as liquid paraffin, ceresin, and squalane; fatty acids such as stearic acid and oleic acid; alcohols such as cetyl alcohol, stearyl alcohol, lanolin alcohol, and hexadecanol; and esters such as isopropyl myristate, isopropyl palmitate and butyl stearate. As examples of surfactants there may be cited anionic surfactants such as sodium stearate, sodium cetylsulfate, polyoxyethylene laurylether phosphate, sodium N-acyl glutamate; cationic surfactants such as stearyldimethylbenzylammonium chloride and stearyltrimethylammonium chloride; ampholytic surfactants such as alkylaminoethylglycine hydrocloride solutions and lecithin; and nonionic surfactants such as glycerin monostearate, sorbitan monostearate, sucrose fatty acid esters, propylene glycol monostearate, polyoxyethylene oleylether, polyethylene glycol monostearate, polyoxyethylene sorbitan monopalmitate, polyoxyethylene coconut fatty acid monoethanolamide, polyoxypropylene glycol (e.g. the materials sold under the trademark "Pluronic"), polyoxyethylene castor oil, and polyoxyethylene lanolin. Examples of humectants include glycerin, 1,3-butylene glycol, and propylene glycol; examples of lower alcohols include ethanol and isopropanol;
examples of thickening agents include xanthan gum, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, polyethylene glycol and sodium carboxymethyl cellulose; examples of antioxidants comprise butylated hydroxytoluene, butylated hydroxyanisole, propyl gallate, citric acid and ethoxyquin; examples of chelating agents include disodium edetate and ethanehydroxy diphosphate; examples of buffers comprise citric acid, sodium citrate, boric acid; borax, and disodium hydrogen phosphate; and examples of preservatives are methyl parahydroxybenzoate, ethyl parahydroxybenzoate, dehydroacetic acid, salicylic acid and benzoic acid.
For preparing ointments, creams, toilet waters, skin milks, and the like, typically from 0.01 to 10% in particular from 0.1 to 5% and more in particular from 0.2 to 2.5% of the active ingredient, e.g., of the hedgehog or ptc therapeutic, will be incorporated in the compositions.
In ointments or creams, the carrier for example consists of 1 to 20%, in particular 5 to 15% of a humectant, 0.1 to 10% in particular from 0.5 to 5% of a thickener and water;
or said carrier may consist of 70 to 99%, in particular 20 to 95% of a surfactant, and 0 to 20%, in particular 2.5 to 1 S% of a fat; or 80 to 99.9% in particular 90 to 99% of a thickener;
or 5 to 15% of a surfactant, 2-15% of a humectant, 0 to 80% of an oil, very small ( < 2%) amounts of preservative, coloring agent and/or perfume, and water. In a toilet water, the carrier for example consists of 2 to 10% of a lower alcohol, 0.1 to 10% or in particular 0.5 to 1 % of a surfactant, 1 to 20%, in particular 3 to 7% of a humectant, 0 to 5% of a buffer, water and small amounts ( < 2%) of preservative, dyestuff andlor perfume. In a skin milk, the Garner typically consists of 10-SO% of oil, 1 to 10% of surfactant, 50-80% of water and 0 to 3%
of preservative and/or perfume. In the aforementioned preparations, all % symbols refer to weight by weight percentage.
Particular compositions for use in the method of the present invention are those wherein the hedgehog or ptc therapeutic is formulated in liposome-containing compositions.
Liposomes are artificial vesicles formed by amphiphatic molecules such as polar lipids, for example, phosphatidyl cholines, ethanolamines and serines, sphingomyelins, cardiolipins, plasmalogens, phosphatidic acids and cerebiosides. Liposomes are formed when suitable amphiphathic molecules are allowed to swell in water or aqueous solutions to form liquid crystals usually of multilayer structure comprised of many bilayers separated from each other by aqueous material (also referred to as coarse liposomes). Another type of liposome known to be consisting of a single bilayer encapsulating aqueous material is referred to as a unilamellar vesicle. If water-soluble materials are included in the aqueous phase during the swelling of the lipids they become entrapped in the aqueous layer between the lipid bilayers.
Water-soluble active ingredients such as, for example, various salt forms of a hedgehog polypeptide, are encapsulated in the aqueous spaces between the molecular layers. The lipid soluble active ingredient of hedgehog or ptc therapeutic, such as an organic mimetic. is predominantly incorporated into the lipid layers, although polar head groups may protude from the layer into the aqueous space. The encapsulation of these compounds can be achieved by a number of methods. The method most commonly used involves casting a thin film of phospholipid onto the walls of a flask by evaporation from an organic solvent.
When this film is dispersed in a suitable aqueous medium, multilamellar liposomes are formed.
Upon suitable sonication, the coarse liposomes form smaller similarly closed vesicles.
Water-soluble active ingredients are usually incorporated by dispersing the cast film with an aqueous solution of the compound. The unencapsulated compound is then removed by centrifugation, chromatography, dialysis or other art-known suitable procedures. The lipid-soluble active ingredient is usually incorporated by dissolving it in the organic solvent with the phospholipid prior to casting the film. If the solubility of the material in the lipid phase is not exceeded or the amount present is not in excess of that which can be bound to the lipid, liposomes prepared by the above method usually contain most of the material bound in the lipid bilayers; separation of the liposomes from unencapsulated material is not required.
A particularly convenient method for preparing liposome formulated forms of hedgehog and ptc therapeutics is the method described in EP-A-253,619, incorporated herein by reference. In this method, single bilayered liposomes containing encapsulated active ingredients are, prepared by dissolving the lipid component in an organic medium, injecting the organic solution of the lipid component under pressure into an aqueous component while simultaneously mixing the organic and aqueous components with a high speed homogenizer or mixing means; whereupon the liposomes are formed spontaneously.
The single bilayered liposomes containing the encapsulated hedgehog or ptc therapeutic can be employed directly or they can be employed in a suitable pharmaceutically acceptable carrier for topical administration. The viscosity of the liposomes can be increased by the addition of one or more suitable thickening agents such as, for example xanthan gum.

wo ~no29a pcrnUS9sn2n~

hydroxypropyl cellulose, hydroxypropyl methylcellulose and mixtures thereof.
The aqueous-component may consist of water alone or it may contain electrolytes, buffered systems and other ingredients, such as, for example, preservatives. Suitable electrolytes which can be employed include metal salts such as alkali metal and alkaline earth metal salts. The preferred metal salts are calcium chloride, sodium chloride and potassium chloride. The concentration of the electrolyte may vary from zero to 260 mM, preferably from 5 mM to 160 mM.
The aqueous component is placed in a suitable vessel which can be adapted to effect homogenization by effecting great turbulence during the injection of the organic component.
Homogenization of the two components can be accomplished within the vessel, or, alternatively, the aqueous and organic components may be injected separately into a mixing means which is located outside the vessel. In the latter case, the liposomes are formed in the mixing means and then transferred to another vessel for collection purpose.
The organic component consists of a suitable non-toxic, pharmaceutically acceptable solvent such as, for example ethanol, glycerol, propylene glycol and polyethylene glycol, and a suitable phospholipid which is soluble in the solvent. Suitable phospholipids which can be employed include lecithin, phosphatidylcholine, phosphatydylserine, phosphatidylethanol amine, phosphatidylinositol, lysophosphatidylcholine and phospha-tidyl glycerol, for example.
Other lipophilic additives may be employed in order to selectively modify the characteristics of the liposomes. Examples of such other additives include stearylamine, phosphatidic acid, tocopherol, cholesterol and lanolin extracts.
In addition, other ingredients which can prevent oxidation of the phospholipids may be added to the organic component. Examples of such other ingredients include tocopherol, butylated hydroxyanisole, butylated hydroxytoluene, ascorbyl palmitate and ascorbyl oleate.
Preservatives such a benzoic acid, methyl paraben and propyl paraben may also be added.
Apart from the above-described compositions, use may be made of covers, e.g.
plasters, bandages, dressings, gauze pads and the like, containing an appropriate amount of a hedgehog or ptc therapeutic. In some cases use may be made of plasters, bandages, dressings, gauze pads and the like which have been impregnated with a topical formulation containing the therapeutic formulation.

Exemplification The invention now being generally described, it wily be more readily understood by reference to the following examples which~re included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention.
Purification of hedgehog protein.
Human sonic hedgehog protein (residues 24-197) was expressed in the baculovirus/insect cell system (Roelink et al. (1995) Cell 81:445-455). The conditioned medium was loaded onto Fast Flo SP agarose equilibrated with 50 mM potassium phosphate, 0.5 mM DTT, pH 7Ø The column was washed with this buffer, and then eluted with a gradient to 10. M NaCI. Fractions were assayed for the induction of alkaline phosphatase activity on mesenchymal stem cells (C3H10T1/2 cells, see, e.g., Wang et al.
(1993) Growth Factors 9:57-71 ) and then pooled on the basis of this activity and also by purity on SDS gels.
The pooled material was concentrated on an Amicon ultra filtration unit (PM 10 membrane) and diafiltered against 10 mM Tris, pH 7.4, 0.5 mM DTT. Protein was estimated by the Bradford method using gamma globulin as a standard.
Preparation of collagen sponge.
Collagen sponge was washed extensively in MilliQ water to remove any surfactants and additives from the manufacturer. The sponge was then washed in 70%
ethanol, then dried in vacuo.
Preparation of implants.
Protein was added to 1.0 - 1.5 mm by 8-10 mm pieces of collagen sponge (1.5-3.0 mg in weight). In some cases zinc sulfate was added to a final concentration of 0.2 mM before the hedgehog protein was added to the collagen sponge. The reconstituted sponges were then .
frozen and lyophilized.
Implantation.
Sponges were implanted either subcutaneously in the thoracic region of Sprague Dawley rats (4-8 weeks old) or in the thigh muscle of rabbits (11-14 weeks old). Animals were maintained for 2-5 weeks before removing the implant. The implant was then fixed in 4% formalin or 4% paraformaldehyde and then embedded in JB-4 resin. Sections were stained with toluidine blue (Wang et al. (1988) PNAS 85:9484-9488).
The induction of new hair follicles, sebaceous glands, and other dermal structures were identified by its distinctive morphology. The careful subcutaneous or intramuscular placement of our implants and the careful removal of these implants preclude the possibility of contamination from existing dermal structures. Also, the appearance of more immature hair follicles is seen in the implants of shorter (2 week) duration.
Biopsy slides were obtained from an intramuscular implant taken out of a rabbit muscle at three weeks and stained with hematoxylin and eosin. Similar slides were examined of an intramuscular implant, rabbit muscle, three weeks, stained with toluidine blue. Slides of certain samples revealed a tissue morphology indicating the presence of follicle- and hair-like structures forming in the intramuscular tissue.
Hair induction by Shh As a follow-up to the above experiments, hedgehog-loaded collagen sponges were implanted under the shaved skin of mice. As indicated in Figures 1 A-C, the hedgehog preparations were able to induce hair growth over the implants. Moreover, Ihh protein modified at the C terminus with a Von Willebrand's factor collagen binding site was active in hair growth, indicating a localized inducing activity of the implanted protein.
All of the above-cited references and publications are hereby incorporated by reference.

Equivalents Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, numerous equivalents to the specific polypeptides, nucleic acids, methods, assays and reagents described herein. Such equivalents are considered to be within the scope of this invention.

wo ~noz9s pcrms9snzz2~
SEQUENCE LISTING
(2) INFORMATION FOR SEQ ID NO:1:
(i) SEQUENCE CHARACTERISTICS: .
(A) LENGTH: 1277 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..1275 (xi)SEQUENCE
DESCRIPTION:
SEQ
ID
N0:1:

MetValGlu MetLeuLeu LeuThrArg IleLeuLeu ValGlyPhe Ile CysAlaLeu LeuValSer SerGlyLeu ThrCysGly ProGlyArg Gly IleGlyLys ArgArgHis ProLysLys LeuThrPro LeuAlaTyr Lys GlnPheIle ProAsnVal AlaGluLys ThrLeuGly AlaSerGly Arg TyrGluGly LysIleThr ArgAsnSer GluArgPhe LysGluLeu Thr ProAsnTyr AsnProAsp IleIlePhe LysAspGlu GluAsnThr Gly AlaAspArg LeuMetThr GlnArgCys LysAspLys LeuAsnAla Leu AlaIleSer ValMetAsn GlnTrpPro GlyValLys LeuArgVal Thr GluGlyTrp AspGluAsp GlyHisHis SerGluGlu SerLeuHis Tyr GluGlyArg AlaValAsp IleThrThr SerAspArg AspArgSer Lys -2=
TyrGly MetLeuAla ArgLeuAla ValGluAla GlyPheAsp TrpVal TyrTyr GluSerLys AlaHisIle HisCysSer ValLysAla GluAsn SerVal AlaAlaLys SerGlyGly CysPhePro GlySerAla ThrVal 195 200 ~' 205 HisLeu GluHisGly GlyThrLys LeuValLys AspLeuSer ProGly AspArg ValLeuAla AlaAspAla AspGlyArg LeuLeuTyr SerAsp PheLeu ThrPheLeu AspArgMet AspSerSer ArgLysLeu PheTyr ValIle GluThrArg GlnProArg AlaArgLeu LeuLeuThr AlaAla HisLeu LeuPheVal AlaProGln HisAsnGln SerGluAla ThrGly SerThr SerGlyGln AlaLeuPhe AlaSerAsn ValLysPro GlyGln ArgVal TyrValLeu GlyGluGly GlyGlnGln LeuLeuPro AlaSer ValHis SerValSer LeuArgGlu GluAlaSer GlyAlaTyr AlaPro LeuThr AlaGlnGly ThrIleLeu IleAsnArg ValLeuAla SerCys TyrAla ValIleGlu GluHisSer TrpAlaHis TrpAlaPhe AlaPro PheArg LeuAlaGln GlyLeuLeu AlaAlaLeu CysProAsp GlyAla IlePro ThrAlaAla ThrThrThr ThrGlyIle HisTrpTyr SerArg LeuLeu TyrArgIle GlySerTrp ValLeuAsp GlyAspAla LeuHis Pro Leu Gly Met Val Ala Pro Ala Ser (2) INFORMATION FOR SEQ ID N0:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1190 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
{ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..1191 (xi)SEQUENCE ID
DESCRIPTION: N0:2:
SEQ

