METHODS AND COMPOSITIONS FOR PROMOTING WOUND HEALING

Provided herein are compositions and methods that inhibit expression of Adam12 gene products, such as ADAM12 mRNA and/or ADAM12 polypeptides, as a therapeutic approach for the treatment of, or promotion of healing of, wounds.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS REFERENCE TO RELATED APPLICATIONS

This application is a divisional of application Ser. No. 12/047,106, filed Mar. 12, 2008, which claims the benefit under 35 U.S.C. §119(e) of U.S. Provisional Application No. 60/918,367, filed Mar. 15, 2007, the contents of all of which are incorporated herein in their entireties by reference thereto.

STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH

The United States Government may have certain rights in this invention pursuant to Grant No. GM64750 and NR008029 awarded by the National Institutes of Health.

BACKGROUND

Chronic wounds represent a major health burden world-wide, and a significant drain on medical resources. For example, recent studies have calculated the annual cost of chronic wounds to the NHS in the United Kingdom to be approximately £1 billion (Harding, “The Future of Wound Healing,” In: Leaper et al., Wounds: Biology and Management,” Oxford University Press, 1998 at page 191). Foot ulceration, the most common complication of diabetes requiring hospitalization, is estimated to cost the UK approximately £17 million per year (Currie et al., 1998, Diabetes Care 21:42-48) and the US approximately $150 million per year (Reiber et al., 1992, Diabetes Care 15(suppl. 1):29-31). The total annual cost of diabetic peripheral neuropathy and its complications in the US was estimated to be between $4.6 and $13.7 billion (Gordois, et al., 2003, Diabetes Care 26:1790-1795). Equally, or even more costly are managing and treating pressure ulcers (bed sores)—$55 billion annually—and venous leg ulcers—estimated to be 2.5 to 3 billion US dollars, with a loss of 2 million work days per year (McGuckin, et al., 2001, Adv Skin Wound Care 14(1):33-36; and see decubitus.org/cost/cost.html).

These sums show only the economic costs of managing and treating chronic wounds. They do not reflect the frustration, economic loss and impaired quality of life experienced by patients with chronic wounds. Indeed, deep foot ulcers may be accompanied by cellulites or osteomyelitis, and a severely infected or nonhealing foot ulcer may require amputation of the toe, foot or leg (Gordois, et al., 2003, Diabetes Care 26:1790-1795).

Recent understandings of the biology underlying wound healing have led to the development of several new and experimental treatments. These include: the use of dressings designed to promote wound healing; topical administration of growth factors such as transforming growth factor beta, which is currently being studied for use in venous ulcers, platelet derived growth factor, which has been licensed for the treatment of diabetic foot ulcers (becaplermin, Regranex) and has shown some promise in treating pressure ulcers (Rees, 1997, Wound Repair Regen. 7:141-147), granulocyte colony stimulating factor, which has been evaluated for the treatment of infected diabetic foot ulcers (Gough, 1997, Lancet 350:833-859), fibroblast growth factor, which has been assessed for treating pressure ulcers (Robson et al., 1992, Ann. Surg. 216(40):401-408), and epidermal growth factor, which has been used to treat venous leg ulcers (Falanga et al., 1992, J. Dermatol. Surg. Oncol. 18:604-606); autologous skin grafts, which have been successfully used to treat venous leg ulcers (Mauro, T. M., 1992, West J. Med. 156(2): 191); and bioengineered skin equivalents such as Alloderm (a dermal matrix without immunogenic cells), Integra (a combination of dermal fibroblasts and bovine collagen), Dermagraft (non-immunogenic neonatal fibroblasts cultured in a polyglactin mesh) and Apilgraf (contains both epidermal and dermal components). All of these bioengineered skin grafts have been used to treat burns. Dermagraft has also been used to treat diabetic foot ulcers (Gentzkow et al., 1996, Diabetes Care 19:350-354). Apilgraf has been used to treat diabetic foot ulcers (Veves et al., 2001, Diabetes Care 24:290-295) and venous leg ulcers (Falanga et al., 1998, Arch. Dermatol. 134:293-300).

Unfortunately, clinical results with these new technologies have not been as dramatic as hoped. Accordingly, there remains a need in the art for new strategies and compositions for the treatment of wounds generally, and chronic wounds specifically.

SUMMARY

ADAM12 (a disintegrin and metalloprotease 12) is a membrane anchored metalloprotease that has been implicated in the cleavage of insulin-like growth factor-binding protein-3 in blood during pregnancy (Shi et al., 2000, J. Biol. Chem. 275:18574-18580) and in the progression of several diseases, including human tumorgenic cancer (Iba et al., 1999, Am. J. Pathol. 154:1489-1501), prostate cancer (Peduto et al., 2006, Oncogene, 25:5462-5466), hematological malignancies (Wu et al., 1997, Biochem. Biophys. Res. Commun. 235:437-442) and cardiac hypertrophy (Asakura et al., 2002, Nat. Med. 8:35-40). As will be discussed in more detail below, compelling evidence reported herein suggests that in addition to these biological functions, ADAM12 is an important mediator of wound healing and pathogenesis. For example, it has been discovered that ADAM12 is up-regulated in chronic wounds. Specifically, it has been discovered that the amount of ADAM12 expressed in the non-healing edge of chronic skin ulcers is approximately at least five-fold greater than that observed in normal skin. In addition, studies with skin explants from ADAM12 knock-out mice revealed a statistically significant increase in the migration of keratinocytes as compared to skin explants from wild-type mice in samples that were either treated or untreated with EGF. While not intending to be bound by any theory of operation, these studies suggest that increased expression of ADAM12 in chronic wounds impairs wound healing through the inhibition of keratinocyte migration, and that compounds capable of inhibiting the activity or expression of ADAM12 would provide a new and powerful means of promoting healing of wounds.

Accordingly, in one aspect, the present disclosure provides methods of inhibiting ADAM12 activity or expression as a therapeutic approach towards the treatment and/or promotion of healing of wounds. In some embodiments, the methods comprise administering to a wound an amount of an ADAM12 inhibitory compound effective to treat and/or promote healing of the wound. The ADAM12 inhibitory compound can inhibit an activity of an ADAM12 polypeptide per se or expression of one or more Adam12 gene products. As specific examples, ADAM12 inhibitory compounds that inhibit expression of an Adam12 gene product can inhibit transcription of a gene encoding an ADAM12, thereby blocking synthesis of ADAM12 mRNA, or translation of mRNA encoding an ADAM12, thereby blocking synthesis of ADAM12 polypeptides. Compounds useful for inhibiting an activity of an ADAM12 polypeptide per se include, by way of example and not limitation, small molecules, antibodies, polypeptides, polynucleotides, or other ADAM12 antagonists. Compounds useful for inhibiting expression of Adam 12 gene products include, by way of example and not limitation, antisense, siRNA and miRNA oligonucleotides.

The ADAM12 inhibitory compounds can be administered by any means suitable for the delivery of the specific ADAM12 inhibitory compound being utilized. The treatment can be applied systemically, or alternatively, the ADAM12 inhibitory compound can be applied locally directly to the wound, for example by way of injection directly into the wounded tissue or by way of a topical formulation containing the ADAM12 inhibitory compound. Non-limiting examples of suitable local or topical formulations include sprays, liquids, gels, creams, ointments, transdermal patches and wound dressings. Embodiments in which the wound being treated is an acute surgical wound, or a wound requiring suturing, the ADAM12 inhibitory compound can be administered with surgical sutures or staples that include, or that are coated with, the ADAM12 inhibitory compound.

The ADAM12 inhibitory compound may be administered alone, in combination with one or more additional ADAM12 inhibitory compounds and/or in combination with one or more additional therapeutic agents. Some examples of such therapeutic agents that can be used in combination therapies include, but are not limited to, Regranex, becaplermin, granulocyte colony stimulating factor, fibroblast growth factor, epidermal growth factor, agents that promote angiogenesis, and antibiotics.

In embodiments in which the ADAM12 inhibitory compound is a biological molecule, such as, for example, a peptide, polypeptide, oligonucleotide or polynucleotide, the inhibitory compound per se can be administered to the wound or patient, or alternatively, nucleic acids encoding the ADAM12 inhibitory compound can be administered to the wound or patient. In this latter embodiment, the cellular machinery of the body or wounded tissue is used as a means to express in vivo the ADAM12 inhibitory compound.

It is expected that virtually any type of wound can be treated with ADAM12 inhibitory compound(s) as described herein to promote healing. Of particular significance are wounds occurring in tissues that over-express ADAM12 as compared to unwounded tissue of the same type. Such over expression has been observed in human skin tissue of chronic wounds such as diabetic foot ulcers, venous leg ulcers and pressure ulcers, and is expected to be observed in other tissue types, including but not limited to mouth ulcers, gingiva, and corneal epithelium. Accordingly, it is expected that inhibition of ADAM12 activity and/or expression of one or more Adam 12 gene products as described herein will be useful to treat a broad range of wounds, including both acute wounds such as injury and surgical wounds and chronic wounds such as diabetic foot ulcers, venous leg ulcers, arterial ulcers, pressure ulcers, mouth ulcers, gingival wounds and corneal wounds.

Furthermore, the usefulness of inhibiting ADAM12 activity and/or expression of one or more Adam12 gene products is not limited to the treatment of chronic wounds or tissues overexpressing ADAM12. In some tissues, ADAM12 expression levels may not be observed, and yet inhibitors may be useful to reduce ADAM12 expression to promote epithelialization. For example, inhibition of ADAM12 activity and/or expression of one or more Adam12 gene products as described herein is expected to be useful in treating acute wounds, surgical incisions, sickle cell ulcers, and corticosteroid-induced wounds.

In another aspect, the present disclosure provides compositions useful for administering ADAM12 inhibitory compounds to treat and/or promote healing of wounds as described herein. The compositions generally comprise one or more ADAM12 inhibitory compounds and can be formulated for systemic or local administration. Specific examples of compositions formulated for systemic administration include, by way of example and not limitation, oral, epidermal, transdermal, transmucosal (including vaginally and rectally), pulmonary (including inhalation or insufflation of powders or aerosols, or by nebulizer), intratracheal, intranasal, ophthalmic, intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intraventricular formulations. Specific examples of compositions suitable for local administration include, by way of example and not limitation, transdermal patches, skin coverings, wound dressings, sprays, foams, emulsions, liquids, powders, ointments, creams, lotions, gels, jellies, pastes, salves, tinctures, drops, liposome-containing formulations, suppositories, enemas, coated condoms and gloves.

In some embodiments, the composition is a wound dressing or packing designed to deliver the ADAM12 inhibitory compound directly to the wounded tissue.