MetAla LeuProAla SerLeuLeu ProLcuCys CysLeuAla LeuLeu AlaLeu SerAlaGln SerCysGly ProGlyArg GlyProVal GlyArg ArgArg TyrValArg LysGlnLeu ValProLeu LeuTyrLys GlnPhe ValPro SerMetPro GluArgThr LeuGlyAla SerGiyPro AlaGlu GlyArg ValThrArg GlySerGlu ArgPheArg AspLeuVal ProAsn TyrAsn ProAspIle IlePheLys AspGluGlu AsnSerGly AlaAsp ArgLeu MetThrGlu ArgCysLys GluArgVal AsnAlaLeu AlaIle AlaVal MetAsnMet TrpProGly ValArgLeu ArgValThr GluGly TrpAsp GluAspGly HisHisAla GlnAspSer LeuHisTyr GluGly ArgAla LeuAspIle ThrThrSer AspArgAsp ArgAsnLys TyrGly LeuLeu AlaArgLeu AlaValGlu AlaGlyPhe AspTrpVal TyrTyr GluSer ArgAsnHis IleHisVal SerValLys AlaAspAsn SerLeu 180. 185 190 AlaVal ArgAlaGly GlyCysPhe ProGlyAsn AlaThrVa1 ArgLeu ArgSer GlyGluArg LysGlyLeu ArgGluLeu HisArgGly AspTrp ValLeu AlaAlaAsp AlaAlaGly ArgValVal ProThrPro ValLeu LeuPhe LeuAspArg AspLeuGln ArgArgAla SerPheVal AlaVal GluThr GluArgPro ProArgLys LeuLeuLeu ThrProTrp HisLeu ValPhe AlaAlaArg GlyProAla ProAlaPro GlyAspPhe AlaPro ValPhe AlaArgArg LeuArgAla GlyAspSer ValLeuAla ProGly GlyAsp AlaLeuGln ProAlaArg ValAlaArg ValAlaArg GluGlu AlaVal GlyValPheAla ProLeu ThrAlaHisGly ThrLeu LeuVal AsnAsp ValLeuAlaSer CysTyr AlaValLeuGlu SerHis GlnTrp AlaHis ArgAlaPheAla ProLeu ArgLeuLeuHis AlaLeu GlyAla LeuLeu ProGlyGlyAla ValGln ProThrGlyMet HisTrp TyrSer ArgLeu LeuTyrArgLeu AlaGlu GluLeuMetGly ' (2) INFORMATION FOR SEQ ID N0:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1281 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..1233 (xi)SEQUENCE
DESCRIPTION:
SEQ
ID
N0:3:

MetSer ProAlaTrp LeuArgPro ArgLeuArg PheCysLeu PheLeu LeuLeu LeuLeuLeu ValProAla AlaArgGly CysGlyPro GlyArg ValVal GlySerArg ArgArgPro ProArgLys LeuValPro LeuAla TyrLys GlnPheSer ProAsnVal ProGluLys ThrLeuGly AiaSer GlyArg TyrGluGly LysIleAla ArgSerSer GluArgPhe LysGlu LeuThr ProAsnTyr AsnProAsp IleIlePhe LysAspGlu GluAsn ThrGly AlaAspArg LeuMetThr GlnArgCys LysAspArg LeuAsn SerLeu AlaIleSer ValMetAsn GlnTrpPro GlyValLys LeuArg ValThr GluGlyArg AspGluAsp GlyHisHis SerGluGlu SerLeu HisTyr GluGlyArg AlaValAsp IleThrThr SerAspArg AspArg AsnLys TyrGlyLeu LeuAlaArg LeuAlaVal GluAlaGly PheAsp Trp Val Tyr Tyr Glu Ser Lys Ala His Val His Cys Ser Val Lys Ser GluHis SerAlaAla AlaLysThr GlyGlyCys PheProAla GlyAla GlnVal ArgLeuGlu AsnGlyGlu ArgValAla LeuSerAla ValLys ProGly AspArgVal LeuAlaMet GlyGluAsp GlyThrPro ThrPhe SerAsp ValLeuIle PheLeuAsp ArgGluPro AsnArgLeu ArgAla PheGln ValIleGlu ThrGlnAsp ProProArg ArgLeuAla LeuThr ProAla HisLeuLeu PheIleAla AspAsnHis ThrGluPro AlaAla HisPhe ArgAlaThr PheAlaSer HisValGln ProGlyGln TyrVal LeuVal SerGlyVal ProGlyLeu GlnProAla ArgValAla AlaVal SerThr HisValAla LeuGlySer TyrAlaPro LeuThrArg HisGiy ThrLeu ValValGlu AspValVal AlaSerCys PheAlaAla ValAla AspHis HisLeuAla GlnLeuAla PheTrpPro LeuArgLeu PhePro SerLeu AlaTrpGly SerTrpThr ProSerGlu GlyValHis SerTyr ProGln MetLeuTyr ArgLeuGly ArgLeuLeu LeuGluGlu SerThr CTAACCACTG

PheHis ProLeuGly MetSerGly AlaGlySer ACTGCTGTGC
GTGGATCC

WO 99/20298 PCTIUS98l22227 (2) INFORMATION FOR SEQ ID N0:9:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1313 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..1319 (xi)SEQUENCE ID
DESCRIPTION: N0:4:
SEQ

MetLeu LeuLeuLeuAla ArgCysPheLeu ValIle LeuAla SerSer LeuLeu ValCysProGly LeuAlaCysGly ProGly ArgGly PheGly LysArg ArgHisProLys LysLeuThrPro LeuAla TyrLys GlnPhe IlePro AsnValAlaGlu LysThrLeuGly AlaSer GlyArg TyrGlu AAC

GlyLysIle ThrArgAsn SerGluArg PheLysGlu LeuThrPro Asn TyrAsnPro AspIleIle PheLysAsp GluGluAsn ThrGlyAla Asp ArgLeuMet ThrGlnArg CysLysAsp LysLeuAsn AlaLeuAla Ile SerValMet AsnGlnTrp ProGlyVal ArgLeuArg ValThrGlu Gly TrpAspGlu AspGlyHis HisSerGlu GluSerLeu HisTyrGlu Gly ArgAlaVal AspIleThr ThrSerAsp ArgAspArg SerLysTyr Gly MetLeuAla ArgLeuAla ValGluAla GlyPheAsp TrpValTyr Tyr _g_ GCA

GluSer LysAlaHis IleHisCys SerValLys AlaGluAsn SerVal AlaAla LysSerGly GlyCysPhe ProGlySer AlaThrVal HisLeu GAGCAG G.GCGGCACC AAGCTGGTG AAGGACTTA CGTCCCGGA GACCGC 672 GluGln GlyGlyThr LysLeuVal LysAspLeu ArgProGly AspArg ValLeu AlaAlaAsp AspGlnGly ArgLeuLeu TyrSerAsp PheLeu ThrPhe LeuAspArg AspGluGly AlaLysLysVal PheTyr ValIle GluThr LeuGluPro ArgGluArg LeuLeuLeuThr AlaAla HisLeu LeuPhe ValAlaPro HisAsnAsp SerGlyProThr ProGly ProSer AlaLeu PheAlaSer ArgValArg ProGlyGlnArg ValTyr ValVal AlaGlu ArgGlyGly AspArgArg LeuLeuProAla AlaVal HisSer ValThr LeuArgGlu GluGluAla GlyAlaTyrAla ProLeu ThrAla HisGly ThrIleLeu IleAsnArg ValLeuAlaSer CysTyr AlaVal IleGlu GluHisSer TrpAlaHis ArgAlaPheAla ProPhe ArgLeu AlaHis AlaLeuLeu AlaAlaLeu AlaProAlaArg ThrAsp GlyGly GlyGly GlySerIle ProAlaAla GlnSerAlaThr GluAla ArgGly AlaGlu ProThrAla GlyIleHis TrpTyrSerGln LeuLeu TyrHis IleGly ThrTrpLeu LeuAspSer GluThrMetHis ProLeu GlyMet WO 99/20298 pGT/US98/22227 Ala Val Lys Ser Ser (2) INFORMATION FOR SEQ ID N0:5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1256 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..1257 (xi)SEQUENCE
DESCRIPTION:
SEQ
ID
N0:5:

MetArg LeuLeu ThrArgVal LeuLeuVal SerLeuLeu ThrLeuSer LeuVal ValSer GlyLeuAla CysGlyPro GlyArgGly TyrGlyArg ArgArg HisPro LysLysLeu ThrProLeu AlaTyrLys GlnPheIle ProAsn ValAla GluLysThr LeuGlyAla SerGlyArg TyrGluGly LysIle ThrArg AsnSerGlu ArgPheLys GluLeuThr ProAsnTyr AsnProAspIle IlePheLys AspGluGlu AsnThrGlyAla AspArg LeuMetThrGln ArgCysLys AspLysLeu AsnSerLeuAla IleSer ValMetAsnHis TrpProGly ValLysLeu ArgValThrGlu GlyTrp AspGluAspGly HisHisPhe GluGluSer LeuHisTyrGlu GlyArg
- 10-AlaVal AspIleThr ThrSerAsp ArgAspLys SerLys TyrGlyThr LeuSer ArgLeuAla ValGluAla GlyPheAsp TrpVal TyrTyrGlu SerLys AlaHisIle HisCysSer ValLysAla GluAsn SerValAla AlaLys SerGlyGly CysPhePro GlySerAla Leu.ValSerLeuGln AspGly GlyGlnLys AlaValLys AspLeuAsn ProGly AspLysVal LeuAla AlaAspSer AlaGlyAsn LeuValPhe SerAsp PheIleMet PheThr AspArgAsp SerThrThr ArgArgVal PheTyr ValIleGlu GAA AAG

ThrGln GluProVal GluLysIle ThrLeuThr AlaAla HisLeuLeu PheVal LeuAspAsn SerThrGlu AspLeuHis ThrMet ThrAlaAla TyrAla SerSerVal ArgAlaGly GlnLysVal MetVal ValAspAsp SerGly GlnLeuLys SerValIle ValGlnArg IleTyr ThrGluGlu GlnArg GlySerPhe AlaProVal ThrAlaHis GlyThr IleValVal AspArg IleLeuAla SerCysTyr AlaValIle GluAsp GlnGlyLeu AlaHis LeuAlaPhe AlaProAla ArgLeuTyr TyrTyr ValSerSer PheLeu SerProLys ThrProAla ValGlyPro MetArg LeuTyrAsn ArgArg GlySerThr GlyThrPro GlySerCys HisGln MetGlyThr -ll-Trp Leu Leu Asp Ser Asn Met Leu His Pro Leu Gly Met Ser Val Asn Ser Ser (2) INFORMATION FOR SEQ ID N0:6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1425 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..1425 (xi)SEQUENCE
DESCRIPTION:
SEQ
ID
N0:6:

MetLeuLeuLeu AlaArgCys LeuLeuLeu ValLeuVal SerSer Leu LeuValCysSer GlyLeuAla CysGlyPro GlyArgGly PheGly Lys ArgArgHisPro LysLysLeu ThrProLeu AlaTyrLys GlnPhe Ile ProAsnValAla GluLysThr LeuGlyAla SerGlyArg TyrGlu Gly LysIleSerArg AsnSerGlu ArgPheLys GluLeuThr ProAsn Tyr AsnProAspIle IlePheLys AspGluGlu AsnThrGly AlaAsp Arg LeuMetThrGln ArgCysLys AspLysLeu AsnAlaLeu AlaIle Ser ValMetAsnGln TrpProGly ValLysLeu ArgValThr GluGly Trp AspGluAspGly HisHisSer GluGluSer LeuHisTyr GluGly Arg WO 99rt0298 PCT/US98/22227 ACC

AlaVal AspIleThr ThrSerAsp ArgAsp ArgSerLys TyrGlyMet LeuAla ArgLeuAla ValGluAla GlyPhe AspTrpVal TyrTyrGlu SerLys AlaHisIle HisCysSer Val-i~ysAlaGluAsn SerValAla AlaLys SerGlyGly CysPhePro GlySer AlaThrVal HisLeuGlu GlnGly GlyThrLys LeuValLys AspLeu SerProGly AspArgVal LeuAla AlaAspAsp GlnGlyArg LeuLeu TyrSerAsp PheLeuThr PheLeu AspArgAsp AspGlyAla LysLys ValPheTyr ValIleGlu ThrArg GluProArg GluArgLeu LeuLeu ThrAlaAla HisLeuLeu PheVal AlaProHis AsnAspSer AlaThr GlyGluPro GluAlaSer SerGly SerGlyPro ProSerGlyGly AlaLeuGly ProArg AlaLeu PheAla SerArgVal ArgProGlyGln ArgValTyr ValVal AlaGlu 305 310 ~ 315 320 ArgAsp GlyAspArg ArgLeuLeuPro AlaAlaVal HisSer ValThr LeuSer GluGluAla AlaGlyAlaTyr AlaProLeu ThrAla GlnGly ThrIle LeuIleAsn ArgValLeuAla SerCysTyr AlaVal IleGlu GluHis SerTrpAla HisArgAlaPhe AlaProPhe ArgLeu AlaHis AlaLeu LeuAlaAla LeuAlaProAla ArgThrAsp ArgGly GlyAsp 385 _ 390 395 400 SerGly GlyGly AspArgGlyGly GlyGly GlyArgVal AlaLeuThr AlaPro GlyAla AlaAspAlaPro GlyAla GlyAlaThr AlaGlyIle HisTrp TyrSer GlnLeuLeuTyr GlnIle GlyThrTrp LeuLeuAsp SerGlu AlaLeu HisProLeuGly MetAla ValLysSer SerXaaSer ArgGly AlaGly GlyGlyAlaArg GluGly Ala (2) INFORMATION FOR SEQ ID N0:7:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1622 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 51..1283 (xi) SEQUENCE DESCRIPTION: SEQ ID N0:7:

CCAGGAGACC ATG
TCGCCCGCCG
CTCCCCCGGG

Met Ser ProAla ArgLeu ArgProArgLeu HisPheCys LeuValLeu LeuLeu LeuLeu ValVal ProAlaAlaTrp GlyCysGly ProGlyArg ValVal GlySer ArgArg ArgProProArg LysLeuVal ProLeuAla TyrLys GlnPhe SerPro AsnValProGlu LysThrLeu GlyAlaSer GlyArg wo 99noz9s rc~r~rs9snza2~

Tyr Glu Gly Lys Ile Ala Arg Ser Ser Glu Arg Phe Lys Glu Leu Thr ProAsnTyrAsn ProAspIle IlePheLys AspGluGlu AsnThrGly AlaAspArgLeu MetThrGln ArgCysLys AspArgLeu AsnSerLeu AlaIleSerVal MetAsnGln TrpProGly ValLysLeu ArgValThr GluGlyTrpAsp GluAspGly HisHisSer GluGluSer LeuHisTyr GluGlyArgAla ValAspIle ThrThrSer AspArgAsp ArgAsnLys TyrGlyLeuLeu AlaArgLeu AlaValGlu AlaGlyPhe AspTrpVal 165 1?0 175 TyrTyrGluSer LysAlaHis ValHisCys SerValLys SerGluHis SerAlaAlaAla LysThrGly GlyCysPhe ProAlaGly AlaGlnVal ArgLeuGluSer GlyAlaArg ValAlaLeu SerAlaVal ArgProGly AspArgValLeu AlaMetGly GluAspGly SerProThr PheSerAsp ValLeuIlePhe LeuAspArg GluProHis ArgLeuArg AlaPheGln ValIle GluThr GlnAspProPro ArgArgLeu AlaLeuThr ProAla HisLeu LeuPhe ThrAlaAspAsn HisThrGlu ProAlaAla ArgPhe ArgAla ThrPhe AlaSerHisVal GlnProGly GlnTyrVal LeuVal AlaGly ValPro GlyLeuGlnPro AlaArgVal AlaAlaVal SerThr AAG

HisValAlaLeu GlyAlaTyr AlaProLeu ThrLysHis GlyThr Leu ValValGluAsp ValValAla SerCysPhe AlaAlaVal AlaAsp His HisLeuAlaGln LeuAlaPhe TrpPro ArgLeuPhe HisSer Leu ~eu AlaTrpGlySer TrpThrPro GlyGluGly ValHisTrp TyrPro Gln LeuLeuTyrArg LeuGlyArg LeuLeuLeu GluGluGly SerPhe His CCACCGCTGC

ProLeuGlyMet SerGlyAla GlySer (2) INFORMATION FOR SEQ ID N0:8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1191 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..1191 (xi) SEQUENCE DESCRIPTION: SEQ ID N0:8:

Met Ala Leu Leu Thr Asn Leu Leu Pro Leu Cys Cys Leu Ala Leu Leu Ala Leu Pro Ala Gln Ser Cys Gly Pro Gly Arg Gly Pro Val Gly Arg WO 99/Z0298 PCT/US98lZ2227 Arg Arg Tyr Ala Arg Lys Gln Leu Val Pro Leu Leu Tyr Lys Gln Phe Val Pro Gly Val Pro Glu Arg Thr Leu Gly Ala Ser Gly Pro Ala Glu GGG AGG GTG GCA .AGG GGC TCC GAG CGC TTC CGG GAC CTC GTG CCC AAC 290 Gly Arg Val Ala Arg Gly Ser Glu Arg~Phe Arg Asp Leu Val Pro Asn AAC

TyrAsnPro AspIleIle PheLysAspGlu GluAsnSer GlyAla Asp ArgLeuMet ThrGluArg CysLysGluArg ValAsnAla LeuAla Ile AlaValMet AsnMetTrp ProGlyValArg LeuArgVal ThrGlu Gly TrpAspGlu AspGlyHis HisAlaGlnAsp SerLeuHis TyrGlu Gly ArgAlaLeu AspIleThr ThrSerAspArg AspArgAsn LysTyr Gly LeuLeuAla ArgLeuAla ValGluAlaGly PheAspTrp ValTyr Tyr GluSerArg AsnHisVal HisValSerVal LysAlaAsp AsnSer Leu AlaValArg AlaGlyGly CysPheProGly AsnAlaThr ValArg Leu TrpSerGly GluArgLys GlyLeuArgGlu LeuHisArg GlyAsp Trp ValLeuAla AlaAspAla SerGlyArgVal ValProThr ProVal Leu LeuPheLeu AspArgAsp LeuGlnArgArg AlaSerPhe ValAla Val GluThrGlu TrpProPro ArgLysLeuLeu LeuThrPro TrpHis Leu ValPheAia AlaArgGly ProAlaProAla ProGlyAsp PheAla Pro _ 17_ Val PheAlaArg ArgLeuArg AlaGlyAsp SerValLeu AlaProGly Gly AspAlaLeu ArgProAla ArgValAla ArgValAla ArgGluGlu GCC GTGGGCGTG TTCGCGCCG CTCACC.GCG CACGGGACG CTGCTGGTG 1008 Ala ValGlyVal PheAlaPro LeuThrAla HisGlyThr LeuLeuVal Asn AspValLeu AlaSerCys TyrAlaVal LeuGluSer HisGlnTrp Ala HisRrgAla PheAlaPro LeuArgLeu LeuHisAla LeuGlyAla Leu LeuProGly GlyAlaVal GlnProThr GlyMetHis TrpTyrSer Arg LeuLeuTyr ArgLeuAla GluGluLeu LeuGly (2) INFORMATION FOR SEQ ID N0:9:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1251 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..1248 (xi) SEQUENCE DESCRIPTION: SEQ ID N0:9:

Met Asp Val Arg Leu His Leu Lys Gln Phe Ala Leu Leu Cys Phe Ile Ser Leu Leu Leu Thr Pro Cys Gly Leu Ala Cys Gly Pro Gly Arg Gly Tyr Gly Lys Arg Arg His Pro Lys Lys Leu Thr Pro Leu Ala Tyr Lys ACG

GlnPheIlePro AsnValAla GluLysThr LeuGlyAla SerGly Lys TyrGluGlyLys IleThrArg AsnSerGlu ArgPheLys GluLeu Ile ProAsnTyrAsn ProAspIle IlePhe'Lys AspGluGlu AsnThr Asn AlaAspArgLeu MetThrLys ArgCysLys AspLysLeu AsnSer Leu AlaIleSerVal MetAsnHis TrpProGly ValLysLeu ArgVal Thr GluGlyTrpAsp GluAspGly HisHisLeu GluGluSer LeuHis Tyr GluGlyArgAla ValAspIle ThrThrSer AspArgAsp LysSer Lys TyrGlyMetLeu SerArgLeu AlaValGlu AlaGlyPhe AspTrp Val Tyr~TyrGluSer LysAlaHis IleHisCys SerValLys AlaGlu Asn SerValAlaAla LysSerGly GlyCysPhe ProGlySer GlyThr Val ThrLeuGlyAsp GlyThrArg LysProIle LysAspLeu LysVal Gly AspArgValLeu AlaAlaAsp GluLysGly AsnValLeu IleSer Asp PheIleMetPhe IleAspHis AspProThr ThrArgArg GlnPhe Ile ValIleGluThr SerGluPro PheThrLys LeuThrLeu ThrAla Ala HisLeuValPhe ValGlyAsn SerSerAla AlaSerGly IleThr Ala Thr Phe Ala Ser Asn Val Lys Pro Gly Asp Thr Val Leu Val Trp Glu AGG