BRIEF DESCRIPTION OF THE FIGURES

FIG. 1 is a graphic illustration of the human gene encoding ADAM12 (GENBANK Accession No. NC000010.9) and its two mRNA variants;

FIG. 2 provides the nucleotide sequence (SEQ ID NO: 1) of mRNA encoding the membrane anchored form of ADAM12 (splice variant 1; also called “ADAM12-L”) (FIG. 2A), as well as the amino acid sequence (SEQ ID NO: 2) of the encoded ADAM12-L polypeptide (FIG. 2B);

FIG. 3 provides the nucleotide sequence (SEQ ID NO: 3) of mRNA encoding the secreted, short form of ADAM12 (splice variant 2; also called “ADAM12-S”) (FIG. 3A), as well as the amino acid sequence (SEQ ID NO: 4) of the encoded ADAM12-S polypeptide (FIG. 3B);

FIG. 4 provides photographs of biopsies taken from unwounded skin (FIG. 4A) and the non-healing edge of chronic skin ulcers (FIG. 4B) that were stained with a labeled antibody specific for ADAM12;

FIG. 5 provides photographs of skin explants from ADAM12 knockout (ADAM12−/−) and wild type (WT) mice that were either treated (+EGF) or untreated (−EGF) with epidermal growth factor; and

FIG. 6 provides a graph illustrating average keratinocyte migration distance observed as a function of time in skin explants taken from ADAM12 knockout (ADAM12KO) and wild type (WT) mice that were either treated (EGF+) or untreated (EGF−) with epidermal growth factor.

DETAILED DESCRIPTION

The various inventions described herein are based upon the surprising discovery that one member of the family of metalloprotease disintegrins referred to as ADAMs, a disintegrin and metalloprotease 12 (“ADAM12”), is an important mediator of wound healing and pathogenesis.

The ADAMs are a family of almost 30 known multidomain proteins so named because they contain a disintegrin and metalloprotease domain. ADAMs share high sequence homology and domain organization with snake venom metalloproteases; both contain a metalloprotease-like domain with an associated regulatory prodomain, a disintegrin-like domain, a cysteine-rich domain, and an epidermal growth factor-like domain. In addition, ADAMs contain a transmembrane domain and a cytoplasmic tail (Evans, 2001, BioEssays 23:628-639). The metalloprotease domains are involved in cleavage of the amyloid precursor protein and ectodomain shedding of several membrane-anchored proteins, including tumor necrosis factors. The disintegrin and cysteine-rich domains play important roles in cell-cell and cell-matrix adhesion, cell differentiation, and fusion (Primakoff & Myles, 2000, Trends Genet. 16:83-87). The cytoplasmic tails contain consensus SH3-binding motifs, and have been shown to bind p85, Src, and alpha-actinin-1 and -2, suggesting that the cytoplasmic tail region is involved in signal transduction (Blobel, C. P., 1997, Cell, 90:589-592; Blobel, C. P., 2005, Nat Rev Mol Cell Biol. 6(1):32-43; Cao et al., 2001, Biochem. J. 357:353-361; Galliano et al., 2000, J. Biol. Chem. 275:13933-13939; Kang et al., 2001, J. Biol. Chem. 276:24466-24472; Kang et al., 2000, Biochem. J. 352:883-892; Suzuki et al., 2000, Oncogene, 19:5842-5850).

ADAM12 was first identified in mice because of its involvement in myotube formation. During myoblast differentiation, the cells adhere to each other before their plasma membranes merge forming myotubes. The process of differentiation leading to cell fusion occurs in different tissues including placenta, bone, and muscle. ADAM12 was identified as a myoblast-expressed gene product and showed strong expression in neonatal skeletal muscle and bone (Yagami-Hiromasa et al., 1995, Nature 377:652-656).

The human gene encoding ADAM12 (also called meltrin-alpha) was cloned in 1998 and two isoforms were identified which arise from alternative splicing of a single gene (Gilpin et al., 1998, J. Biol. Chem. 273:157-166). The larger isoform is membrane-bound (designated ADAM12-L) and the shorter isoform (designated ADAM12-S) is secreted. The mRNA of both isoforms is abundant in human placenta and in some tumor cell lines. ADAM12-S has been identified as a possible protease responsible for the cleavage of insulin-like growth factor-binding protein-3 in blood during pregnancy (Shi et al., 2000, J. Biol. Chem. 275:18574-18580). The ADAM12-L transcript is also detected in human adult skeletal, cardiac, and smooth muscle. Human ADAM12 is an active metalloprotease and is synthesized as a zymogen, with the prodomain maintaining the metalloprotease in an a latent form (Loechel et al., 1998, J. Biol. Chem. 273:16993-16997; Loechel et al., 1999, J. Biol. Chem. 274:13427-13433).

The expression pattern of the ADAM12 transcripts examined during mouse embryogenesis revealed that the most prominent expression of ADAM12 is found in regions of bone formation. The ADAM12 gene is not activated in myotomes until myotube formation starts (Kurisaki et al., 1998, Mech. Dev. 73:211-215). The expression patterns of ADAM12 suggest that it is involved in cell fusion, but expression of ADAM12 is not restricted to fusagenic cells, suggesting a more general role for ADAM12. ADAM12 is also constitutively expressed in mouse C2 skeletal muscle cell fibroblasts (Kadota et al., 2000, J. Biochem. (Tokyo) 128:941-949), and ADAM12 mRNA has been detected in osteoblasts (Harris et al., 1997, J Cell. Biochem. 67:136-142).

ADAM12, with its capacity to mediate cell adhesion, has been implicated in the progression of several diseases. ADAM12 expression is up-regulated in human carcinoma tissue and may have a role in human cancer by supporting tumor cell adhesion through the cysteine rich domain of ADAM12 (Iba et al., 1999, Am. J. Pathol. 154:1489-1501). ADAM12 has been reported to play a role in breast tumor progression and stromal cell apoptosis (Kveiborg et al., 2005, Cancer Res., 65(11):4754-4761). Nude mice tumors generated from human rhabdomyosarcoma cells transfected with ADAM12-S contain a pattern of ectopic muscle cell formation as compared with control tumors. These tumors are of murine origin rather than derived from the human tumor cells, providing evidence that ADAM12-S provokes myogenesis in a nude mouse tumor model (Gilpin et al., 1998, J. Biol. Chem. 273:157-166).

The ADAM family of proteins may also be involved in the pathology associated with haematological malignancies. The cells derived from leukemia (HL60 and Jurkat), erythroleukemia (K562), lymphoma (U937 and Cupillo), and myeloma (U266B1) express mRNA of four members of the ADAM family, including ADAM12. It has been suggested that the metalloproteinase domains may be involved in egression of leukemic cells from the bone marrow into the peripheral blood, and that the integrin-like domain may be involved in adhesion-mediated migration mechanisms, such as that seen between bone marrow fibroblasts and erythroleukemia cell lines (Wu et al., 1997, Biochem. Biophys. Res. Commun. 235:437-442).

Cardiac hypertrophy is an adaptive response of the heart that occurs in various cardiovascular diseases, but prolonged hypertrophy typically culminates in chronic heart failure or sudden cardiac death. Inhibitors of metalloproteinases, in particular inhibitors of ADAM12, have been suggested as a therapeutic approach for cardiac hypertrophy. In cultured rat cardiomyocytes and in vivo, ADAM12 has been reported to cleave the membrane-anchored heparin-binding epidermal growth factor (HB-EGF) so that HB-EGF can induce a signaling cascade via G-protein coupled receptors leading to cardiac hypertrophy (Asakura et al., 2002, Nat. Med. 8:35-40).

Wound healing involves a complex interaction between epidermal and dermal cells, the extracellular matrix, controlled angiogenesis, and plasma derived proteins, all coordinated by an array of cytokines and growth factors. This dynamic process has been classically divided into several overlapping phases: inflammation, proliferation, migration and remodeling.

These various phases have been extensively studied and reviewed elsewhere. (See Martin, et al., 1997, Science, 276(5309):75-81). Germane to the instant disclosure are the proliferation and migration stages. The combination of new tissue and contraction of surrounding tissues is essential for the healing of chronic skin ulcers (Clark, 1993, Dermatol. Clin. 11:657-666). Fibroblasts are the key cells involved in the production of new extracellular matrix (in addition to producing collagen they produce tenascin, fibronectin, and proteoglycans such as hyaluronic acid). While this new matrix is synthesized, existing matrix in and around the wound region is degraded by several enzyme systems, including matrix metalloproteinases and plasminogen activators. The effect of metalloproteinases is regulated by tissue inhibitors, which are believed to be important in healing by preventing excessive matrix degradation (March et al., 1994, Arch. Dermatol. Res. 287:107-114). At an injury site, keratinocytes are also a part of the primary response to injury, releasing a signal and mobilizing other cell types (macrophages, platelets, endothelial cells and fibroblasts) to the site of injury. In addition, keratinocytes respond to cellular signals by undergoing two processes: migration and proliferation. Both of these processes are important for complete epithelialization and wound closure. During healing, some keratinocytes at the wound edge proliferate. Others undergo a marked transformation to enable them to phagocytose debris and migrate across the wound bed. Keratinocyte migration, coupled with wound contraction, results in re-epithelialization and wound closure. However, the epidermal morphology of chronic wounds differs from the morphology of normal epidermis and suggests that keratinocytes do not successfully complete activation or differentiation in chronic wounds (Stojadinovic, et al., 2005, Am J Pathol, 167:59-69; Tomic-Canic, Wounds, Suppl:3-5; Morasso, et al., 2005, Biol Cell, 97:173-183; Brem, et al., 2007, Molecular Medicine, in press). Instead, keratinocytes are caught in a ‘loop’ of trying, but not succeeding, to accomplish either of the two processes. Keratinocytes at the non-healing edge of chronic wounds appear to be hyperproliferative but non-migratory, suggesting that lack of migration leads to inability to epithelialize and plays important role in pathogenesis of chronic ulcers.

Although ADAM12 has been implicated in tissue remodeling and activation/inactivation of various growth factors implicated in wound healing, such as the membrane-anchored heparin-binding epidermal growth factor (HB-EGF) and insulin-like growth factor (IGF) binding proteins, it has not been recognized as a key target or mediator of wound healing.

Compelling evidence reported herein suggests that ADAM12 plays a significant role in wound healing, and that inhibition of ADAM12 activity and/or expression represents a powerful new mechanism-based approach towards the treatment of chronic wounds, and may accelerate wound healing in general. For example, as reported in Example 1, microarray analyses carried out with the non-healing edge of chronic human skin ulcers revealed a five-fold increase in ADAM12 expression as compared to normal skin tissue. The results of these microarray analyses were confirmed in immunohistochemical assays using an antibody specific for ADAM12 (Example 2). A pronounced (between 5- and 6-fold) increase in signal from both membrane-bound and cytoplasmic ADAM12 was found in the epidermis of chronic wounds as compared to normal skin controls. These studies confirm that ADAM12 is upregulated in chronic skin ulcers as compared to normal skin.

ADAM12 also inhibits migration of keratinocytes, an important step in the proliferation phase of wound healing. As reported in Example 3, skin explants taken at birth from ADAM12 knock-out (ADAM12−/−) and wild-type (WT) mice placed in tissue culture for a period of seven days both in the presence and absence of epidermal growth factor (EGF) showed statistically significant differences in keratinocytes migration. Statistically significant increases in migration of ADAM12−/− keratinocytes as compared to WT keratinocytes were observed in both untreated (p=0.0001) and EGF-treated (p=0.028) samples.

These studies provide compelling evidence that ADAM12 is an important mediator of wound healing, and that inhibition of ADAM12 activity or expression provides a new, powerful therapeutic approach towards promoting healing of wounds in general, and in particular healing of chronic skin ulcers. The compositions and methods described herein take advantage of this surprising and new-found role of ADAM12.

While not intending to be bound by any theory of operation, it is hypothesized that the increased expression of ADAM12 in wound tissue reported herein impairs healing of the wound through inhibition of keratinocyte migration, an important step in the proliferation cycle of wound healing. Inhibition of ADAM12 activity and/or expression is expected to modulate this important step in the wound healing process, promoting healing of wounds.

Although the observations reported herein were observed in skin tissue from non-healing chronic wounds, it is expected that ADAM12 also plays an important role in mediating the healing of wound in other tissue types, such as, for example mouth tissue, gingiva, and corneal epithelium, and in tissues of non-chronic wounds. Accordingly, the ADAM12 inhibitory compounds described herein are expected to be useful for promoting healing of virtually any type of wound, including but not limited to, chronic skin ulcers such as diabetic foot ulcers, arterial ulcers, venous leg ulcers, and pressure ulcers (bed sores), mouth ulcers, sickle cell ulcers, and corticosteroid-induced wounds and acute wounds of the skin, caused from insults like injury and/or surgery.

In some embodiments, the ADAM12 inhibitory compounds described herein are used to treat and/or promote healing of wounded tissues that over-express an Adam12 gene product. As used herein, wound tissue “over-expresses an Adam 12 gene product” if the wound tissue contains at least about 2-fold, and in some embodiments at least about 3-fold, 4-fold, 5-fold, or even higher, levels of an Adam 12 gene product, such as, for example, mRNA encoding an ADAM12 polypeptide, and/or an ADAM12 polypeptide per se, as compared to unwounded tissue of the same type.

In some embodiments, the ADAM12 inhibitory compounds described herein are used to treat or promote healing of wounded tissues that do not over-express an Adam12 gene product.

As mentioned above, human ADAM12 is expressed in two forms arising from two different splicings of a single gene: a larger, membrane-bound form (designated herein as “ADAM12-L”) and a shorter, secreted form (designated herein as “ADAM12-S”) (Gilpin et al., 1998, J. Biol. Chem. 273:157-166). As used herein, the expression “ADAM12” is intended to encompass the ADAM12-L and/or ADAM12-S forms of the ADAM12 polypeptide. Thus, the expression “ADAM12” or “ADAM12 polypeptide” includes either or both the ADAM12-L and ADAM12-S forms of the polypeptide expressed by the Adam12 gene, and the expression “Adam12 gene product” includes mRNA encoding ADAM12-L, mRNA encoding ADAM12-S, or mRNA encoding both ADAM12-L and ADAM12-S.

In some embodiments, wound tissue that over-expresses an Adam12 gene product contains at least about 2-, 3-, 4- or 5-fold more total ADAM12 mRNA as compared to unwounded tissue of the same type. In some embodiments, wound tissue that over-expresses an Adam12 gene product contains at least about 2-, 3-, 4- or 5-fold more total ADAM12 polypeptide as compared to unwounded tissue of the same type.

The nucleotide sequence of the human Adam 12 gene can be obtained from public data bases (GENBANK Accession No. NC000010.9). Two mRNA isoforms are illustrated in FIG. 1. The nucleotide and amino acid sequences of human ADAM12-L mRNA and polypeptides are illustrated in FIGS. 2A and 2B, respectively. Amino acids comprising the leader sequence of the pro-form of the ADAM 12-L polypeptide (“pro-ADAM 12-L” or “pro-ADAM12-L polypeptide”) are underlined. The nucleotide and amino acid sequences of human ADAM12-S mRNA and polypeptide are illustrated in FIGS. 3A and 3B, respectively. Amino acids comprising the leader sequence of the pro-form of the ADAM12-S polypeptide (“pro-ADAM12-S” or “pro-ADAM12-S polypeptide”) are underlined. In FIGS. 2A and 3A, the start codon is indicated in bold and underlined. In FIGS. 2B and 3B, the predicted cleavage site between the signal sequence and the pro-domain in the pro-ADAM12-L and pro-ADAM12-S forms is indicated with an arrowhead, the amino acid residues comprising the predicted mature ADAM12-L and ADAM12-S polypeptides and several predicted domains, (e.g., metalloprotease, disintegrin, cysteine-rich and transmembrane domains) are indicated, and the catalytic site is double-underlined.

Methods of determining amounts of mRNA gene expression products are well-known, and include by way of example and not limitation, microarray analyses (Brazma & Vilo, 2000, FEBS Lett. 480:17-24; Celis et al., 2000, FEBS Lett. 480:2-16), serial analysis of gene expression (“SAGE;” Madden et al., 2000, Drug Discovery Today 5:415-425), restriction enzyme amplification of digested cDNAs (“READS;” Prashar & Weissman, 1999, Methods Enzymol. 303:258-272), total gene expression analysis (“TOGA;” Sutcliffe et al., 2000, Proc. Nat'l Acad. Sci. USA 97:1976-81), expressed sequence tag sequencing (Celis et al., 2000, FEBS Lett. 480:2-16; Larsson et al., 2000, J. Biotechnol. 80:143-157), subtractive RNA fingerprinting (“SURF;” Fuchs et al., 2000, Anal. Biochem. 286:91-98; Larson et al., 2000, Cytometry 41:203-208), subtractive cloning differential display (“DD;” Jurecic & Belmont, 2000, Curr. Opin. Microbiol. 3:316-321), comparative genomic hybridization (Carulli et al., 1998, J. Cell. Biochem. Supp. 31:286-96) and quantitative real-time RT PCR (“A-Z of Quantitative PCR,” Bustin, Ed., INL Biotechnology Series, International University Line, La Jolla, Calif. 2004). Reagents for carrying out these assays can be designed from the nucleotide sequences illustrated in FIGS. 2-3 using well-known, routine means. Specific reagents and methods of quantifying Adam 12 gene transcription (mRNA) products via microarray analysis are described in Example 1. Specific probes and primers for quantifying Adam 12 gene transcription (mRNA) products via quantitative real-time PCR are provided in a later section.

Methods of determining amounts of polypeptide expression gene products are well-known, and include by way of example and not limitation, protein arrays and proteomics (Celis et al., 2000, FEBS. Lett. 480:2-16; Jungblat et al., 1999, Electrophoresis 20:2100-2110), fluorescent in situ hybridization (“FISH;” Going & Guskerron, 1999, Eur. J. Cancer 35:1895-1904) and mass spectrometry (review in TO, 2000, Comb. Chem. High Throughput Science. 3:235-241; see also ITRAQ® product literature from Applied Biosystems). Reagents for carrying out these assays are either commercially available or can be designed from the ADAM12 polypeptide sequences illustrated in FIGS. 2 and 3 using well-known, routine means. Specific reagents and methods of quantifying Adam12 gene translation products (ADAM12 polypeptide) are described in Example 2. Anti-human ADAM12 antibody is described in (Kveiborg et al., 2005, Cancer Res., 65(11):4754-4761).

The various methods and compositions described herein utilize ADAM12 inhibitory compounds. As used herein, an “ADAM12 inhibitory compound” is a compound that inhibits an activity of an ADAM12 polypeptide, or expression of an Adam12 gene product, such as for example synthesis of mRNA encoding an ADAM12 polypeptide (transcription) and/or synthesis of an ADAM12 polypeptide from ADAM12 mRNA (translation).

Compounds that inhibit an activity of an ADAM12 polypeptide can inhibit the protease activity of the metalloprotease domain of ADAM12-L and/or ADAM12-S. The chemical nature of the compounds can vary, and can range from small organic molecules, to small biological molecules such as peptides, hormones, oligonucleotides, to large biological molecules such as polypeptides, enzymes, antibodies and polynucleotides. It will be appreciated that this list is merely illustrative of representative types of ADAM12 inhibitory compounds that can be employed in the methods and compositions described herein, and is not intended to be limiting.

In some embodiments, the ADAM12 inhibitory compound inhibits the protease activity of the metalloprotease domain of an ADAM12 polypeptide. Such ADAM12 inhibitory compounds are known in the art, and include by way of example and not limitation, the class of inhibitors called “hydroxamate inhibitors,” which specifically intersect in a bidentate manner via the hydroxyl and carbonyl oxygens of their hydroxamic group with the zinc ion in the catalytic site (Grams et al., 1995, Biochem. 34:14012-14020; Bode et al., 1994, EMBO J. 13:1263-1269).

Such hydroxymate inhibitors are typically composed of a carbon back-bone (See, e.g., WO 95/29892, WO 97/24117, WO 97/49679 and EP 0780386), a peptidyl back-bone (WO 9005719, WO 93/20047, WO 95/09841 and WO 96/06074) a peptidomimetic back-bone (Schwartz et al., 1992, Progr. Med. Chem. 29: 271-334; Rasmussen et al., 1997, Pharmacol. Ther. 75:69-75; Denis et al., 1997, Invest. New Drugs 15:175-185). Alternatively, they contain a sulfonamide sulfonyl group which is bonded on one side to a phenyl ring and a sulfonamide nitrogen which is bonded to an hydroxamate group via a chain of one to four carbon atoms (See, e.g., EP 0757984).

Other peptide-based matrix metalloprotease inhibitors that can be used as ADAM12 inhibitory compounds in the methods described herein include thiol amides which exhibit collagenase inhibition activity (U.S. Pat. No. 4,595,700), N-carboxyalkyl derivatives containing a biphenylethylglycine which inhibit MMP-3, MMP-2 and collagenase (Durette, et al., WO 95/29689), lactam derivatives which inhibit matrix metalloproteases, TNF-alpha and aggrecanase (see U.S. Pat. No. 6,495,699), tricyclic sulfonamide compounds (see U.S. Pat. No. 6,492,422), the compound ONO-4817 (Ono Pharmaceutical Co. Ltd., Osaka, Japan; see also Mori, et al., 2002, Anticancer Res., 22(6C):3985-8) and the collagenase inhibitors GM6001 (trade name Galardin) and GM1489 (a derivative of GM6001) (see U.S. Pat. No. 6,759,432). Specific examples of hydroxamic acid-based metalloprotease inhibitors include, but are not limited to, the compounds “5A” [NHOHCOCH2 CH(i-Bu)CO-tryptophan-NHMe], “21A” [NHOHCOCH2 CH(i-Bu)CO-tryptophan-NHCHMePh], “39A” [HOOCCH2 CH(i-Bu)CO-tryptophan-NHCHMePh], “S1209” [NHOHCOCH2 CH(i-Bu)CO-tyrosine-OMeNHMe], UL001 [HSCH2 CH(CH2 CH(CH3)2)CO-Phe-Ala-NH2] and MP506 (Elastin Products Company, Inc.) (see U.S. Pat. Nos. 5,773,438 and 5,892,112).