AspThr CysGluSer LeuLysSer ValThrVal LysArg IleTyrThr GluGlu HisGluGly SerPheAla ProValThr AlaHis GlyThrIle IleVal AspGlnVal LeuAlaSer Cys_ Ala ValIle GluAsnHis Tyr LysTrp AlaHisTrp AlaPheAla ProValArg LeuCys HisLysLeu MetThr TrpLeuPhe ProAlaArg GluSerAsn ValAsn PheGlnGlu AspGly IleHisTrp TyrSerAsn MetLeuPhe HisIle GlySerTrp LeuLeu AspArgAsp SerPheHis ProLeuGly IleLeu HisLeuSer (2) INFORMATION FOR SEQ ID NO:10:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 425 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID N0:10:
Met Val Glu Met Leu Leu Leu Thr Arg Ile Leu Leu Val Gly Phe Ile Cys Ala Leu Leu Val Ser Ser Gly Leu Thr Cys Gly Pro Gly Arg Gly Ile Gly Lys Arg Arg His Pro Lys Lys Leu Thr Pro Leu Ala Tyr Lys Gln Phe Ile Pro Asn Val Ala Glu Lys Thr Leu Gly Ala Ser Gly Arg Tyr Glu Gly Lys Ile Thr Arg Asn Ser Glu Arg Phe Lys Glu Leu Thr Pro Asn Tyr Asn Pro Asp Ile Ile Phe Lys Asp Glu Glu Asn Thr Gly Ala Asp Arg Leu Met Thr Gln Arg Cys Lys Asp Lys Leu Asn Ala Leu Ala Ile Ser Val Met Asn Gln Trp Pro Gly Val Lys Leu Arg Val Thr Glu Gly Trp Asp Glu Asp Gly His His Ser Glu Glu Ser Leu His Tyr Glu Gly Arg Ala Val Asp Ile Thr Thr Ser Asp Arg Asp Arg Ser Lys 145 150 .155 160 Tyr Gly Met Leu Ala Arg Leu Ala Val Glu Ala Gly Phe Asp Trp Val Tyr Tyr Glu Ser Lys Ala His Ile His Cys Ser Val Lys Ala Glu Asn Ser Val Ala Ala Lys Ser Gly Gly Cys Phe Pro Gly Ser Ala Thr Val His Leu Glu His Gly Gly Thr Lys Leu Val Lys Asp Leu Ser Pro Gly Asp Arg Val Leu Ala Ala Asp Ala Asp Gly Arg Leu Leu Tyr Ser Asp Phe Leu Thr Phe Leu Asp Arg Met Asp Ser Ser Arg Lys Leu Phe Tyr Val Ile Glu Thr Arg Gln Pro Arg Ala Arg Leu Leu Leu Thr Ala Ala His Leu Leu Phe Val Ala Pro Gln His Asn Gln Ser Glu Ala Thr Gly Ser Thr Ser Gly Gln Ala Leu Phe Ala Ser Asn Val Lys Pro Gly Gln Arg Val Tyr Val Leu Gly Glu Gly Gly Gln Gln Leu Leu Pro Ala Ser Val His Ser Val Ser Leu Arg Glu Glu Ala Ser Gly Ala Tyr Ala Pro Leu Thr Ala Gln Gly Thr Ile Leu Ile Asn Arg Val Leu Ala Ser Cys Tyr Ala Val Ile Glu Glu His Ser Trp Ala His Trp Ala Phe Ala Pro Phe Arg Leu Ala Gln Gly Leu Leu Ala Ala Leu Cys Pro Asp Gly Ala Ile Pro Thr Ala Ala Thr Thr Thr Thr Gly Ile His Trp Tyr Ser Arg Leu Leu Tyr Arg Ile Gly Ser Trp Val Leu Asp Gly Asp Ala Leu His Pro Leu Gly Met Val Ala Pro Ala Ser WO 99IZ029$ PCTNS98/22227 (2) INFORMATION FOR SEQ ID N0:11:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 396 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID N0:11:
Met Ala Leu Pro Ala Ser Leu Leu Pro Leu Cys Cys Leu A1-a Leu Leu Ala Leu Ser Ala Gln Ser Cys Gly Pro Gly Arg Gly Pro Val Gly Arg Arg Arg Tyr Val Arg Lys Gln Leu Val Pro Leu Leu Tyr Lys Gln Phe Val Pro Ser Met Pro Glu Arg Thr Leu Gly Ala Ser Gly Pro Ala Glu Gly Arg Val Thr Arg Gly Ser Glu Arg Phe Arg Asp Leu Val Pro Asn 65 ~ 70 75 80 Tyr Asn Pro Asp Ile Ile Phe Lys Asp Glu Glu Asn Ser Gly Ala Asp Arg Leu Met Thr Glu Arg Cys Lys Glu Arg Val Asn Ala Leu Ala Ile Ala Val Met Asn Met Trp Pro Gly Val Arg Leu Arg Val Thr Glu Gly Trp Asp Glu Asp Gly His His Ala Gln Asp Ser Leu His Tyr Glu Gly Arg Ala Leu Asp Ile Thr Thr Ser Asp Arg Asp Arg Asn Lys Tyr Gly Leu Leu Ala Arg Leu Ala Val Glu Ala Gly Phe Asp Trp Val Tyr Tyr Glu Ser Arg Asn His Ile His Val Ser Val Lys Ala Asp Asn Ser Leu Ala Val Arg Ala Gly Gly Cys Phe Pro Gly Asn Ala Thr Val Arg Leu Arg Ser Gly Glu Arg Lys Gly Leu Arg Glu Leu His Arg Gly Asp Trp Val Leu Ala Ala Asp Ala Ala Gly Arg Val Val Pro Thr Pro Val Leu Leu Phe Leu Asp Arg Asp Leu Gln Arg Arg Ala Ser Phe Val Ala Val Glu Thr Glu Arg Pro Pro Arg Lys Leu Leu Leu Thr Pro Trp His Leu Val Phe Ala Ala Arg Gly Pro Ala Pro Ala Pro Gly Asp Phe Ala Pro Val Phe Ala Arg Arg Leu Arg Ala Gly Asp Ser Val Leu Ala Pro Gly Gly Asp Ala Leu Gln Pro Ala Arg Val Ala Arg Val Ala Arg Glu Glu Ala Val Gly Val Phe Ala Pro Leu Thr Ala His Gly Thr Leu Leu Val Asn Asp Val Leu Ala Ser Cys Tyr Ala Val Leu Glu Ser His Gln Trp Ala His Arg Ala Phe Ala Pro Leu Arg Leu Leu His Ala Leu Gly Ala Leu Leu Pro Gly Gly Ala Val Gln Pro Thr Gly Met His Trp Tyr Ser Arg Leu Leu Tyr Arg Leu Ala Glu Glu Leu Met Gly (2) INFORMATION FOR SEQ ID N0:12:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 411 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID N0:12:
Met Ser Pro Ala Trp Leu Arg Pro Arg Leu Arg Phe Cys Leu Phe Leu Leu Leu Leu Leu Leu Val Pro Ala Ala Arg Gly Cys Gly Pro Gly Arg Val Val Gly Ser Arg Arg Arg Pro Pro Arg Lys Leu Val Pro Leu Ala Tyr Lys Gln Phe Ser Pro Asn Val Pro Glu Lys Thr Leu Gly Ala Ser Gly Arg Tyr Glu Gly Lys Ile Ala Arg Ser Ser Glu Arg Phe Lys Glu Leu Thr Pro Asn Tyr Asn Pro Asp Ile Ile Phe Lys Asp Glu Glu Asn Thr Gly Ala Asp Arg Leu Met Thr Gln Arg Cys Lys Asp Arg Leu Asn Ser Leu Ala Ile Ser Val Met Asn Gln Trp Pro Gly Val Lys Leu Arg Val Thr Glu Gly Arg Asp Glu Asp Gly His His Ser Glu Glu Ser Leu His Tyr Glu Gly Arg Ala Val Asp Ile Thr Thr Ser Asp Arg Asp Arg Asn Lys Tyr Gly Leu Leu Ala Arg Leu Ala Val Glu Ala Gly Phe Asp Trp Val Tyr Tyr Glu Ser Lys Ala His-Val His Cys Ser Val Lys Ser Glu His Ser Ala Ala Ala Lys Thr Gly Gly Cys Phe Pro Ala Gly Ala Gln Val Arg Leu Glu Asn Gly Glu Arg Val Ala Leu Ser Ala Val Lys Pro Gly Asp Arg Val Leu Ala Met Gly Glu Asp Gly Thr Pro Thr Phe Ser Asp Val Leu Ile Phe Leu Asp Arg Glu Pro Asn Arg Leu Arg Ala Phe Gln Val Ile Glu Thr Gln Asp Pro Pro Arg Arg Leu Ala Leu Thr Pro Ala His Leu Leu Phe Ile Ala Asp Asn His Thr Glu Pro Ala Ala His Phe Arg Ala Thr Phe Ala Ser His Val Gln Pro Gly Gln Tyr Val Leu Val Ser Gly Val Pro Gly Leu Gln Pro Ala Arg Val Ala Ala Val Ser Thr His Val Ala Leu Gly Ser Tyr Ala Pro Leu Thr Arg His Gly Thr Leu Val Val Glu Asp Val Val Ala Ser Cys Phe Ala Ala Val Ala Asp His His Leu Ala Gln Leu Ala Phe Trp Pro Leu Arg Leu Phe Pro Ser Leu Ala Trp Gly Ser Trp Thr Pro Ser Glu Gly Val His Ser Tyr Pro Gln Met Leu Tyr Arg Leu Gly Arg Leu Leu Leu Glu Glu Ser Thr Phe His Pro Leu Gly Met Ser Gly Ala Gly Ser (2) INFORMATION FOR SEQ ID N0:13:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 937 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear wo 99n0298 PCT/US98/22227 (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID N0:13:
Met Leu Leu Leu Leu Ala Arg Cys Phe Leu Val Ile Leu Ala Ser Ser Leu Leu Val Cys Pro Gly Leu Ala Cys Gly Pro Gly Arg Gly Phe Gly Lys Arg Arg His Pro Lys Lys Leu Thr Pro Leu Ala Tyr Lys Gln Phe Ile Pro Asn Val Ala Glu Lys Thr Leu Gly Ala Ser Gly Arg Tyr Glu Gly Lys Ile Thr Arg Asn Ser Glu Arg Phe Lys Glu Leu Thr Pro Asn Tyr Asn Pro Asp Ile Ile Phe Lys Asp Glu Glu Asn Thr Gly Ala Asp Arg Leu Met Thr Gln Arg Cys Lys Asp Lys Leu Asn Ala Leu Ala Ile Ser Val Met Asn Gln Trp Pro Gly Val Arg Leu Arg Val Thr Glu Gly Trp Asp Glu Asp Gly His His Ser Glu Glu Ser Leu His Tyr Glu Gly Arg Ala Val Asp Ile Thr Thr Ser Asp Arg Asp Arg Ser Lys Tyr Gly Met Leu Ala Arg Leu Ala Val Glu Ala Gly Phe Asp Trp Val Tyr Tyr Glu Ser Lys Ala His Ile His Cys Ser Val Lys Ala Glu Asn Ser Val Ala Ala Lys Ser Gly Gly Cys Phe Pro Gly Ser Ala Thr Val His Leu Glu Gln Gly Gly Thr Lys Leu Val Lys Asp Leu Arg Pro Gly Asp Arg Val Leu Ala Ala Asp Asp Gln Gly Arg Leu Leu Tyr Ser Asp Phe Leu Thr Phe Leu Asp Arg Asp Glu Gly Ala Lys Lys Val Phe Tyr Val Ile Glu Thr Leu Glu Pro Arg Glu Arg Leu Leu Leu Thr Ala Ala His Leu Leu Phe Val Ala Pro His Asn Asp Ser Gly Pro Thr Pro Gly Pro Ser Ala Leu Phe Ala Ser Arg Val Arg Pro Gly Gln Arg Val Tyr Val Val Ala Glu Arg Gly Gly Asp Arg Arg Leu Leu Pro Ala Ala Val His Ser Val Thr Leu Arg Glu Glu Glu Ala Gly Ala Tyr Ala Pro Leu Thr Ala His Gly Thr Ile Leu Ile Asn Arg Val Leu Ala Ser Cys Tyr Ala Val Ile Glu Glu His Ser Trp Ala His Arg-Ala Phe Ala Pro Phe Arg Leu Ala His Ala Leu Leu Ala Ala Leu Ala Pro Ala Arg Thr Asp Gly Gly Gly Gly Gly Ser Ile Pro Ala Ala Gln Ser Ala Thr Glu Ala Arg Gly Ala Glu Pro Thr Ala Gly Ile His Trp Tyr Ser Gln Leu Leu Tyr His Ile Gly Thr Trp Leu Leu Asp Ser Glu Thr Met His Pro Leu Gly Met Ala Val Lys Ser Ser (2) INFORMATION FOR SEQ ID N0:14:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 918 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID N0:14:
Met Arg Leu Leu Thr Arg Val Leu Leu Val Ser Leu Leu Thr Leu Ser Leu Val Val Ser Gly Leu Ala Cys Gly Pro Gly Arg Gly Tyr Gly Arg Arg Arg His Pro Lys Lys Leu Thr Pro Leu Ala Tyr Lys Gln Phe Ile Pro Asn Val Ala Glu Lys Thr Leu Gly Ala Ser Gly Arg Tyr Glu Gly Lys Ile Thr Arg Asn Ser Glu Arg Phe Lys Glu Leu Thr Pro Asn Tyr Asn Pro Asp Ile Ile Phe Lys Asp Glu Glu Asn Thr Gly Ala Asp Arg Leu Met Thr Gln Arg Cys Lys Asp Lys Leu Asn Ser Leu Ala Ile Ser Val Met Asn His Trp Pro Gly Val Lys Leu Arg Val Thr Glu Gly Trp Asp Glu Asp Gly His His Phe Glu Glu Ser Leu His Tyr Glu Gly Arg Ala Val Asp Ile Thr Thr Ser Asp Arg Asp Lys Ser Lys Tyr Gly Thr Leu Ser Arg Leu Ala Val Glu Ala Gly Phe Asp Trp Val Tyr Tyr Glu Ser Lys Ala His Ile His Cys Ser Val Lys Ala Glu Asn Ser Val AIa Ala Lys Ser Gly Gly Cys Phe Pro Gly Ser Ala Leu Val Ser Leu Gln Asp Gly Gly Gln Lys Ala Val Lys Asp Leu Asn Pro Gly Asp Lys Val Leu Ala Ala Asp Ser Ala Gly Asn Leu Val Phe Ser Asp Phe Ile Met Phe Thr Asp Arg Asp Ser Thr Thr Arg Arg Val Phe Tyr Val Ile Glu Thr Gln Glu Pro Val Glu Lys Ile Thr Leu Thr Ala Ala His Leu Leu Phe Val Leu Asp Asn Ser Thr Glu Asp Leu His Thr Met Thr Ala Ala Tyr Ala Ser Ser Val Arg Ala Gly Gln Lys Val Met Val Val Asp Asp Ser Gly Gln Leu Lys Ser Val Ile Val Gln Arg Ile Tyr Thr Glu Glu Gln Arg Gly Ser Phe Ala Pro Val Thr Ala His Gly Thr Ile Val Val Asp Arg Ile Leu Ala Ser Cys Tyr Ala Val Ile Glu Asp Gln Gly Leu Ala His Leu Ala Phe Ala Pro Ala Arg Leu Tyr Tyr Tyr Val Ser Ser Phe Leu Ser Pro Lys Thr Pro Ala Val Gly Pro Met Arg Leu Tyr Asn Arg Arg Gly Ser Thr Gly Thr Pro Gly Ser Cys His Gln Met Gly Thr Trp Leu Leu Asp Ser Asn Met Leu His Pro Leu Gly Met Ser Val Asn Ser Ser (2) INFORMATION FOR SEQ ID N0:15:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 975 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEø ID N0:15:
Met Leu Leu Leu Ala Arg Cys Leu Leu Leu Val Leu Val Ser Ser Leu Leu Val Cys Ser Gly Leu Ala Cys Gly Pro Gly Arg Gly Phe Gly Lys Arg Arg His Pro Lys Lys Leu Thr Pro Leu Ala Tyr Lys Gln Phe Ile Pro Asn Val Ala Glu Lys Thr Leu Gly Ala Ser Gly Arg Tyr Glu Gly Lys Ile Ser Arg Asn Ser Glu Arg Phe Lys Glu Leu Thr Pro Asn Tyr Asn Pro Asp Ile Ile Phe Lys Asp Glu Glu Asn Thr Gly Ala Asp Arg Leu Met Thr Gln Arg Cys Lys Asp Lys Leu Asn Ala Leu Ala Ile Ser Val Met Asn Gln Trp Pro Gly Val Lys Leu Arg Val Thr Glu Gly Trp Asp Glu Asp Gly His His Ser Glu Glu Ser Leu His Tyr Glu Gly Arg Ala Val Asp Ile Thr Thr Ser Asp Arg Asp Arg Ser Lys Tyr Gly Met Leu Ala Arg Leu Ala Val Glu Ala Gly Phe Asp Trp Val Tyr Tyr Glu Ser Lys Ala His Ile His Cys Ser Val Lys Ala Glu Asn Ser Val Ala Ala Lys Ser Gly Gly Cys Phe Pro Gly Ser Ala Thr Val His Leu Glu Gln Gly Gly Thr Lys Leu Val Lys Asp Leu Ser Pro Gly Asp Arg Val Leu Ala Ala Asp Asp Gln Gly Arg Leu Leu Tyr Ser Asp Phe Leu Thr Phe Leu Asp Arg Asp Asp Gly Ala Lys Lys Val Phe Tyr Val Ile Glu Thr Arg Glu Pro Arg Glu Arg Leu Leu Leu Thr Ala Ala His Leu Leu Phe Val Ala Pro His Asn Asp Ser Ala Thr Gly Glu Pro Glu Ala Ser Ser Gly Ser Gly Pro Pro Ser Gly Gly Ala Leu Gly Pro Arg Ala Leu Phe Ala Ser Arg Val Arg Pro Gly Gln Arg Val Tyr Val Val Ala Glu Arg Asp Gly Asp Arg Arg Leu Leu Pro-Ala Ala Val His Ser Val Thr Leu Ser Glu Glu Ala Ala Gly Ala Tyr Ala Pro Leu Thr Ala Gln Gly Thr Ile Leu Ile Asn Arg Val Leu Ala Ser Cys Tyr Ala Val Ile Glu Glu His Ser Trp Ala His Arg Ala Phe Ala Pro Phe Arg Leu Ala His Ala Leu Leu Ala Ala Leu Ala Pro Ala Arg Thr Asp Arg Gly Gly Asp Ser Gly Gly Gly Asp Arg Gly Gly Gly Giy Gly Arg Val Ala Leu Thr Ala Pro Gly Ala Ala Asp Ala Pro Gly Ala Gly Ala Thr Ala Gly Ile His Trp Tyr Ser Gln Leu Leu Tyr Gln Ile Gly Thr Trp Leu Leu Asp Ser Glu Ala Leu His Pro Leu Gly Met Ala Val Lys Ser Ser Xaa Ser Arg Gly Ala Gly Gly Gly Ala Arg Glu Gly Ala (2) INFORMATION FOR SEQ ID N0:16:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 411 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID N0:16:
Met Ser Pro Ala Arg Leu Arg Pro Arg Leu His Phe Cys Leu Val Leu Leu Leu Leu Leu Val Val Pro Ala Ala Trp Gly Cys Gly Pro Gly Arg Val Val Gly Ser Arg Arg Arg Pro Pro Arg Lys Leu Val Pro Leu Ala Tyr Lys Gln Phe Ser Pro Asn Val Pro Glu Lys Thr Leu Gly Ala Ser Gly Arg Tyr Glu Gly Lys Ile Ala Arg Ser Ser Glu Arg Phe Lys Glu Leu Thr Pro Asn Tyr Asn Pro Asp Ile Ile Phe Lys Asp Glu Glu Asn Thr Gly Ala Asp Arg Leu Met Thr Gln Arg Cys Lys Asp Arg Leu Asn Ser Leu Ala Ile Ser Val Met Asn Gln-Trp Pro Gly Val Lys Leu Arg Val Thr Glu Gly Trp Asp Glu Asp Gly His His Ser Glu Glu Ser Leu His Tyr Glu Gly Arg Ala Val Asp Ile Thr Thr Ser Asp Arg Asp Arg Asn Lys Tyr Gly Leu Leu Ala Arg Leu Ala Val Glu Ala Gly Phe Asp Trp Val Tyr Tyr Glu Ser Lys Ala His Val His Cys Ser Val Lys Ser Glu His Ser Ala Ala Ala Lys Thr Gly Gly Cys Phe Pro Ala Gly Ala Gln Val Arg Leu Glu Ser Gly Ala Arg Val Ala Leu Ser Ala Val Arg Pro Gly Asp Arg Val Leu Ala Met Gly Glu Asp Gly Ser Pro Thr Phe Ser Asp Val Leu Ile Phe Leu Asp Arg Glu Pro His Arg Leu Arg Ala Phe Gln Val Ile Glu Thr Gln Asp Pro Pro Arg Arg Leu Ala Leu Thr Pro Ala His Leu Leu Phe Thr Ala Asp Asn His Thr Glu Pro Ala Ala Arg Phe Arg Ala Thr Phe Ala Ser His Val Gln Pro Gly Gln Tyr Val Leu Val Ala Gly Val Pro Gly Leu Gln Pro Ala Arg Val Ala Ala Val Ser Thr His Val Ala Leu Gly Ala Tyr Ala Pro Leu Thr Lys His Gly Thr Leu Val Val Glu Asp Val Val Ala Ser Cys Phe Ala Ala Val Ala Asp His His Leu Ala Gln Leu Ala Phe Trp Pro Leu Arg Leu Phe His Ser Leu Ala Trp Gly Ser Trp Thr Pro Gly Glu Gly Val His Trp Tyr Pro Gln Leu Leu Tyr Arg Leu Gly Arg Leu Leu Leu Glu Glu Gly Ser Phe His Pro Leu Gly Met Ser Gly Ala Gly Ser (2) INFORMATION FOR SEQ ID N0:17:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 396 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID N0:1?:
Met Ala Leu Leu Thr Asn Leu Leu Pro Leu Cys Cys Leu Ala Leu Leu Ala Leu Pro Ala Gln Ser Cys Gly Pro Gly Arg Gly Pro Val Gly Arg Arg Arg Tyr Ala Arg Lys Gln Leu Val Pro Leu Leu Tyr Lys Gln Phe Val Pro Gly Val Pro Glu Arg Thr Leu Gly Ala Ser Gly Pro Ala Glu Gly Arg Val Ala Arg Gly Ser Glu Arg Phe Arg Asp Leu Val Pro Asn Tyr Asn Pro Asp Ile Ile Phe Lys Asp Glu Glu Asn Ser Gly Ala Asp Arg Leu Met Thr Glu Arg Cys Lys Glu Arg Val Asn Ala Leu Ala Ile Ala Val Met Asn Met Trp Pro Gly Val Arg Leu Arg Val Thr Glu Gly Trp Asp Glu Asp Gly His His Ala Gln Asp Ser Leu His Tyr Glu Gly Arg Ala Leu Asp Ile Thr Thr Ser Asp Arg Asp Arg Asn Lys Tyr Gly Leu Leu Ala Arg Leu Ala Val Glu Ala Gly Phe Asp Trp Val Tyr Tyr Glu Ser Arg Asn His Val His Val Ser Val Lys Ala Asp Asn Ser Leu Ala Val Arg Ala Gly Gly Cys Phe Pro Gly Asn Ala Thr Val Arg Leu Trp Ser Gly Glu Arg Lys Gly Leu Arg Glu Leu His Arg Gly Asp Trp Val Leu Ala Ala Asp Ala Ser Gly Arg Val Val Pro Thr Pro Val Leu Leu Phe Leu Asp Arg Asp Leu Gln Arg Arg Ala Ser Phe Val Ala Val wo 99noz98 rc r~s9smn~