Other compounds, such as KB-R7785, have also been reported to antagonize ADAM12 processing of HB-EGF (Asakura et al., 2002, Nat. Med. 8:35-40) and to act as inhibitors of ADAM12 (Oh et al., 2004, Bioorg Med Chem. Lett., 14(24):6071-6074).

Recently, a mechanism-based matrix metalloprotease inhibitor, SB-3CT, was designed according to the X-ray crystallographic information of the MMP active site (Brown et al., 2000, J. Am. Chem. Soc., 122:6799-6800). X-ray absorption studies revealed that binding of this molecule to the catalytic zinc reconstructs the conformational environment around the active site metal ion back to that of the pro-enzyme (Kleifeld et al., 2001, J. Biol. Chem. 276: 17125-17131). This SB-3CT compound is also expected to be useful as an ADAM12 inhibitory compound in the compositions and methods described herein.

Antibodies and antibody fragments capable of inhibiting the activity of matrix metalloproteases, and which are expected to be useful as ADAM12 inhibitory compounds in the compositions and methods described herein, are also known in the art. For example, one class of natural inhibitors is monoclonal antibodies. Several antibodies which may inhibit the activity of matrix metalloprotease have been raised against specific peptide sequences within the catalytic domain MMP-1 (Galvez et al., 2001, J. Biol. Chem. 276:37491-37500). Antibodies directed against ADAM12 are also described in Kawaguchi et al., 2003, J Cell Sci. 116(19):3893-904, incorporated herein by reference. Similarly, skilled artisans can raise antibodies against polypeptide sequences within the catalytic domain of ADAM12 to be used as ADAM12 inhibitory compounds.

The catalytic site of matrix metalloproteases includes a coordinated metal ion which becomes available for substrate binding following enzyme activation. Antibodies which recognize both electronic and structural determinants of the catalytic site have been reported to be potent inhibitors of matrix metalloproteases (See, for example, PCT Publication WO 04/087042).

Additional metalloprotease inhibitors expected to be useful as ADAM12 inhibitory compounds in the compositions and methods described herein include the various compounds disclosed in U.S. Pat. Nos. 6,500,847; 6,268,379; 5,968,795; 5,892,112; 5,872,152; 4,681,894; 4,943,587 and WO 06/014903, the disclosures of which are incorporated herein by reference.

Such ADAM12 inhibitory compounds will generally exhibit IC50s of ADAM12 protease activity of less than about 1 mM, as measured in a standard in vitro inhibition assay, such as, for example the ADAM12 inhibition assay described by Horiuchi et al., (Horiuchi et al., 2007, Mol Biol Cell, 18:176-188), although ADAM12 inhibitory compounds that exhibit higher IC50s may also find use in the methods and compositions described herein. In some embodiments, such ADAM12 inhibitory compounds will exhibit IC50s in the range of 100 μM, 75 μM, 50 μM, 25 μM, 10 μM, 1 μM, 100 nM, 75 nM, 50 nM, 25 nM, 10 nM, 1 nM, or even lower. As will be appreciated by skilled artisans, in many contexts, ADAM12 inhibitory compounds having IC50s in the micromolar, nanomolar or even subnanomolar range may be desirable.

The suitability of any compound that inhibits the metalloprotease activity of a matrix metalloprotease, including the above-described compounds for use as an ADAM12 inhibitory compound in the compositions and methods as described herein can be accessed in routine assays with ADAM12.

Animal models used to assay wound healing are well-known. Specific representative examples are taught in U.S. Pat. No. 7,060,795. For example, the healing rate of an ADAM12 inhibitory compound can be assessed by introducing the ADAM12 inhibitory compound into a wound and measuring whether the healing rate is altered by the presence of the inhibitor. The rate of wound healing in the presence and absence of an inhibitor can be determined. While any wound may be used, a wound model with predictable properties is desirable. Two animal chronic wound models commonly used are an ischemic rabbit ear model, and an induced diabetic rat model. Optimal epithelialization and wound healing may require some ADAM12 activity. Hence, the compositions and formulations provided herein do not necessarily promote maximal inhibition of ADAM12. Instead, the activity of the inhibitor formulation may be varied as needed to optimize epithelialization and wound healing, and may be varied according to whether the wound is a chronic or acute wound.

As will be recognized by skilled artisans, matrix metalloprotease inhibitors such as those described above may be “promiscuous” or “non-selective,” in that they may inhibit more than one matrix metalloprotease. While such promiscuous or non-selective inhibitors having requisite activity against ADAM12 are expected to be useful in the compositions and methods described herein, in some embodiments, compounds that specifically or selectively inhibit the activity of ADAM12 are preferred. Compounds can be tested for any desired degree of specificity or selectively for ADAM12 using routine screening assays.

In some embodiments, the ADAM12 inhibitory compound is a compound that inhibits expression of an Adam 12 gene product. Such ADAM12 inhibitory compounds can inhibit Adam12 gene expression by inhibiting the transcription of the Adam12 gene (i.e., by inhibiting synthesis of ADAM12 mRNA) and/or by inhibiting the translation of ADAM12 mRNA (i.e., by inhibiting synthesis of ADAM12 polypeptides). In some embodiments, such ADAM12 inhibitory compounds are oligonucleotides that are complementary to, or that specifically hybridize with, one or more nucleic acids encoding ADAM12, such as the Adam 12 gene or ADAM12 mRNA. The hybridization of the oligonucleotide with the nucleic acid encoding ADAM12 interferes with the normal function of the encoding nucleic acid.

As used herein, “hybridization” means hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleoside or nucleotide bases (nucleobases). For example, adenine and thymine are complementary nucleobases which pair through the formation of Watson-Crick hydrogen bonds. “Complementary,” as used herein, refers to the capacity for precise pairing between two nucleotides. For example, if a nucleotide at a certain position of an oligonucleotide is capable of hydrogen bonding with a nucleotide at the same position of a DNA or RNA molecule, then the oligonucleotide and the DNA or RNA are considered to be complementary to each other at that position. The oligonucleotide and the DNA or RNA are complementary to each other when a sufficient number of corresponding positions in each molecule are occupied by nucleotides which can hydrogen bond with each other. Thus, “specifically hybridizable” and “complementary” are terms which are used to indicate a sufficient degree of complementarity or precise base pairing such that stable and specific binding occurs between the oligonucleotide and the DNA or RNA. It is understood in the art that the sequence of an oligonucleotide need not be 100% complementary to its target nucleic acid to be specifically hybridizable.

An antisense compound is specifically hybridizable when binding of the compound to a target DNA or RNA molecule interferes with the normal function of the target DNA or RNA to cause a loss of activity, and there is a sufficient degree of complementarity to avoid non-specific binding of the oligonucleotide compound to non-target sequences under conditions in which specific binding is desired, i.e., under physiological conditions in the case of in vivo assays or therapeutic treatment, and in the case of in vitro assays, under conditions in which the assays are performed.

Oligonucleotide compounds capable of specifically hybridizing to nucleic acids encoding polypeptides are well-known in the art, and include by way of example, and not limitation, antisense oligonucleotides, small interfering RNAs (siRNAs), micro RNA (miRNA), short hairpin RNAs (shRNAs) and antisense expression vectors. Antisense oligonucleotides, siRNA and miRNA capable of inhibiting expression of Adam12 gene products can be designed from the sequences of polynucleotides encoding ADAM12, such as the Adam12 gene and ADAM12 mRNA sequences illustrated in FIGS. 1-3, using principles that are well known in the art. For example, antisense ADAM12 inhibitory compounds can be designed from polynucleotide sequences encoding ADAM12 as described in Yagami-Hiromasa et al., Nature, 1995, 377:652-656; Abe et al., 1999, Calcif. Tissue Int. 64:508-515. siRNA ADAM12 inhibitory compounds can be designed from ADAM12 encoding polynucleotides as described in Cao et al., 2003, Mol. Cell. Biol. 123(19):6725-6738. miRNA ADAM12 inhibitory compounds can be designed from ADAM12 encoding polynucleotides as described on several commercial websites (see, for example, world wide web at dharmacon.com). As will be recognized by skilled artisans, the lengths of such antisense, siRNA and miRNA molecules can vary, as can their degree of complementarity to the ADAM12 encoding polynucleotide. In some embodiments, antisense oligonucleotide ADAM12 inhibitory compounds will contain from 8-80, 8-70, 8-60, 8-50, 8-40, 8-35, 8-30, 8-25, 8-20, 8-15 or 8-10 nucleotides; siRNA ADAM12 will contain from 20 to 22 nucleotides; and miRNA ADAM12 inhibitory compounds will typically contain from 21 to 24 nucleotides. Oligonucleotide ADAM12 inhibitory compounds, including antisense, siRNA and miRNA oligonucleotides, need not exhibit 100% complementarity to the target region of the ADAM12-encoding nucleic acid to be effective in the methods and compositions described herein. Depending upon their length, oligonucleotides exhibiting between 80-100% complimentarity are expected to be effective, although in some circumstances oligonucleotides having even lower percentages of complementarity will be effective. The only requirement is that the oligonucleotide be capable of specifically hybridizing to its target region of the ADAM12-encoding nucleic acid. The correlation between the length of an oligonucleotide, its degree or percentage of complementary to a target nucleic acid sequence and its ability to specifically hybridize to the target sequence under various different hybridization conditions is well understood (see, e.g., Nucleic Acid Hybridization: A Practical Approach, Haines & Higgins, Eds., IRL Press, Oxford, 1988). Oligonucleotide ADAM12 inhibitory compounds having the requisite degree of complementarity as a function of length to be useful in the methods and compositions described herein can be designed using these well-known principles.

In some embodiments, the oligonucleotide ADAM12 inhibitory compound is 90-100% complementary to a target region of ADAM12-encoding nucleic acid, with a degree of complementarity of greater than 95, 96, 97, 98 or 99% being preferred. In some embodiments the oligonucleotide ADAM12 inhibitory compound is 100% complementary (“completely complementary”) to a target region of an ADAM12-encoding nucleic acid. Percentage of complementarity of an oligonucleotide to a region of a target nucleic acid can be determined routinely using basic sequence alignment search tools (BLAST programs; see, e.g., Altschul et al., 1990, J. Mol. Biol. 215:403-410; Zhang & Madden, 1997, Genome Res. 7:649-656).

The target region may be located in the untranslated region of the ADAM12-encoding nucleic acid or in the coding region, or may span the untranslated and coding regions. The targeted region can include the nucleic acid sequence surrounding the AUG start codon, which can include the AUG codon itself; sequences containing and/or surrounding the splice donor and/or acceptor sites in the introns of the Adam 12 encoding unprocessed mRNA transcript; sequences containing and/or surrounding the splice donor and/or acceptor sites in the exons of the Adam 12 unprocessed mRNA transcript; sequences spanning the intro-exon junctions of the unprocessed Adam 12 mRNA transcript; and the 3′ untranslated regions of the Adam12 mRNA. In some embodiments, an Adam12 specific antisense oligonucleotide can target the splicing acceptor or donor regions to cause exon skipping, thereby inhibiting expression of the ADAM12 polypeptide. These approaches for inhibiting expression are described in, among others, U.S. Pat. No. 6,784,291; U.S. Pat. No. 6,210,892; U.S. Pat. No. 6,653,466; and U.S. patent publication 2002/0055481; all publications incorporated herein by reference.

A variety of different antisense oligonucleotides that can be used as ADAM12 inhibitory compounds in the methods and compositions described herein are also described in US 2004/0002467, published Jan. 1, 2004, the disclosure of which is incorporated herein by reference. Specific antisense oligonucleotides contemplated to be useful in the methods and compositions described herein are provided in TABLE 1 of US 2004/0002467 as SEQ ID NOS:8-44, respectively. The nucleotide sequences of these oligonucleotides, as well as the specific antisense oligonucleotides containing these sequences described in Example 15 of US 2004/0002467, are incorporated herein by reference. The nucleotide sequences of these antisense oligonucleotides are reproduced below:

TABLE 1 TARGET SEQ SEQ ID TARGET % ID REGION NO SITE SEQUENCE INHIB NO: UTR 4 4026 ttgactagatctggttaatg 77 8 UTR 4 4152 ctacgaataatgatagcaga 89 9 UTR 4 3776 tttctatcaagcaccgcctt 71 10 UTR 4 4265 gttgcaagtgtttaagaaat 94 11 UTR 4 3815 tgctctagaaaggacagcga 89 12 UTR 4 3399 gactgctttccaataaggcc 83 13 UTR 4 4541 acactcagagtgaattatgt 90 14 UTR 4 3174 agcacagcacagcactgacg 84 15 Coding 4 2514 tcttatttgtaaacagcagt 82 16 UTR 4 4689 atactatgttgcctagatac 87 17 UTR 4 3462 gattctacttgggatctctt 87 18 UTR 4 4818 gctttacattttaaagaact 92 19 UTR 4 4530 gaattatgtaactgttgtca 82 20 UTR 4 4340 ccacatctcataatatttag 30 21 Coding 4 2915 ggtcctggccaaggcatgag 93 22 UTR 4 4545 taaaacactcagagtgaatt 75 23 UTR 4 3502 tggctctggcctgagatggg 69 24 UTR 4 3230 acattctgcataaattaata 62 25 UTR 4 4570 ttgaccccaaaagaaggctt 89 26 UTR 4 3733 aaacatgctcacggctggtc 93 27 UTR 4 4007 ggttcacatctgaaaatagt 85 28 UTR 4 4016 ctggttaatggttcacatct 93 29 UTR 4 3737 ttccaaacatgctcacggct 85 30 UTR 4 4870 aatcagttacagtaattctg 86 31 UTR 4 4617 caagattcttggtacatatt 87 32 UTR 4 4467 atggcattttaccttatctg 78 33 UTR 4 3844 tcccaagctaaataacctac 77 34 UTR 4 3328 tcaagttcactaaaatacta 37 35 UTR 4 4988 atgccagatctaaggatttg 81 36 UTR 4 4789 tagaataaacagatgcatgc 90 37 Coding 4 2843 gtttggcttacaggtcccct 93 38 UTR 4 3643 ctgtctgcaacccaggttct 90 39 UTR 4 4419 tcactttaaagaccttgtga 69 40 UTR 4 4589 tcaaagcataggaaaactgt 83 41 Coding 4 2544 gcacacaccttagtttttca 44 42 UTR 4 3339 agcaggatatttcaagttca 74 43 UTR 4 4683 tgttgcctagatacagtggg 84 44

In TABLE 1, the numbers in the % inhibition column are the percentage inhibition data reported in the '2467 publication for the antisense oligonucleotides described in Example 15 of the '2467 publication.

As indicated in TABLE 1, SEQ. ID NOS: 8-20, 22-34, 36-41 and 43-44 inhibited synthesis of human ADAM12 mRNA by at least 60% in quantitative real-time PCR experiments using the following PCR probes and primers:

(SEQ. ID NO: 5) Forward Primer: GCTCCACAATATCCACACCAAGT  (SEQ ID NO: 6) Reverse Primer: TGAAAAAAGGTGTCGGCTTCTC  (SEQ ID NO: 7) Probe: FAM-CCCAGATCCACCCACACCGCCTATATTAA-TAMRA 

as described in Examples 13-15 of US 2004/0002467, the disclosure of which is incorporated herein by reference.

In some embodiments, oligonucleotide ADAM12 inhibitory compounds have nucleotide sequences selected from SEQ ID NOS: 8-20, 22-34, 36-41 and 43-44.

In some embodiments, the oligonucleotide ADAM12 inhibitory compound will inhibit synthesis of ADAM12 mRNA by at least 70%, as measured in the quantitative real-time PCR assay described above. Specific examples of such ADAM12 inhibitory compounds include oligonucleotides having nucleotide sequences corresponding to SEQ ID NOS: 8-20, 22-23, 26-34, 36-39, 41 and 43-44.

In some embodiments, the oligonucleotide ADAM12 inhibitory compounds will inhibit synthesis of ADAM12 mRNA by at least 80%, as measured in the quantitative real-time PCR assay described above. Specific examples of such ADAM12 inhibitory compounds include oligonucleotides having sequences corresponding to SEQ ID NOS:9, 11-20, 22, 26-32, 36-39, 41 and 44.

In some embodiments, the ADAM12 inhibitory compound will inhibit synthesis of ADAM12 mRNA by at least 90%, as measured in the quantitative real-time PCR assay described above. Specific examples of such ADAM12 inhibitory compounds include oligonucleotides having nucleotide sequences corresponding to SEQ ID NOS: 11, 14, 19, 22, 27, 29 and 37-39.

The sites or regions of the ADAM12-encoding nucleic acids to which oligonucleotide ADAM12 inhibitory compounds are complementary are referred to herein as “target sites” or “target regions.” The nucleotide sequences of such target sites or target regions are referred to herein as “target sequences.” The sequences of the target sites complementary to ADAM12 oligonucleotide inhibitory compounds having sequences corresponding to SEQ ID NOS: 8-20, 22-34, 36-41 and 43-44 are provided in TABLE 2, infra, as SEQ ID NOS:45-78:

TABLE 2 REV SEQ  TARGET COMP OF ID  SITE SEQUENCE SEQ ID ACTIVE IN NO: 4026 cattaaccagatctagtcaa 11 H. sapiens 45 4152 tctgctatcattattcgtag 12 H. sapiens 46 3776 aaggcggtgcttgatagaaa 13 H. sapiens 47 4265 atttcttaaacacttgcaac 14 H. sapiens 48 3815 tcgctgtcctttctagagca 15 H. sapiens 49 3399 ggccttattggaaagcagtc 16 H. sapiens 50 4541 acataattcactctgagtgt 17 H. sapiens 51 3174 cgtcagtgctgtgctgtgct 18 H. sapiens 52 2514 actgctgtttacaaataaga 19 H. sapiens 53 4689 gtatctaggcaacatagtat 20 H. sapiens 54 3462 aagagatcccaagtagaatc 21 H. sapiens 55 4818 agttctttaaaatgtaaagc 22 H. sapiens 56 4530 tgacaacagttacataattc 23 H. sapiens 57 2915 ctcatgccttggccaggacc 25 H. sapiens 58 4545 aattcactctgagtgtttta 26 H. sapiens 59 3502 cccatctcaggccagagcca 27 H. sapiens 60 3230 tattaatttatgcagaatgt 28 H. sapiens 61 4570 aagccttcttttggggtcaa 29 H. sapiens 62 3733 gaccagccgtgagcatgttt 30 H. sapiens 63 4007 actattttcagatgtgaacc 31 H. sapiens 64 4016 agatgtgaaccattaaccag 32 H. sapiens 65 3737 agccgtgagcatgtttggaa 33 H. sapiens 66 4870 cagaattactgtaactgatt 34 H. sapiens 67 4617 aatatgtaccaagaatcttg 35 H. sapiens 68 4467 cagataaggtaaaatgccat 36 H. sapiens 69 3844 gtaggttatttagcttggga 37 H. sapiens 70 4988 caaatccttagatctggcat 39 H. sapiens 71 4789 gcatgcatctgtttattcta 40 H. sapiens 72 2843 aggggacctgtaagccaaac 41 H. sapiens 73 3643 agaacctgggttgcagacag 42 H. sapiens 74 4419 tcacaaggtctttaaagtga 43 H. sapiens 75 4589 acagttttcctatgctttga 44 H. sapiens 76 3339 tgaacttgaaatatcctgct 46 H. sapiens 77 4683 cccactgtatctaggcaaca 47 H. sapiens 78

By virtue of the demonstrated inhibitory activity of their respective complementary oligonucleotides, these targets have been experimentally found to be “open to,” and accessible for, hybridization with oligonucleotides. Those of skill will be able to design further embodiments of oligonucleotides capable of specifically hybridizing with these target sites to inhibit expression of ADAM12 using routine skill and experimentation. Accordingly, in some embodiments, the oligonucleotide ADAM12 inhibitory compound is an oligonucleotide capable of specifically hybridizing with a target sequence selected from SEQ ID NOS:45-78 under physiological conditions.

Those of skill will also be able to identify additional target sites and sequences, and oligonucleotides capable of specifically hybridizing thereto, using routine skill and experimentation. Specific examples of antisense RNAs and siRNAs effective in reducing ADAM12 levels are described in Kawaguchi, supra; Lafuste et al., 2005, Mol Biol Cell. 16(2):861-70; and Cao et al., 2003, Mol Cell Biol. 23(19):6725-6738.

As used herein, the expression “oligonucleotide” is intended to encompass not only oligoribonucleic acids and oligo 2′-deoxyribonucleic acids, that are composed of naturally occurring and/or encoding nucleotide bases (“nucleobases,” e.g., adenine, guanine, thymine, cytocine and uracil) sugars and phosphate ester internucloside linkages, but also oligomers composed of one or more non-natural nucleobases, sugars and/or internucleoside linkages that retain their ability to specifically hybridize to a target site of an ADAM12-encoding nucleic acid. In some embodiments, such modified oligonucleotides are preferred over native forms owing to certain desirable properties, such as, for example, enhanced cellular uptake, enhanced affinity for the ADAM12-encoding nucleic acid under physiological or other conditions of use, and/or increased stability in the presence of degradatory enzyme, such as, for example, nucleases.