Glu Thr Glu Trp Pro Pro Arg Lys Leu Leu Leu Thr Pro Trp His Leu Val Phe Ala Ala Arg Gly Pro Ala Pro Ala Pro Gly Asp Phe Ala Pro Val Phe Ala Arg Arg Leu Arg Ala Gly Asp Ser Val Leu Ala Pro Gly Gly Asp Ala Leu Arg Pro Ala Arg Val-~11a Arg Val Ala Arg Glu Glu Ala Val Gly Val Phe Ala Pro Leu Thr Ala His Gly Thr Leu Leu Val Asn Asp Val Leu Ala Ser Cys Tyr Ala Val Leu Glu Ser His Gln Trp Ala His Arg Ala Phe Ala Pro Leu Arg Leu Leu His Ala Leu Gly Ala Leu Leu Pro Gly Gly Ala Val Gln Pro Thr Gly Met His Trp Tyr Ser Arg Leu Leu Tyr Arg Leu Ala Glu Glu Leu Leu Gly (2) INFORMATION FOR SEQ ID N0:18:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 416 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID N0:18:
Met Asp Val Arg Leu His Leu Lys Gln Phe Ala Leu Leu Cys Phe Ile Ser Leu Leu Leu Thr Pro Cys Gly Leu Ala Cys Gly Pro Gly Arg Gly Tyr Gly Lys Arg Arg His Pro Lys Lys Leu Thr Pro Leu Ala Tyr Lys Gln Phe Ile Pro Asn Val Ala Glu Lys Thr Leu Gly Ala Ser Gly Lys Tyr Glu Gly Lys Ile Thr Arg Asn Ser Glu Arg Phe Lys Glu Leu Ile Pro Asn Tyr Asn Pro Asp Ile Ile Phe Lys Asp Glu Glu Asn Thr Asn Ala Asp Arg Leu Met Thr Lys Arg Cys Lys Asp Lys Leu Asn Ser Leu Ala Ile Ser Val Met Asn His Trp Pro Gly Val Lys Leu Arg Val Thr WO 99/20298 PGT/US98n2227 Glu Gly Trp Asp Glu Asp Gly His His Leu Glu Glu Ser Leu His Tyr Glu Gly Arg Ala Val Asp Ile Thr Thr Ser Asp Arg Asp Lys Ser Lys Tyr Gly Met Leu Ser Arg Leu Ala Val Glu Ala Gly Phe Asp Trp Val 165 x.70 175 Tyr Tyr Glu Ser Lys Ala His Ile His Cys Ser Val Lys Ala Glu Asn Ser Val Ala Ala Lys Ser Gly Gly Cys Phe Pro Gly Ser Gly Thr Val Thr Leu Gly Asp Gly Thr Arg Lys Pro Ile Lys Asp Leu Lys Val Gly Asp Arg Val Leu Ala Ala Asp Glu Lys Gly Asn Val Leu Ile Ser Asp Phe Ile Met Phe Ile Asp His Asp Pro Thr Thr Arg Arg Gln Phe Ile Val Ile Glu Thr Ser Glu Pro Phe Thr Lys Leu Thr Leu Thr Ala Ala His Leu Val Phe Val Gly Asn Ser Ser Ala Ala Ser Gly Ile Thr Ala Thr Phe Ala Ser Asn Val Lys Pro Gly Asp Thr Val Leu Val Trp Glu Asp Thr Cys Glu Ser Leu Lys Ser Val Thr Val Lys Arg Ile Tyr Thr Glu Glu His Glu Gly Ser Phe Ala Pro Val Thr Ala His Gly Thr Ile Ile Val Asp Gln Val Leu Ala Ser Cys Tyr Ala Val Ile Glu Asn His Lys Trp Ala His Trp Ala Phe Ala Pro Val Arg Leu Cys His Lys Leu Met Thr Trp Leu Phe Pro Ala Arg Glu Ser Asn Val Asn Phe Gln Glu Asp Gly Ile His Trp Tyr Ser Asn Met Leu Phe His Ile Gly Ser Trp Leu Leu Asp Arg Asp Ser Phe His Pro Leu Gly Ile Leu His Leu Ser (2) INFORMATION FOR SEQ ID N0:19:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1416 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: both (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..1413 (xi)SEQUENCE ID
DESCRIPTION: N0:19:
SEQ

MetAsp AsnHisSer SerValPro TrpAlaSer AlaAlaSer ValThr CysLeu SerLeuGly CysGlnMet ProGlnPhe GlnPheGln PheGln LeuGln IleArgSer GluLeuHis LeuArgLys ProAlaArg ArgThr GlnThr MetArgHis IleAlaHis ThrGlnArg CysLeuSer ArgLeu ThrSer LeuValAla LeuLeuLeu IleValLeuPro MetVal PheSer ProAla HisSerCys GlyProGly ArgGlyLeuGly ArgHis ArgAla ArgAsn LeuTyrPro LeuValLeu LysGlnThrIle ProAsn LeuSer GluTyr ThrAsnSer AlaSerGly ProLeuGluGly ValIle ArgArg AspSer ProLysPhe LysAspLeu ValProAsnTyr AsnArg AspIle LeuPhe ArgAspGlu GluGlyThr GlyAlaAspGly LeuMet SerLys ArgCys LysGluLys LeuAsnVal LeuAlaTyrSer ValMet AsnGlu TrpPro GlyIleArg LeuLeuVal ThrGluSerTrp AspGlu AspTyr HisHis GlyGlnGlu SerLeuHis TyrGluGlyArg AlaVal ThrIle Ala Thr Ser Asp Arg Asp Gln Ser Lys Tyr Gly Met Leu Ala Arg Leu Ala Val Glu Ala Gly Phe Asp Trp Val Ser Tyr Val Ser Arg Arg His IleTyr CysSerVal LysSerAsp SerSerIle SerSerHis ValHis GlyCys PheThrPro GluSerThr AlaLeuLeu GluSerGly ValArg LysPro LeuGlyGlu LeuSerIle GlyAspArg ValLeuSer MetThr AlaAsn GlyGlnAla ValTyrSer GluValIle LeuPheMet AspArg AsnLeu GluGlnMet GlnAsnPhe ValGlnLeu HisThrAsp GlyGly AlaVal LeuThrVal ThrProAla HisLeuVal SerValTrp GlnPro GluSer GlnLysLeu ThrPheVal PheAlaHis ArgIleGlu GluLys AsnGln ValLeuVal ArgAspVal GluThrGly GluLeuArg ProGln ArgVal ValLysLeu GlySerVal ArgSerLys GlyValVal AlaPro LeuThr ArgGluGly ThrIleVal ValAsnSer ValAlaAla SerCys TyrAla ValIleAsn SerGlnSer LeuAlaHis TrpGlyLeu AlaPro MetArg LeuLeuSer ThrLeuGlu AlaTrpLeu ProAlaLys GluGln LeuHis SerSerPro LysValVal SerSerAla GlnGlnGln AsnGly Ile His Trp Tyr Ala Asn Ala Leu Tyr Lys Val Lys Asp Tyr Val Leu Pro Gln Ser Trp Arg His Asp (2) INFORMATION FOR SEQ ID N0:20:
(i} SEQUENCE CHARACTERISTICS:
(A) LENGTH: 971 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID N0:20:
Met Asp Asn His Ser Ser Val Pro Trp Ala Ser Ala Ala Ser Val Thr Cys Leu Ser Leu Gly Cys Gln Met Pro Gln Phe Gln Phe Gln Phe Gln Leu Gln Ile Arg Ser Glu Leu His Leu Arg Lys Pro Ala Arg Arg Thr Gln Thr Met Arg His Ile Ala His Thr Gln Arg Cys Leu Sex Arg Leu Thr Ser Leu Val Ala Leu Leu Leu Ile Val Leu Pro Met 'lal Phe Ser Pro Ala His Ser Cys Gly Pro Gly Arg Gly Leu Gly Arg His Arg Ala Arg Asn Leu Tyr Pro Leu Val Leu Lys Gln Thr Ile Pro Asn Leu Ser Glu Tyr Thr Asn Ser Ala Ser Gly Pro Leu Glu Gly Val Ile Arg Arg Asp Ser Pro Lys Phe Lys Asp Leu Val Pro Asn Tyr Asn Arg Asp Ile Leu Phe Arg Asp Glu Glu Gly Thr~Gly Ala Asp Gly Leu Met Ser Lys Arg Cys Lys Glu Lys Leu Asn Val Leu Ala Tyr Ser Val Met Asn Glu Trp Pro Gly Ile Arg Leu Leu Val Thr Glu Ser Trp Asp Glu Asp Tyr His His Gly Gln Glu Ser Leu His Tyr Glu Gly Arg Ala Val Thr Ile Ala Thr Ser Asp Arg Asp Gln Ser Lys Tyr Gly Met Leu Ala Arg Leu wo 99noz9s rcnus9snZZZ~

Ala Val Glu Ala Gly Phe Asp Trp Val Ser Tyr Val Ser Arg Arg His Ile Tyr Cys Ser Val Lys Ser Asp Ser Ser Ile Ser Ser His Val His Gly Cys Phe Thr Pro Glu Ser Thr Ala Leu Leu Glu Ser Gly Val Arg Lys Pro Leu Gly Glu Leu Ser Ile Gly-Asp Arg Val Leu Ser Met Thr Ala Asn Gly Gln Ala Val Tyr Ser Glu Val Ile Leu Phe Met Asp Arg Asn Leu Glu Gln Met Gln Asn Phe Val Gln Leu His Thr Asp Gly Gly Ala Val Leu Thr Val Thr Pro Ala His Leu Val Ser Val Trp Gln Pro Glu Ser Gln Lys Leu Thr Phe Val Phe Ala His Arg Ile Glu Glu Lys Asn Gln Val Leu Val Arg Asp Val Glu Thr Gly Glu Leu Arg Pro Gln Arg Val Val Lys Leu Gly Ser Val Arg Ser Lys Gly Val Val AIa Pro Leu Thr Arg Glu Gly Thr Ile Val Val Asn Ser Val Ala Ala Ser Cys Tyr Ala Val Ile Asn Ser Gln Ser Leu Ala His Trp Gly Leu Ala Pro Met Arg Leu Leu Ser Thr Leu Glu Ala Trp Leu Pro Ala Lys Glu Gln 920 425 ~ 430 Leu His Ser Ser Pro Lys Val Val Ser Ser Ala Gln Gln Gln Asn Gly Ile His Trp Tyr Ala Asn Ala Leu Tyr Lys Val Lys Asp Tyr Val Leu Pro Gln Ser Trp Arg His Asp (2) INFORMATION FOR SEQ ID N0:21:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 221 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide (v) FRAGMENT TYPE: internal (xi) SEQUENCE DESCRIPTION: SEQ ID N0:21:

wo r~noz9s rcT~s9snzzz~

Cys Gly Pro Gly Arg Gly Xaa Gly Xaa Arg Arg His Pro Lys Lys Leu Thr Pro Leu Ala Tyr Lys Gln Phe Ile Pro Asn Val Ala Glu Lys Thr Leu Gly Ala Ser Gly Arg Tyr Glu Gly Lys Ile Xaa Arg Asn Ser Glu Arg Phe Lys Glu Leu Thr Pro Asri Tyr Asn Pro Asp Ile Ile Phe Lys Asp Glu Glu Asn Thr Gly Ala Asp Arg Leu Met Thr Gln Arg Cys Lys Asp Lys Leu Asn Xaa Leu Ala Ile Ser Val Met Asn Xaa Trp Pro Gly Val Xaa Leu Arg Val Thr Glu Gly Trp Asp Glu Asp Gly His His Xaa Glu Glu Ser Leu His Tyr Glu Gly Arg Ala Val Asp Ile Thr Thr Ser Asp Arg Asp Xaa Ser Lys Tyr Gly Xaa Leu Xaa Arg Leu Ala Val Glu Ala Gly Phe Asp Trp Val Tyr Tyr Glu Ser Lys Ala His Ile His Cys Ser Val Lys Ala Glu Asn Ser Val Ala Ala Lys Ser Gly Gly Cys Phe Pro Gly Ser Ala Xaa Val Xaa Leu Xaa Xaa Gly Gly Xaa Lys Xaa Val Lys Asp Leu Xaa Pro Gly Asp Xaa Val Leu Ala Ala Asp Xaa Xaa Gly Xaa Leu Xaa Xaa Ser Asp Phe Xaa Xaa Phe Xaa Asp Arg (2) INFORMATION FOR SEQ ID N0:22:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 167 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide (v) FRAGMENT TYPE: internal (xi) SEQUENCE DESCRIPTION: SEQ ID N0:22:
Cys Gly Pro Gly Arg Gly Xaa Xaa Xaa Arg Arg Xaa Xaa Xaa Pro Lys Xaa Leu Xaa Pro Leu Xaa Tyr Lys Gln Phe Xaa Pro Xaa Xaa Xaa Glu Xaa Thr Leu Gly Ala Ser Gly Xaa Xaa Glu Gly Xaa Xaa Xaa Arg Xaa Ser Glu Arg Phe Xaa Xaa Leu Thr Pro Asn Tyr Asn Pro Asp Ile Ile Phe Lys Asp Glu Glu Asn Xaa Gly Ala Asp Arg Leu Met Thr Xaa Arg Cys Lys Xaa Xaa Xaa Asn Xaa Leu Ala Ile Ser Val Met Asn Xaa Trp Pro Gly Val Xaa Leu Arg Val Thr Glu Gly Xaa Asp Glu Asp Gly His His Xaa Xaa Xaa Ser Leu His Tyr Glu Gly Arg Ala Xaa Asp Ile Thr Thr Ser Asp Arg Asp Xaa Xaa Lys Tyr Gly Xaa Leu Xaa Arg Leu Ala Val Glu Ala Gly Phe Asp Trp Val Tyr Tyr Glu Sex Xaa Xaa His Xaa His Xaa Ser Val Lys Xaa Xaa

Claims (68)