Representative examples of modified internucleoside linkages (“backbones”) that include a phosphorous atom and that can comprise the oligonucleotide ADAM12 inhibitory compounds include, but are not limited to, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotri-esters, methyl and other alkyl phosphonates including 3′-alkylene phosphonates, 5′-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3′-amino phosphoramidate, aminoalkylphosphoramidates, phosphorodiamidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates and borano-phosphates having normal 3′-5′ linkages, 2′-5′ linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a 3′ to 3′, 5′ to 5′, or 2′ to 2′ linkage. In some embodiments, oligonucleotides having inverted polarity comprise a single 3′ to 3′ linkage at the 3′-most internucleotide linkage i.e., a single inverted nucleoside residue, which may be abasic (the nucleobase is missing or has a hydroxyl group in place thereof).

Methods of synthesizing oligonucleotides including one or more such modified internucleoside linkages are well-known. Specific representative methods are taught in U.S. Pat. Nos. 3,687,808; 4,469,863; 4,476,301; 5,023,243; 5,177,196; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,306; 5,550,111; 5,563,253; 5,571,799; 5,587,361; 5,194,599; 5,565,555; 5,527,899; 5,721,218; 5,672,697 and 5,625,050, the disclosures of which are incorporated herein by reference.

Representative examples of modified backbases that do not include a phosphorous atom and that comprise the oligonucleotide ADAM12 inhibitory compounds include, but are not limited to, oligonucleotides in which the backbone is formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages. These include those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; riboacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH2 component parts.

Methods of synthesizing oligonucleotides including one or more such modified internucleoside linkages are well-known. Specific representative examples are taught in U.S. Pat. Nos. 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; 5,792,608; 5,646,269 and 5,677,439, the disclosures of which are incorporated herein by reference.

Representative examples of modified nucleobases that comprise the ADAM12 oligonucleotide inhibitory compounds include, but are not limited to, 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine and other alkynyl derivatives of pyrimidine bases, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 2-F-adenine, 2-amino-adenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3-deazaguanine and 3-deazaadenine. Further modified nucleobases include tricyclic pyrimidines such as phenoxazine cytidine(1H-pyrimido[5,4-b][1,4]benzoxazin-2(3H)-one), phenothiazine cytidine (1H-pyrimido[5,4-b][1,4]benzothiazin; 2(3H)-one), G-clamps such as a substituted phenoxazine cytidine (e.g. 9-(2-aminoethoxy)-H-pyrimido[5,4-b][1,4]benzoxazin-2(3H)-one), carbazole cytidine (2H-pyrimido[4,5-b]indol-2-one), pyridoindole cytidine (H-pyrido[3′,′:4,5]pyrrolo[2,3-d]pyrimidin-2-one). Modified nucleobases may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example 7-deaza-adenine, 7-deazaguanosine, 2-aminopyridine and 2-pyridone. Further nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in The Concise Encyclopedia Of Polymer Science And Engineering, pages 858-859, Kroschwitz, J. I., ed. John Wiley & Sons, 1990, those disclosed by Englisch et al., 1991, Angewandte Chemie, International Edition, 30:613, and those disclosed by Sanghvi Chapter 15, Antisense Research and Applications, pages 289-302, Crooke, S. T. and Lebleu, B. ed., CRC Press, 1993. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligonucleotide ADAM12 inhibitory compounds. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-12° C. (Sanghvi, et al., ed., Antisense Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278) and in some embodiments are preferred base substitutions, especially when combined with 2′-O-methoxyethyl sugar modifications.

Methods of synthesizing oligonucleotides including one or more modified nucleobases are well-known. Specific representative examples are taught in U.S. Pat. Nos. 3,687,808; 4,845,205; 5,130,302; 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,457,187; 5,459,255; 5,484,908; 5,502,177; 5,525,711; 5,552,540; 5,587,469; 5,594,121, 5,596,091; 5,614,617; 5,645,985; 5,830,653; 5,763,588; 6,005,096; and 5,681,941, the disclosures of which are incorporated herein by reference.

Representative examples of modified sugars that can comprise the oligonucleotide ADAM12 inhibitory compounds include, but are not limited to, ribose that is substituted at the 2′-position with a group selected from OH, SH, SCH3, OCN, CN, CF3, OCF3, SOCH3, SO2CH3, ONO2, NO2, N3,NH2, Cl, Br, F, alkyl, O-alkyl, S-alkyl, NH-alkyl, alkenyl, O-alkenyl, S-alkenyl, NH-alkenyl, alkynyl, O-alkynyl, S-alkynyl, NH, alkynyl, O-alkyl-O-alkyl, alkyl, alkylaryl, O-alkylaryl, arylalkyl, O-arylalkyl, heterocycloalkyl, heterocycloalkylaryl, aminoalkylamino, polyalkylamino and substituted silyl where the alkyl group contains from 1-10 carbon atoms and the alkyl and alkynyl groups contain from 2-10 carbon atoms, and wherein the alkyl, alkenyl and alkynyl groups are optionally substituted. Specific examples of such 2′-substituent groups include, but are not limited to, O[(CH2)nO]m CH3, O(CH2)nOCH3,O(CH2)n NH2, O(CH2)n CH3, O(CH2)nONH2 and O(CH2)nON(CH2)nCH3]2, where n and m are each, independently of one another, integers ranging from 1 to 10; OCH2CH2OCH3, also known as 2′-methoxyethoxy, 2′-O-(2-methoxyethyl) or 2′-MOE (Martin et al., 1995, Helv. Chim. Acta. 78:486-504); OCH2)2ON(CH3)2, also known as 2′-DMAOE; OCH2OCH2N(CH3)2, also known as 2′-dimethyl-aminoethoxyethoxy, 2′-O-dimethyl 1-amino-ethoxy-ethyl or 2′-DMAEOE); OCH3; OCH2CH2CH2NH2; allyl (CH2CH═CH2); O-allyl (O—CH2CH═CH2); and F.

The 2′-modification may be in the arabino (up) position or the ribo (down) position.

Further representative examples of modified sugars that can comprise the oligonucleotide ADAM12 inhibitory compounds include ribose substituted at the 3′-position or 5′-position of the terminal nucleotide with groups such as those exemplified above for the 2′-position, 2′-5′ linked ribose or deoxyribose sugars, cyclobutyl groups, etc.

Methods of synthesizing oligonucleotides including one or more such modified sugars are well-known. Specific representative examples are taught in U.S. Pat. Nos. 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811; 5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,053; 5,639,873; 5,646,265; 5,658,873; 5,670,633; 5,792,747; and 5,700,920, the disclosures of which are incorporated herein by reference.

Another representative example of a modified sugar that can comprise the oligonucleotide ADAM12 inhibitory compound is ribose in which the 2′-hydroxyl group is linked to the 3′ or 4′-position, thereby forming a bicyclic sugar. The linkage bridging the 2′-oxygen atom and the 3′- or 4′-carbon atom is typically an alkylene group, for example, containing one or two methylene moieties (e.g., —CH2— or —CH2CH2—). Oligonucleotides containing such bicyclic sugars are commonly referred to as locked nucleic acids or “LNAs.” Methods for synthesizing LNAs are described in WO 98/39352 and WO 99/14226, the disclosures of which are incorporated herein by reference.

In some embodiments, modified oligonucleotides include compounds in which both the sugar and phosphate ester groups are replaced with non-sugar phosphate linkages (referred to herein as “oligonucleotide mimetics”). The nucleobases are retained for hybridization with the ADAM12-encoding nucleic acid. One representative example of such an oligonucleotide mimetic that has been shown to have excellent hybridization and other properties is referred to as a peptide nucleic acid (“PNA”). In PNAs, the sugar-phosphate backbone of an oligonucleotide is replaced with an amide containing backbone, the most common of which is an aminoethylglycine backbone (see, e.g., Nielsen et al., 1991, Science 254:1497-1500). Methods of synthesizing PNA compounds are described, for example, in U.S. Pat. Nos. 5,539,082; 5,714,331; and 5,719,262, the disclosures of which are incorporated herein by reference.

In some embodiments, oligonucleotide ADAM12 inhibitory compounds are oligonucleotides with phosphorothioate internucleosidic linkages, heteroatomic internucleosidiclinkages or combinations thereof. Specific exemplary heteroatomic interlinkages include, but are not limited to, —CH2—NH—O—CH2—, —CH2—N(CH3)—O—CH3— (known as a methylenene (methylimino) or MMI backbone, —CH2—O—N(CH3)—CH2—CH2, amides and morpholino. These internucleoside backbones, as well as methods of synthesizing oligonucleotides including them, are described, for example, in U.S. Pat. Nos. 5,489,677; 5,602,240; and 5,034,506, the disclosures of which are incorporated herein by reference.

Oligonucleotide ADAM12 inhibitory compounds can include one or more moieties or conjugates covalently bound to appropriate functional groups of the oligonucleotide moiety of the compound. Conjugate groups or moieties can be selected to impart the oligonucleotide ADAM12 inhibitory compound with specified properties as compared to an un-conjugated oligonucleotide, such as, for example, cellular uptake, cellular distribution, cellular activity, pharmacokinetic activities, etc. Exemplary conjugating groups include, but are not limited to, polyamines, polyamides, polyethylene glycols, polyethers, groups that enhance the pharmacodynamic properties of oligomers, and groups that enhance the pharmacokinetic properties of oligomers. Typical conjugate groups include cholesterols, lipids, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes. Groups that enhance the pharmacodynamic properties, in the context of this invention, include groups that improve oligomer uptake, enhance oligomer resistance to degradation, and/or strengthen sequence-specific hybridization with RNA. Groups that enhance the pharmacokinetic properties, in the context of this invention, include groups that improve oligomer uptake, distribution, metabolism or excretion. Representative conjugate groups are disclosed in International Patent Application PCT/US92/09196, filed Oct. 23, 1992 the disclosure of which is incorporated herein by reference. Conjugate moieties include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al., 1989, Proc. Natl. Acad. Sci. USA 86:6553-6556), cholic acid (Manoharan et al., 1994, Bioorg. Med. Chem. Let. 4:1053-1060), a thioether, e.g., hexyl-5-tritylthiol (Manoharan et al., 1992, Ann. N.Y. Acad. Sci., 660:306-309; Manoharan et al., 1993, Bioorg. Med. Chem. Let. 3:2765-2770), a thiocholesterol (Oberhauser et al., 1992, Nucl. Acids Res. 20:533-538), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et al., 1991, EMBO J. 10:1111-1118; Kabanov et al., 1990, FEBS Lett., 259 327-330; Svinarchuk et al., 1993, Biochimie 75:49-54), a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethyl-ammonium 1,2-di-O-hexadecyl-rac-gly-cero-3-H-phosphonate (Manoharan et al., 1995, Tetrahedron Lett., 36:3651-3654; Shea et al., 1990, Nucl. Acids Res. 18:3777-3783), a polyamine or a polyethylene glycol chain (Manoharan et al., 1995, Nucleosides & Nucleotides 14:969-973), or adamantane acetic acid (Manoharan et al., 1995, Tetrahedron Lett. 36:3651-3654), a palmityl moiety (Mishra et al., 1995, Biochim. Biophys. Acta 1264:229-237), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., 1996, J. Pharmacol. Exp. Ther. 277, 923-937). Oligonucleotide ADAM12 inhibitory compounds may also be conjugated to active drug substances, for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fenbufen, ketoprofen, (S)-(+)-pranoprofen, carprofen, dansylsarcosine, 2,3,5-triiodobenzoic acid, flufenamic acid, folinic acid, a benzothiadiazide, chlorothiazide, a diazepine, indomethicin, a barbiturate, a cephalosporin, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic. Oligonucleotide-drug conjugates and their preparation are described in U.S. Pat. No. 6,656,730 (filed Jun. 15, 1999) which is incorporated herein by reference in its entirety.