We claim:
1. A method for modulating the growth state of an epithelial cell comprising ectopically contacting the epithelial cell with an amount of an agent effective to alter the rate of proliferation of the epithelial cell, wherein the agent is selected from the group consisting of a hedgehog therapeutic and a ptc therapeutic.
2. A method for modulating the growth state of an epithelial tissue comprising ectopically contacting the tissue with an amount of an agent effective to alter the rate of proliferation of the epithelial cells in the tissue, wherein the agent is selected from the group consisting of a hedgehog therapeutic and a ptc therapeutic.
3. A method for inducing the formation of skin, comprising treating the skin with an amount of an agent effective to induce the formation of new skin tissue, wherein the agent is selected from the group consisting of a hedgehog therapeutic and a ptc therapeutic.
4. A method for inducing growth of hair on an animal, comprising treating the animal with an amount of an agent effective to induce growth of hair, wherein the agent is selected from the group consisting of a hedgehog therapeutic and a ptc therapeutic which induce proliferation of hair follicle keratinocytes.
5. The method of claim 1 or 2, wherein the agent increases the rate of proliferation of epithelial cells.
6. The method of claim 1 or 2, wherein the agent decreases the rate of proliferation of epithelial cells.
7. The method of claim 1 or 2, wherein the epithelial cell is a cutaneous epithelial cell.
8. The method of claim 7, wherein the epithelial cell is a dermal keratinocyte.
9. The method of claim 7, wherein the epithelial cell is a mucosal epithelial cell.
10. The method of claim 7, wherein the epithelial cell is an epithelial stem cell.
11. The method of claim 7, wherein the epithelial cell is a hair follicle stem cell.
12. The method of claim 1, wherein the cell is in culture, and the agent is provided as a cell culture additive.
13. The method of claim 1, wherein the cell is treated in an animal and the agent is administered to the animal as a therapeutic composition.
14. The method of claim 1, wherein the epithelial tissue is in tissue culture, and the agent is provided as a tissue culture additive.
15. The method of claim 1, wherein the epithelial tissue is treated in an animal and the agent is administered to the animal as a therapeutic composition.
16. The method of claim 3, 4, 13 or 15, wherein the agent is applied topically.
17. The method of claim 1, 2, 3 or 4, wherein the agent is a hedgehog therapeutic.
18. The method of claim 17, wherein the hedgehog therapeutic is a polypeptide including a hedgehog polypeptide sequence of at least a bioactive extracellular portion of a hedgehog protein.
19. The method of claim 18, wherein the polypeptide includes at least 50 amino acids residues of an N-terminal half of the hedgehog protein.
20. The method of claim 18, wherein the polypeptide includes at least 100 amino acids of an extracellular domain of the hedgehog protein.
21. The method of claim 18, wherein the polypeptide includes at least a portion of the hedgehog protein corresponding to a l9kd fragment of an extracellular domain of the hedgehog protein.
22. The method of claim 18, wherein the hedgehog protein is encoded by a gene of a vertebrate organism.
23. The method of claim 18, wherein the polypeptide includes a hedgehog polypeptide sequence represented in the general formula of SEQ ID No. 21.
24. The method of claim 18, wherein the polypeptide includes a hedgehog polypeptide sequence represented in the general formula of SEQ ID No. 22.
25. The method of claim 18, wherein the hedgehog protein is encoded by a human hedgehog gene.
26. The method of claim 18, wherein the hedgehog polypeptide sequence is at least 60 percent identical to an amino acid sequence of a hedgehog protein selected from the group consisting of SEQ ID No:9, SEQ ID No:10, SEQ ID No:11, SEQ ID No:12, SEQ
ID No:13, SEQ ID No:14, SEQ ID No:15 and SEQ ID No:16.
27. The method of claim 26, wherein the hedgehog polypeptide sequence is at least 75 percent identical to an amino acid sequence of a hedgehog protein selected from the group consisting of SEQ ID No:9, SEQ ID No:10, SEQ ID No:11, SEQ ID No:12, SEQ
ID No:13, SEQ ID No:14, SEQ ID No:15 and SEQ ID No:16.
28. The method of claim 26, wherein the hedgehog polypeptide sequence is at least 85 percent identical to an amino acid sequence of a hedgehog protein selected from the group consisting of SEQ ID No:9, SEQ ID No:10, SEQ ID No:11, SEQ ID No:12, SEQ
ID No:13, SEQ ID No:14, SEQ ID No:15 and SEQ ID No:16.
29. The method of claim 26, wherein the hedgehog polypeptide sequence is encodable by a nucleotide sequence which hybridizes under stringent conditions to a sequence selected from the group consisting of SEQ ID No:1, SEQ ID No:2, SEQ ID No:3, SEQ ID
No:4, SEQ ID No:5, SEQ ID No:6, SEQ ID No:7 and SEQ ID No:8.
30. The method of claim 23, wherein the hedgehog polypeptide sequence is an amino acid sequence of a hedgehog protein selected from the group consisting of SEQ ID
No:9, SEQ ID No:10, SEQ ID No:11, SEQ ID No:12, SEQ ID No:13, SEQ ID No:14, SEQ ID
No:15 and SEQ ID No:16.
31. The method of claim 18, wherein the hedgehog polypeptide sequence is an amino acid sequence of a Sonic hedgehog protein.
32. The method of claim 18, wherein the hedgehog polypeptide sequence is an amino acid sequence of an Indian hedgehog protein.
33. The method of claim 18, wherein the hedgehog polypeptide sequence is an amino acid sequence of a Desert hedgehog protein.
34. The method of claim 18, wherein the hedgehog polypeptide sequence includes an amino acid sequence corresponding approximately to residues 24-193 of SEQ ID No:15.
35. The method of claim 18, wherein the polypeptide is purified to at least 80% by dry weight.
36. The method of claim 18, wherein the polypeptide is a recombinantly produced polypeptide.
37. The method of claim 18, wherein the polypeptide is a chemically synthesized polypeptide.
38. The method of claim 17, wherein the hedgehog therapeutic is a peptidomimetic of a hedgehog polypeptide sequence.
39. The method of claim 1, 2, 3 or 4, wherein the agent is a ptc therapeutic.
40. The method of claim 39, wherein the ptc therapeutic is a small organic molecule which binds to a patched protein and derepresses patched-mediated inhibition of mitosis.
41. The method of claims 39, wherein the ptc therapeutic binds to patched and mimics hedgehog-mediated patched signal transduction.
42. The method of claim 41, wherein the ptc therapeutic is a small organic molecule.
43. The method of claim 41, wherein the binding of the ptc therapeutic to patched results in upregulation of patched and/or gli expression.
44. The method of claim 39, wherein the ptc therapeutic is a small organic molecule which interacts with epithelial cells to induce hedgehog-mediated patched signal transduction.
45. The method of claim 44, wherein the ptc therapeutic induces hedgehog-mediated patched signal transduction by altering the localization, protein-protein binding and/or enzymatic activity of an intracellular protein involved in a patched signal pathway.
46. The method of claim 39, wherein the ptc therapeutic alters the level of expression of a hedgehog protein, a patched protein or a protein involved in the intracellular signal transduction pathway of patched.
47. The method of claim 46, wherein the ptc therapeutic is an antisense construct which inhibits the expression of a protein which is involved in the signal transduction pathway of patched and the expression of which antagonizes hedgehog-mediated signals.
48. The method of claim 47, wherein the antisense construct is an oligonucleotide of about 20-30 nucleotides in length and having a GC content of at least 50 percent.
49. The method of claim 48, wherein the antisense oligonucleotide is selected from the group consisting of-. 5'-GTCCTGGCGCCGCCGCCGCCGTCGCC;
5'-TTCCGATGACCGGCCTTTCGCGGTGA; and 5'-GTGCACGGAAAGGTGCAGGCCACACT
50. The method of claims 44, wherein the ptc therapeutic is a small organic molecule which binds to patched and regulates patched dependent gene expression.
51. The method of claim 44, wherein the ptc therapeutic is an inhibitor of protein kinase A
(PKA).
52. The method of claim 51, wherein the PKA inhibitor is a 5-isoquinolinesulfonamide
53. The method of claim 51, wherein the PKA inhibitor is represented in the general formula:
wherein, R1 and R2 each can independently represent hydrogen, and as valence and stability permit a lower alkyl, a lower alkenyl, a lower alkynyl, a carbonyl (such as a carboxyl, an ester, a formate, or a ketone), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an amino, an acylamino, an amido, a cyano, a nitro, an azido, a sulfate, a sulfonate, a sulfonamido, -(CH2)m-R8, -(CH2)m OH, -(CH2)m O-lower alkyl, -(CH2)m O-lower alkenyl, -.
(CH2)n-O-(CH2)m-R8, -(CH2)m-SH, -(CH2)m S-lower alkyl, -(CH2)n; S-lower alkenyl, -(CH2)n-S-(CH2)m-R8, or R1 and R2 taken together with N form a heterocycle (substituted or unsubstituted);
R3 is absent or represents one or more substitutions to the isoquinoline ring such as a lower alkyl, a lower alkenyl, a lower alkynyl, a carbonyl (such as a carboxyl, an ester, a formate, or a ketone), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an amino, an acylamino, an amido, a cyano, a nitro, an azido, a sulfate, a sulfonate, a sulfonamido, -(CH2)m-R8, -(CH2)m-OH, -(CH2)m-O-lower alkyl, -(CH2)m-O-lower alkenyl, -(CH2)n-O-(CH2)m R8, -(CH2)m-SH, -(CH2)m-S-lower alkyl, -(CH2)m S-lower alkenyl, -(CH2)n-S-(CH2)m-R8;
R8 represents a substituted or unsubstituted aryl, aralkyl, cycloalkyl, cycloalkenyl, or heterocycle; and n and m are independently for each occurrence zero or an integer in the range of 1 to 6.
54. The method of claim 51, wherein the PKA inhibitor is cyclic AMP analog.
55. The method of claim 51, wherein the PKA inhibitor is selected from the group consisting of N-[2-((p-bromocinnamyl)amino)ethyl]-5-isoquinolinesulfonamide, 1-(5-isoquinoline-sulfonyl)-2-methylpiperazine, KT5720, 8-bromo-cAMP, dibutyryl-cAMP and PKA
Heat Stable Inhibitor isoform .alpha..
56. A method for promoting the proliferation of skin epithelial cells, comprising ectopically contacting the cells with a polypeptide in an amount effective to increase the rate of proliferation of the epithelial cells, which polypeptide comprises an amino acid sequence including at least a bioactive portion of the N-terminal half of a hedgehog protein.
57. The method of claim 56, which method is used as part of a treatment to control a wound healing process.
58. The method of claim 56, wherein the treatment is selected from a group consisting of burn treatment, skin regeneration, skin grafting, pressure sore treatment, dermal ulcer treatment, post surgery scar reduction and treatment of ulcerative colitis.
59. The method of claim 56, which method is used as part of a treatment of alopecia.
60. A method for inhibiting the proliferation of skin epithelial cells, comprising ectopically contacting the cells with agent, in an amount effective to decrease the rate of proliferation of the epithelial cells, which inhibits binding of a hedgehog protein to patched.
61. The method of claim 60, wherein the epithelial cells are hair follicle cells.
62. The method of claim 61, which method inhibits hair growth.
63. A preparation of a polypeptide comprising a hedgehog polypeptide sequence including a bioactive fragment of a hedgehog protein, which polypeptide is formulated for topical application to epithelial tissue.
64. The preparation of claim 63, wherein the polypeptide is formulated for topical application to skin.
65. The preparation of claim 63, wherein the polypeptide includes at least 50 amino acids residues of an N-terminal half of the hedgehog protein.
66. The preparation of claim 63, wherein the polypeptide includes at least 100 amino acids of an extracellular domain of the hedgehog protein.
67. The preparation of claim 63, wherein the polypeptide includes at least a portion of the hedgehog protein corresponding to a 19kd fragment of an extracellular domain of the hedgehog protein.
68. The preparation of claim 63, wherein the hedgehog protein is encoded by a gene of a vertebrate organism.
CA2307322A 1997-10-20 1998-10-20 Regulation of epithelial tissue by hedgehog-like polypeptides, and formulations and uses related thereto Expired - Lifetime CA2307322C (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US95555297A 1997-10-20 1997-10-20
US08/955,552 1997-10-20
US09/151,999 US6639051B2 (en) 1997-10-20 1998-09-11 Regulation of epithelial tissue by hedgehog-like polypeptides, and formulations and uses related thereto
US09/151,999 1998-09-11
PCT/US1998/022227 WO1999020298A1 (en) 1997-10-20 1998-10-20 Regulation of epithelial tissue by hedgehog-like polypeptides, and formulations and uses related thereto

Publications (2)

Publication Number Publication Date
CA2307322A1 true CA2307322A1 (en) 1999-04-29
CA2307322C CA2307322C (en) 2017-02-28

Family

ID=26849158

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2307322A Expired - Lifetime CA2307322C (en) 1997-10-20 1998-10-20 Regulation of epithelial tissue by hedgehog-like polypeptides, and formulations and uses related thereto

Country Status (16)

Country Link
US (5) US6639051B2 (en)
EP (1) EP1028741B1 (en)
JP (1) JP2001520202A (en)
KR (1) KR20010031266A (en)
CN (1) CN1306436A (en)
AT (1) ATE257391T1 (en)
AU (1) AU760939B2 (en)
CA (1) CA2307322C (en)
CY (1) CY2016003I1 (en)
DE (1) DE69821021T2 (en)
DK (1) DK1028741T3 (en)
ES (1) ES2214744T3 (en)
HK (1) HK1030365A1 (en)
LU (1) LU92977I2 (en)
PT (1) PT1028741E (en)
WO (1) WO1999020298A1 (en)

Families Citing this family (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6639051B2 (en) * 1997-10-20 2003-10-28 Curis, Inc. Regulation of epithelial tissue by hedgehog-like polypeptides, and formulations and uses related thereto
TW570805B (en) 1998-09-01 2004-01-11 Hoffmann La Roche Water-soluble pharmaceutical composition in an ionic complex
EP1109569A2 (en) 1998-09-11 2001-06-27 President And Fellows Of Harvard College Regulation of lung tissue by hedgehog-like polypeptides, and formulations and uses related thereto
WO2000018428A2 (en) * 1998-09-11 2000-04-06 Biogen, Inc. Hedgehog and patched antagonists for inhibiting cell and tissue growth and differentiation and uses therefor
US6159950A (en) * 1998-10-16 2000-12-12 Cornell Research Foundation, Inc. Method of modulating hair growth
EP1133519B1 (en) * 1998-11-02 2008-08-13 Curis, Inc. Functional antagonists of hedgehog activity
US6884427B1 (en) * 1999-02-08 2005-04-26 Aderans Research Institute, Inc. Filamentary means for introducing agents into tissue of a living host
IL146842A0 (en) * 1999-06-01 2002-07-25 Biogen Inc Polymer conjugates of hedgehog proteins and uses
JP2003501395A (en) * 1999-06-08 2003-01-14 ローランティス・リミテッド Therapeutic use
US20070021493A1 (en) 1999-09-16 2007-01-25 Curis, Inc. Mediators of hedgehog signaling pathways, compositions and uses related thereto
CA2385736C (en) * 1999-09-16 2011-11-15 Curis, Inc. Mediators of hedgehog signaling pathways, compositions and uses related thereto
US6903073B2 (en) * 1999-12-10 2005-06-07 The General Hospital Corporation Methods to stimulate insulin production by pancreatic beta-cells
EP1254369B1 (en) * 2000-02-08 2010-10-06 Sangamo BioSciences, Inc. Cells for drug discovery
US7115653B2 (en) 2000-03-30 2006-10-03 Curis, Inc. Small organic molecule regulators of cell proliferation
US8852937B2 (en) 2000-03-30 2014-10-07 Curis, Inc. Small organic molecule regulators of cell proliferation
AU2001257012A1 (en) * 2000-04-14 2001-10-30 Genencor International, Inc. Methods for selective targeting
US20030022819A1 (en) * 2000-06-16 2003-01-30 Ling Leona E. Angiogenesis-modulating compositions and uses
AU2001279209B2 (en) * 2000-08-08 2006-08-24 Aderans Research Institute, Inc. Scaffolds for tissue engineered hair
US7708998B2 (en) 2000-10-13 2010-05-04 Curis, Inc. Methods of inhibiting unwanted cell proliferation using hedgehog antagonists
CA2425356C (en) 2000-10-13 2015-10-06 Curis, Inc. Hedgehog antagonists, methods and uses related thereto
US20040068284A1 (en) * 2002-01-29 2004-04-08 Barrows Thomas H. Method for stimulating hair growth and kit for carrying out said method
US20060009409A1 (en) 2002-02-01 2006-01-12 Woolf Tod M Double-stranded oligonucleotides
CA2475003A1 (en) 2002-02-01 2003-08-07 Sequitur, Inc. Double-stranded oligonucleotides
EP1476175A2 (en) * 2002-02-20 2004-11-17 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Hedgehog-related prophylaxis, therapy and diagnosis of gi tract carcinogenesis
JP2003267966A (en) * 2002-03-13 2003-09-25 Pola Chem Ind Inc Flavan derivative, skin fibroblast proliferation inhibitor and external preparation for skin
WO2005042740A1 (en) * 2003-10-31 2005-05-12 Renomedix Institute Inc. Method of enhancing hematopoiesis-supporting ability of immortalized human bone marrow stroma cell by transferring indian hedgehog gene
DK1730280T3 (en) * 2004-03-26 2019-02-04 Curis Inc RNA interference modulators for hedgehog signaling and applications thereof
US7597885B2 (en) 2004-03-26 2009-10-06 Aderans Research Institute, Inc. Tissue engineered biomimetic hair follicle graft
DE102004025881A1 (en) * 2004-05-19 2006-01-05 Beiersdorf Ag Oligoribonucleotides for influencing hair growth
AR050212A1 (en) * 2004-08-13 2006-10-04 Aderans Res Inst Inc ORGANOGENESIS FROM DISCELLED CELLS
US20070281040A1 (en) * 2004-09-30 2007-12-06 The University Of Chicago Combination therapy of hedgehog inhibitors, radiation and chemotherapeutic agents
US20060105950A1 (en) * 2004-10-25 2006-05-18 Caritas St. Elizabeth's Medical Center Of Boston, Inc. Morphogen compositions and use thereof to treat wounds
US20060293228A1 (en) * 2005-06-24 2006-12-28 Bhatnagar Rajendra S Therapeutic compositions and methods using transforming growth factor-beta mimics
US20060293227A1 (en) * 2005-06-24 2006-12-28 Bhatnagar Rajendra S Cosmetic compositions and methods using transforming growth factor-beta mimics
CA2626199A1 (en) * 2005-10-17 2007-04-26 Aderans Research Institute, Inc. Method of delivering hair follicle progenitor cells to the skin
WO2007062386A2 (en) 2005-11-22 2007-05-31 Aderans Research Institute, Inc. Hair follicle graft from tissue engineered skin
WO2007064945A2 (en) 2005-12-01 2007-06-07 Pronai Therapeutics, Inc. Cancer therapies and pharmaceutical compositions used therein
GB2439377A (en) * 2006-06-09 2007-12-27 Pasteur Institut Korea Methods of identifying modulators of and modulating GPCR cellular distribution and medical uses thereof
EP2046753A2 (en) 2006-07-06 2009-04-15 Brystol-Myers Squibb Company Pyridone/hydroxypyridine 11-beta hydroxysteroid dehydrogenase type i inhibitors
WO2008057468A1 (en) 2006-11-02 2008-05-15 Curis, Inc. Small organic molecule regulators of cell proliferation
US7922688B2 (en) 2007-01-08 2011-04-12 Restoration Robotics, Inc. Automated delivery of a therapeutic or cosmetic substance to cutaneous, subcutaneous and intramuscular tissue regions
US7985537B2 (en) 2007-06-12 2011-07-26 Aderans Research Institute, Inc. Methods for determining the hair follicle inductive properties of a composition
EP2617415B1 (en) * 2010-09-16 2016-04-20 Catholic Kwandong University Industry Foundation Use of a compound for inducing differentiation of mesenchymal stem cells into cartilage cells
WO2014071406A1 (en) 2012-11-05 2014-05-08 Pronai Therapeutics, Inc. Methods of using biomarkers for the treatment of cancer by modulation of bcl2|expression
WO2015073691A1 (en) 2013-11-14 2015-05-21 The Board Of Trustees Of The Leland Stanford Junior University Methods for treating cancer by activation of bmp signaling
WO2015075166A1 (en) 2013-11-22 2015-05-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for treatment of a bacterial infection
US9765332B2 (en) 2014-01-29 2017-09-19 Inserm (Institut National De La Sante Et De La Recherche Medicale) Oligonucleotides and methods for inhibiting or reducing bacterial biofilms
CA3043886A1 (en) * 2016-11-16 2018-05-24 Aivita Biomedical, Inc. Use of cell membrane-bound signaling factors
CN110392686A (en) 2016-12-30 2019-10-29 频率治疗公司 1H- pyrrole-2,5-diones compound and making be used to induction it is dry/method of ancestral's sertoli cell self-renewing
WO2019048898A1 (en) 2017-09-05 2019-03-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Pharmaceutical compositions for the treatment of endothelial dysfunction
US11261445B2 (en) 2017-10-17 2022-03-01 Inserm (Institut National De La Sante Et De La Recherche Medicale) Combination treatment for cystic fibrosis
WO2019099850A1 (en) * 2017-11-16 2019-05-23 Aivita Biomedical, Inc. Use of cell membrane-bound signaling factors
KR102176832B1 (en) * 2017-11-29 2020-11-10 주식회사 파이안바이오테크놀로지 A microparticle comprising hedgehog protein and biocompatible material and a composition for preventing or treating hair loss comprising the same
WO2021005223A1 (en) 2019-07-10 2021-01-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of epilepsy
WO2021074657A1 (en) 2019-10-17 2021-04-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Combination treatment for cystic fibrosis
WO2021099394A1 (en) 2019-11-19 2021-05-27 INSERM (Institut National de la Santé et de la Recherche Médicale) Antisense oligonucleotides and their use for the treatment of cancer
BR112023000428A2 (en) 2020-07-10 2023-03-14 Inst Nat Sante Rech Med METHODS AND COMPOSITIONS TO TREAT EPILEPSY
JPWO2022244502A1 (en) * 2021-05-21 2022-11-24
WO2023152369A1 (en) 2022-02-14 2023-08-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Nucleic acid mir-9 inhibitor for the treatment of cystic fibrosis
WO2024017990A1 (en) 2022-07-21 2024-01-25 Institut National de la Santé et de la Recherche Médicale Methods and compositions for treating chronic pain disorders