Representative United States patents that teach the preparation of such oligonucleotide conjugates include, but are not limited to, U.S. Pat. Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538; 5,578,717, 5,580,731; 5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4,587,044; 4,605,735; 4,667,025; 4,762,779; 4,789,737; 4,824,941; 4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082,830; 5,112,963; 5,214,136; 5,082,830; 5,112,963; 5,214,136; 5,245,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241, 5,391,723; 5,416,203, 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810; 5,574,142; 5,585,481; 5,587,371; 5,595,726; 5,597,696; 5,599,923; 5,599,928 and 5,688,941, the disclosures of which are incorporated herein by reference.

It is not necessary for all positions in a given oligonucleotide ADAM12 inhibitory compound to be uniformly modified, and in fact more than one of the aforementioned modifications may be incorporated in a single compound or even at a single nucleoside within an oligonucleotide. The oligonucleotide ADAM12 inhibitory compounds also include oligonucleotides which are chimeric compounds. “Chimeric” oligonucleotides or “chimeras,” in the context of this disclosure, are oligonucleotides that contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of an unmodified oligonucleotide compound. These oligonucleotides typically contain at least one region wherein the oligonucleotide is modified so as to confer upon the oligonucleotide increased resistance to nuclease degradation, increased cellular uptake, increased stability and/or increased binding affinity for the target nucleic acid. An additional region of the oligonucleotide may serve as a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids. By way of example, RNAse H is a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of oligonucleotide inhibition of gene expression. The cleavage of RNA:RNA hybrids can, in like fashion, be accomplished through the actions of endoribonucleases, such as interferon-induced RNAseL which cleaves both cellular and viral RNA. Consequently, comparable results can often be obtained with shorter oligonucleotides when chimeric oligonucleotides are used, compared to phosphorothioate deoxyoligonucleotides hybridizing to the same target region. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.

Chimeric oligonucleotides can be synthesized as composite structures of two or more oligonucleotides, modified oligonucleotides, oligonucleosides and/or oligonucleotide mimetics as described above. Such compounds have also been referred to in the art as hybrids or gapmers. Representative United States patents that teach the synthesis of such hybrid structures include, but are not limited to, U.S. Pat. Nos. 5,013,830; 5,149,797; 5,220,007; 5,256,775; 5,366,878; 5,403,711; 5,491,133; 5,565,350; 5,623,065; 5,652,355; 5,652,356; and 5,700,922, the disclosures of which are incorporated herein by reference.

The oligonucleotide ADAM12 inhibitory compounds described herein may be conveniently and routinely made through the well-known technique of solid phase synthesis. Equipment for such synthesis is sold by several vendors including, for example, Applied Biosystems (Foster City, Calif.). Any other means for such synthesis known in the art may additionally or alternatively be employed. It is well known to use similar techniques to prepare oligonucleotides such as the phosphorothioates and alkylated derivatives.

Non-oligonucleotide ADAM12 inhibitory compounds described herein can likewise be synthesized using standard techniques. For example, small organic ADAM12 inhibitory compounds can be synthesized using standard techniques of organic chemistry. Protecting groups suitable for use in such syntheses are described, for example, in Greene & Wuts, “Protective Groups in Organic Chemistry,” latest edition.

The ADAM12 inhibitory compounds may also be admixed, encapsulated, conjugated or otherwise associated with other molecules, molecule structures or mixtures of compounds, as for example, liposomes, receptor-targeted molecules, oral, rectal, topical or other formulations, for assisting in uptake, distribution and/or absorption. Representative United States patents that teach the preparation of such uptake, distribution and/or absorption-assisting formulations include, but are not limited to, U.S. Pat. Nos. 5,108,921; 5,354,844; 5,416,016; 5,459,127; 5,521,291; 5,543,158; 5,547,932; 5,583,020; 5,591,721; 4,426,330; 4,534,899; 5,013,556; 5,108,921; 5,213,804; 5,227,170; 5,264,221; 5,356,633; 5,395,619; 5,416,016; 5,417,978; 5,462,854; 5,469,854; 5,512,295; 5,527,528; 5,534,259; 5,543,152; 5,556,948; 5,580,575; and 5,595,756, the disclosures of which are incorporated herein by reference.

The ADAM12 inhibitory compounds described herein encompass any pharmaceutically acceptable salts, esters, or salts of such esters, or any other compound which, upon administration to an animal, including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof. Accordingly, for example, the disclosure is also drawn to prodrugs and pharmaceutically acceptable salts of the ADAM12 inhibitory compounds, compounds pharmaceutically acceptable salts of such prodrugs, and other bioequivalents.

The term “prodrug” indicates a therapeutic agent that is prepared in an inactive form that is converted to an active form (i.e., drug) within the body or cells thereof by the action of endogenous enzymes or other chemicals and/or conditions. In particular, prodrug versions of the oligonucleotides ADAM12 inhibitory compounds described herein are prepared as SATE [(S-acetyl-2-thioethyl)phosphate] derivatives according to the methods disclosed in WO 93/24510 to Gosselin et al., or in WO 94/26764 and U.S. Pat. No. 5,770,713 to Imbach et al., the disclosures of which are incorporated herein by reference.

The term “pharmaceutically acceptable salts” refers to physiologically and pharmaceutically acceptable salts of the compounds of the invention: i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto.

Pharmaceutically acceptable base addition salts are formed with metals or amines, such as alkali and alkaline earth metals or organic amines. Examples of metals used as cations are sodium, potassium, magnesium, calcium, and the like. Examples of suitable amines are N,N′-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, dicyclohexylamine, ethylenediamine, N-methylglucamine, and procaine (see, for example, Berge et al., “1977, Pharmaceutical Salts,” J. of Pharma Sci. 66:1-19). The base addition salts of said acidic compounds are prepared by contacting the free acid form with a sufficient amount of the desired base to produce the salt in the conventional manner. The free acid form may be regenerated by contacting the salt form with an acid and isolating the free acid in the conventional manner. The free acid forms differ from their respective salt forms somewhat in certain physical properties such as solubility in polar solvents, but otherwise the salts are equivalent to their respective free acid for purposes of the present invention. As used herein, a “pharmaceutical addition salt” includes a pharmaceutically acceptable salt of an acid form of one of the components of the compositions described herein. These include organic or inorganic acid salts of the amines. In some embodiments, acid salts are hydrochlorides, acetates, salicylates, nitrates and phosphates. Other suitable pharmaceutically acceptable salts are well known to those skilled in the art and include basic salts of a variety of inorganic and organic acids, such as, for example, with inorganic acids, such as for example hydrochloric acid, hydrobromic acid, sulfuric acid or phosphoric acid; with organic carboxylic, sulfonic, sulfo or phospho acids or N-substituted sulfamic acids, for example acetic acid, propionic acid, glycolic acid, succinic acid, maleic acid, hydroxymaleic acid, methylmaleic acid, fumaric acid, malic acid, tartaric acid, lactic acid, oxalic acid, gluconic acid, glucaric acid, glucuronic acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, salicylic acid, 4-aminosalicylic acid, 2-phenoxybenzoic acid, 2-acetoxybenzoic acid, embonic acid, nicotinic acid or isonicotinic acid; and with amino acids, such as the 20 alpha-amino acids involved in the synthesis of proteins in nature, for example glutamic acid or aspartic acid, and also with phenylacetic acid, methanesulfonic acid, ethanesulfonic acid, 2-hydroxyethanesulfonic acid, ethane-1,2-disulfonic acid, benzenesulfonic acid, 4-methylbenzenesulfonic acid, naphthalene-2-sulfonic acid, naphthalene-1,5-disulfonic acid, 2- or 3-phosphoglycerate, glucose-6-phosphate, N-cyclohexylsulfamic acid (with the formation of cyclamates), or with other acid organic compounds, such as ascorbic acid. Pharmaceutically acceptable salts of compounds may also be prepared with a pharmaceutically acceptable cation. Suitable pharmaceutically acceptable cations are well known to those skilled in the art and include alkaline, alkaline earth, ammonium and quaternary ammonium cations. Carbonates or hydrogen carbonates are also possible.

For oligonucleotide ADAM12 inhibitory compounds, specific examples of pharmaceutically acceptable salts include, but are not limited to (a) salts formed with cations such as sodium, potassium, ammonium, magnesium, calcium, polyamines such as spermine and spermidine, etc.; (b) acid addition salts formed with inorganic acids, for example hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid and the like; (c) salts formed with organic acids such as, for example, acetic acid, oxalic acid, tartaric acid, succinic acid, maleic acid, fumaric acid, gluconic acid, citric acid, malic acid, ascorbic acid, benzoic acid, tannic acid, palmitic acid, alginic acid, polyglutamic acid, naphthalenesulfonic acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acid, polygalacturonic acid, and the like; and (d) salts formed from elemental anions such as chlorine, bromine, and iodine.

When administered to patients as a therapeutic approach towards the treatment of and/or promotion of wound healing, the ADAM12 inhibitory compounds can be administered as the compound per se, or in the form of pharmaceutical compositions.

The compounds and/or pharmaceutical compositions may be administered in a number of ways depending upon whether local or systemic treatment is desired.

Administration may be topical (including ophthalmic and to mucous membranes including vaginal and rectal delivery), pulmonary, e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, epidermal and transdermal, oral or parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration. Oligonucleotide ADAM12 inhibitory compounds with at least one 2′-O-methoxyethyl modification are believed to be particularly useful for oral administration.

Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. Coated condoms, gloves and the like may also be useful. Preferred topical formulations include those in which the oligonucleotides of the invention are in admixture with a topical delivery agent such as lipids, liposomes, fatty acids, fatty acid esters, steroids, chelating agents and surfactants. Preferred lipids and liposomes include neutral (e.g. dioleoylphosphatidyl DOPE ethanolamine, dimyristoylphosphatidyl choline DMPC, distearolyphosphatidyl choline) negative (e.g. dimyristoylphosphatidyl glycerol DMPG) and cationic (e.g. dioleoyltetramethylaminopropyl DOTAP and dioleoylphosphatidyl ethanolamine DOTMA). ADAM12 inhibitory compounds may be encapsulated within liposomes or may form complexes thereto, in particular to cationic liposomes. Alternatively, the compounds may be complexed to lipids, for example cationic lipids. Exemplary fatty acids and esters include but are not limited to, arachidonic acid, oleic acid, eicosanoic acid, lauric acid, caprylic acid, capric acid, myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, monoolein, dilaurin, glyceryl 1-monocaprate, 1-dodecylazacycloheptan-2-one, an acylcarnitine, an acylcholine, or a C1-10 alkyl ester (e.g. isopropylmyristate IPM), monoglyceride, diglyceride or pharmaceutically acceptable salt thereof. Topical formulations are described in detail in U.S. patent application Ser. No. 09/315,298 filed on May 20, 1999 which is incorporated herein by reference in its entirety.