Family Cites Families (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US456687A (en) * 1891-07-28 William bader
US4456687A (en) 1978-11-16 1984-06-26 President And Fellows Of Harvard College Agents for promoting growth of epithelial cells
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
DE3786714T2 (en) 1986-06-16 1994-03-10 Daiichi Seiyaku Co Therapeutic agent for skin ulcers.
JPH0745383B2 (en) 1986-09-30 1995-05-17 第一製薬株式会社 Hair formulation containing cyclic adenosine derivative
AU1362488A (en) * 1987-01-28 1988-08-24 Peter H. Proctor Topical composition and method for stimulating hair growth with stable free radicals
AU4308689A (en) 1988-09-02 1990-04-02 Protein Engineering Corporation Generation and selection of recombinant varied binding proteins
DE3942114C2 (en) 1988-12-26 1997-09-04 Hiroyoshi Hidaka Compounds with an effect for a uniform vascular muscle relaxation and pharmaceutical composition containing them
JPH02273610A (en) * 1989-04-17 1990-11-08 Chugai Pharmaceut Co Ltd Trichogenous promoter
US5789650A (en) * 1990-08-29 1998-08-04 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
JPH04305528A (en) * 1991-02-22 1992-10-28 Dai Ichi Seiyaku Co Ltd Proliferation stimulant for keratinocyte and dermal fibroblast
CA2105300C (en) 1991-03-01 2008-12-23 Robert C. Ladner Process for the development of binding mini-proteins
US5223408A (en) 1991-07-11 1993-06-29 Genentech, Inc. Method for making variant secreted proteins with altered properties
AU3920693A (en) * 1992-03-18 1993-10-21 General Hospital Corporation, The Four novel receptors of the TGF-beta receptor family
JP2893029B2 (en) 1992-08-10 1999-05-17 旭化成工業株式会社 Cardioprotectant
US6333168B1 (en) 1993-05-20 2001-12-25 The Trustees Of Columbia University In The City Of New York Cloning, expression and uses of dorsalin-1
US5519035A (en) 1993-07-02 1996-05-21 Cornell Research Foundation, Inc. Treatment of stroke or in anticipation of the occurrence of brain ischemia
US6610656B1 (en) * 1993-12-30 2003-08-26 President And Fellows Of Harvard College Method of promoting chondrocyte differentiation with hedgehog related polypeptides
US5789543A (en) 1993-12-30 1998-08-04 President And Fellows Of Harvard College Vertebrate embryonic pattern-inducing proteins and uses related thereto
EP0741783B1 (en) 1993-12-30 2009-03-18 President And Fellows Of Harvard College Vertebrate embryonic pattern-inducing hedgehog-like proteins
US6261786B1 (en) * 1993-12-30 2001-07-17 Imperial Cancer Res. Technology Screening assays for hedgehog agonists and antagonists
US6884775B1 (en) * 1993-12-30 2005-04-26 President And Fellows Of Harvard College Methods and compositions for regulating skeletogenic formation
US7060450B1 (en) * 1993-12-30 2006-06-13 President And Fellows Of Harvard College Screening assays for agonists and antagonists of the hedgehog signaling pathway
US6165747A (en) * 1993-12-30 2000-12-26 President & Fellows Of Harvard College Nucleic acids encoding hedgehog proteins
US6384192B1 (en) * 1993-12-30 2002-05-07 President & Fellows Of Harvard College Vertebrate embryonic pattern-inducing proteins
US20030186357A1 (en) * 1993-12-30 2003-10-02 Philip W. Ingham Vertebrate embryonic pattern-inducing proteins, and uses related thereto
WO1995023223A1 (en) 1994-02-25 1995-08-31 The Trustees Of Columbia University In The City Of New York Dna encoding the vertebrate homolog of hedgehog, vhh-1, expressed by the notochord, and uses thereof
US5585087A (en) 1994-06-08 1996-12-17 President And Fellows Of Harvard College Assay for identifying extracellular signaling proteins
US5554608A (en) 1994-09-28 1996-09-10 Ahluwalia; Gurpreet S. Inhibition of hair growth
AU4001195A (en) 1994-10-07 1996-05-02 Board Of Trustees Of The Leland Stanford Junior University Patched genes and their use
US6946257B1 (en) * 1994-10-07 2005-09-20 Regents Of The University Of California Patched genes and uses related thereto
US6429354B1 (en) * 1994-10-07 2002-08-06 The Board Of Trustees Of The Leland Stanford Junior University Patched genes and uses related thereto
US6027882A (en) * 1994-10-07 2000-02-22 The Regents Of The University Of California Patched genes and their use for diagnostics
US6281332B1 (en) 1994-12-02 2001-08-28 The Johns Hopkins University School Of Medicine Hedgehog-derived polypeptides
US5643915A (en) 1995-06-06 1997-07-01 Andrulis Pharmaceuticals Corp. Treatment of ischemia/reperfusion injury with thalidomide alone or in combination with other therapies
AU7115996A (en) 1995-09-21 1997-04-09 Trustees Of Columbia University In The City Of New York, The Uses of bone morphogenetic proteins
IL128999A0 (en) 1996-09-20 2000-02-17 Harvard College Hedgehog interacting proteins and uses related thereto
US5990281A (en) 1996-09-30 1999-11-23 Genentech, Inc. Vertebrate smoothened proteins
IL129295A0 (en) 1996-10-07 2000-02-17 Univ Johns Hopkins Med Novel hedgehog-derived polypeptides
US5759811A (en) 1996-11-13 1998-06-02 The Regents Of The University Of California Mutant human hedgehog gene
JPH10194987A (en) 1997-01-14 1998-07-28 Youtai Iwamoto Composition for ossification and chondrogenesis
US6713065B2 (en) * 1997-02-10 2004-03-30 President And Fellows Of Harvard College Methods of using hedgehog proteins to modulate hematopoiesis and vascular growth
JPH114695A (en) 1997-04-25 1999-01-12 Hayashibara Biochem Lab Inc Hedgehog protein
US6051400A (en) 1997-05-23 2000-04-18 Smithkline Beecham Plc Patched-2 polypeptides and polynucleotides
AU8173098A (en) 1997-06-27 1999-01-19 Ontogeny, Inc. Neuroprotective methods and reagents
US6218144B1 (en) 1997-06-30 2001-04-17 The Board Of Trustees Of The Leland Stanford Junior University Costal2 genes and their uses
AU8380898A (en) 1997-07-01 1999-01-25 Ingham, Philip W Vertebrate smoothened gene, gene products, and uses related thereto
US7144997B2 (en) 1997-07-24 2006-12-05 Curis, Inc. Vertebrate embryonic patterning-inducing proteins, compositions and uses related therto
EP1009424A2 (en) 1997-08-29 2000-06-21 Ontogeny, Inc. Regulation of muscle tissues by hedgehog-like polypeptides, and formulations and uses related thereto
US6639051B2 (en) * 1997-10-20 2003-10-28 Curis, Inc. Regulation of epithelial tissue by hedgehog-like polypeptides, and formulations and uses related thereto
EP1109569A2 (en) * 1998-09-11 2001-06-27 President And Fellows Of Harvard College Regulation of lung tissue by hedgehog-like polypeptides, and formulations and uses related thereto
WO2000018428A2 (en) * 1998-09-11 2000-04-06 Biogen, Inc. Hedgehog and patched antagonists for inhibiting cell and tissue growth and differentiation and uses therefor
US6159950A (en) * 1998-10-16 2000-12-12 Cornell Research Foundation, Inc. Method of modulating hair growth
US7498304B2 (en) * 2000-06-16 2009-03-03 Curis, Inc. Angiogenesis-modulating compositions and uses
US20030022819A1 (en) * 2000-06-16 2003-01-30 Ling Leona E. Angiogenesis-modulating compositions and uses
US7708998B2 (en) * 2000-10-13 2010-05-04 Curis, Inc. Methods of inhibiting unwanted cell proliferation using hedgehog antagonists
CA2425356C (en) * 2000-10-13 2015-10-06 Curis, Inc. Hedgehog antagonists, methods and uses related thereto

Also Published As

Publication number Publication date
PT1028741E (en) 2004-05-31
CA2307322C (en) 2017-02-28
DE69821021D1 (en) 2004-02-12
CN1306436A (en) 2001-08-01
US20020151460A1 (en) 2002-10-17
US20090118187A1 (en) 2009-05-07
HK1030365A1 (en) 2001-05-04
EP1028741B1 (en) 2004-01-07
US20030104970A1 (en) 2003-06-05
DE69821021T2 (en) 2004-10-21
WO1999020298A1 (en) 1999-04-29
US6639051B2 (en) 2003-10-28
US20070110698A1 (en) 2007-05-17
JP2001520202A (en) 2001-10-30
AU1108999A (en) 1999-05-10
US20120238500A1 (en) 2012-09-20
KR20010031266A (en) 2001-04-16
CY2016003I2 (en) 2019-07-10
AU760939B2 (en) 2003-05-22
EP1028741A1 (en) 2000-08-23
ATE257391T1 (en) 2004-01-15
DK1028741T3 (en) 2004-05-17
LU92977I2 (en) 2016-04-11
ES2214744T3 (en) 2004-09-16
CY2016003I1 (en) 2019-07-10

Similar Documents

Publication Publication Date Title
AU760939B2 (en) Regulation of epithelial tissue by hedgehog-like polypeptides, and formulations and uses related thereto
US20110124580A1 (en) Regulation of lung tissue by hedgehog-like polypeptides, and formulations and uses related thereto
US6767888B1 (en) Neuroprotective methods and reagents
EP1067917B1 (en) Method of treating dopaminergic and gaba-nergic disorders
CA2302780A1 (en) Regulation of muscle tissues by hedgehog-like polypeptides, and formulations and uses related thereto
EP1646395B1 (en) Methods and compositions for regulating lymphocyte activity
US6951839B1 (en) Methods and compositions for regulating lymphocyte activity
EP1135411B1 (en) Methods and compositions for treating disorders involving excitotoxicity
AU2003200161B2 (en) Method of treating dopaminergic and gaba-nergic disorders

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20181022