Compositions and formulations for oral administration include powders or granules, microparticulates, nanoparticulates, suspensions or solutions in water or non-aqueous media, capsules, gel capsules, sachets, tablets or minitablets. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders may be desirable. Preferred oral formulations are those in which the oligonucleotide is administered in conjunction with one or more penetration enhancers surfactants and chelators. Exemplary surfactants include fatty acids and/or esters or salts thereof, bile acids and/or salts thereof. Exemplary bile acids/salts include chenodeoxycholic acid (CDCA) and ursodeoxychenodeoxycholic acid (UDCA), cholic acid, dehydrocholic acid, deoxycholic acid, glucholic acid, glycholic acid, glycodeoxycholic acid, taurocholic acid, taurodeoxycholic acid, sodium tauro-24,25-dihydro-fusid-ate and sodium glycodihydrofusidate. Exemplary fatty acids include arachidonic acid, undecanoic acid, oleic acid, lauric acid, caprylic acid, capric acid, myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, monoolein, dilaurin, glyceryl 1-monocaprate, 1-dodecylazacycloheptan-2-one, an acylcarnitine, an acylcholine, or a monoglyceride, a diglyceride or a pharmaceutically acceptable salt thereof (e.g. sodium). In some embodiments, oligonucleotide or other ADAM12 inhibitory compounds are administered with combinations of penetration enhancers, for example, fatty acids/salts in combination with bile acids/salts. An exemplary combination is the sodium salt of lauric acid, capric acid and UDCA. Further penetration enhancers include polyoxyethylene-9-lauryl ether, polyoxyethylene-20-cetyl ether.

ADAM12 inhibitory compounds may be delivered orally, in granular form including sprayed dried particles, or complexed to form micro or nanoparticles. For oligonucleotide ADAM12 inhibitory compounds, complexing agents include poly-amino acids; polyimines; polyacrylates; polyalkylacrylates, polyoxethanes, polyalkylcyanoacrylates; cationized gelatins, albumins, starches, acrylates, polyethyleneglycols (PEG) and starches; polyalkylcyanoacrylates; DEAE-derivatized polyimines, pollulans, celluloses and starches. Particularly preferred complexing agents include chitosan, N-trimethylchitosan, poly-L-lysine, polyhistidine, polyornithine, polyspermines, protamine, polyvinylpyridine, polythiodiethylamino-methylethylene P(TDAE), polyaminostyrene (e.g. p-amino), poly(methylcyanoacrylate), poly(ethylcyanoacrylate), poly(butylcyanoacrylate), poly(isobutylcyanoacrylate), poly(isohexylcynaoacrylate), DEAE-methacrylate, DEAE-hexylacrylate, DEAE-acrylamide, DEAE-albumin and DEAE-dextran, polymethylacrylate, polyhexylacrylate, poly(D,L-lactic acid), poly(DL-lactic-co-glycolic acid (PLGA), alginate, and polyethyleneglycol (PEG). Oral formulations for oligonucleotide ADAM12 inhibitory compounds and their preparation are described in detail in U.S. application Ser. No. 08/886,829 (filed Jul. 1, 1997), Ser. No. 09/108,673 (filed Jul. 1, 1998), Ser. No. 09/256,515 (filed Feb. 23, 1999), Ser. No. 09/082,624 (filed May 21, 1998) and Ser. No. 09/315,298 (filed May 20, 1999), each of which is incorporated herein by reference in their entirety.

In some particularly convenient embodiments, the ADAM12 inhibitory compounds may usefully be incorporated into wound dressings for topical administration. Such dressings may be nonabsorbable or absorbable, air- and/or water-permeable, impermeable, or semipermeable, and may optionally effect controlled and/or sustained local release of the ADAM12 inhibitory compound contained therein. The wound dressings may include actives additional to one or more ADAM12 inhibitory compounds, such as antimicrobials and hemostatic agents.

Wound dressings comprising therapeutic actives suitable for use with ADAM12 inhibitory compounds are well-known.

For example, PCT Publication WO 07/024,972 discloses devices and methods for the treatment of wounds comprising an absorbent wound dressing material having non-leachable antimicrobial and anti-protease agents as well as a releasable antimicrobial agent and a releasable anti-protease agent that are ionically stabilized within the device so as to be released from the device in a controlled manner. PCT Publication WO 07/011,612 and US Patent Application publication No. 2007/0020320 disclose anti-microbial wound dressings comprising a water-soluble film-forming polymer carrier and a water-soluble glass-encapsulated anti-microbial agent that is embedded within the carrier and/or coated on a surface of the carrier, as well as methods of making said wound dressings. U.S. Patent Application publication No. 2007/0009586 discloses wound dressings with antimicrobial and elastase sequestration properties which provide enhanced wound fluid absorption, comprising a phosphorylated polysaccharide substrate complexed with at least one transition metal and an alginate coating.

Wound dressings suitable for delivery of ADAM12 inhibitory compounds to wounds include passive or activated matrices, which may be fully or partially conformable to a wound shape.

For example, ADAM12 inhibitory compounds may usefully be included within activated matrices, such as beads, microspheres, films, sponges, or wafers which may be used therapeutically by placement within the body. Such matrices may be used to coat surgical devices such as suture materials or implants. ADAM12 inhibitory compounds may be included within activated matrices such as those disclosed in U.S. Pat. No. 5,962,427, European Patent No. EP0892644, and European Patent Application EP1510224, which disclose compositions in which a therapeutic agent, such as a nucleic acid encoding a protein of interest, is associated with or impregnated within a matrix to form a gene activated matrix. Once the gene activated matrix is prepared, it may be stored for future use or placed immediately at the site of the wound.

Matrices may include either biological or synthetic matrices, or combinations thereof. Biocompatible matrices may be formed from both natural or synthetic materials. In some embodiments, the matrices are non-biodegradable, for use in treatments in which it is clinically desirable to leave permanent structures in the body. In some embodiments, the matrices are biodegradable, for use in treatments in which the expression or elaboration of the ADAM12 inhibitory compound is desired for a short duration of time.

The matrices may take the form of sponges, implants, tubes, telfa pads, band-aids, bandages, pads, lyophilized components, gels, patches, powders or nanoparticles. In addition, matrices may usefully be designed to allow for sustained release of the ADAM12 inhibitory compounds over prolonged periods of time.

In embodiments in which the matrices are to be maintained in contact with the wound for extended periods of time, non-biodegradable matrices may be employed, such as sintered hydroxyapatite, bioglass, aluminates, other bioceramic materials and metal materials, such as titanium. A suitable ceramic delivery system is described in U.S. Pat. No. 4,596,574. The bioceramics may be altered in composition, such as in calcium-aluminate-phosphate; and they may be processed to modify particular physical and chemical characteristics, such as pore size, particle size, particle shape, and biodegradability. Polymeric matrices may also be employed, including acrylic ester polymers and lactic acid polymers, as disclosed in U.S. Pat. Nos. 4,521,909, and 4,563,489, respectively. Particular examples of useful polymers are those of orthoesters, anhydrides, propylene-cofumarates, or a polymer of one or more γ-hydroxy carboxylic acid monomers, e.g., γ-hydroxy auric acid (glycolic acid) and/or γ-hydroxy propionic acid (lactic acid).

Wound dressings suitable for delivery of ADAM12 inhibitory compounds may also take the form of compressible and noncompressible bandages, plasters, and patches as are well known.

For example, PCT Publication WO 07/002,696 describes decompressive thermogenic bandage for delivery of pharmacological compositions for treatment of tissue wounds via a permeable dermal patch positioned inside a self-adhering vessel having a one-way valve allowing evacuation of said vessel, wherein the dermal patch is impregnated with a heat activated pharmacological composition. Application of vacuum to the treated tissue disinfects the tissue while providing a bacterial infiltration barrier.

In embodiments in which the ADAM12 inhibitory compounds are to be used to promote healing of surgical wounds, the compound can be incorporated into, or onto, a suture, staple, or other device that will be used to close the wound. Drug-containing and/or coated sutures, staples and other devices that can be used at wound sites with routine modification to deliver locally ADAM12 inhibitory compounds are known in the art. Representative examples are described in US Patent Application Nos. 20070010856 and 20060182778, for example.

As will be appreciated by skilled artisans, ADAM12 inhibitory compounds that are biological in nature, for example, peptides, polypeptides, oligonucleotides and polynucleotides, can be administered as the inhibiting compound per se, or alternatively nucleic acids encoding such inhibitory compounds can be administered, and the inhibitory compound expressed, either by transcription or translation, in vivo. Methods for administering nucleic acids encoding entire therapeutic agents are well known, and are described, for example, in US Patent Application Nos. 20040086486 and 20020151516, the disclosures of which are incorporated herein by reference. Uptake of nucleic acids by mammalian cells may be enhanced using several known transfection techniques including the use of transfection agents. The formulation which is administered may contain such agents. Example of these agents include cationic agents (for example calcium phosphate and DEAE-dextran) and lipofectants (for example LIPOFECTAM™ and TRANSFECTAM™).

The amount of ADAM12 inhibitory compound administered will depend upon a variety of factors, such as the mode of administration, the activity of the compound, the height, age, weight and general condition etc., of the patient, the bioavailabilty of the compound, the type and/or location of the wound to be treated, and other criteria apparent to the treating physician. Typically, an amount of compound effective to achieve a desired level of ADAM12 inhibition in the wounded tissue will be administered. Initial dosages can be estimated initially from in vitro assays and adjusted for specific desired ADAM12 inhibitory compounds using routine methods. For example, an initial dosage for use in animals may be formulated to achieve a circulating blood or serum concentration of active compound that inhibits about 25% or more of ADAM12 activity, or a process associated therewith, such as keratinocyte migration, as measured in an in vitro assay. Alternatively, an initial dosage for use in animals may be formulated to achieve a circulating blood or serum concentration of active compound that is equal to or greater than the IC50 as measured in an in vitro assay. A suitable dose may be from 0.1 to 100 mg/kg body weight such as 1 to 40 mg/kg body weight. Calculating dosages to achieve such circulating blood or serum concentrations taking into account the bioavailability of the particular active compound is well within the capabilities of skilled artisans. For guidance, the reader is referred to Fingl & Woodbury, “General Principles,” In: The Pharmaceutical Basis of Therapeutics, Chapter 1, pp. 1-46, 1975, and the references cited therein.

EXAMPLES

The working examples that follow, which are intended to be illustrative and not limiting, highlight various features of the invention(s) described herein.

Example 1 ADAM12 mRNA is Up-Regulated in Chronic Skin Ulcers

Preparation of Samples. Chronic Wound Skin Biopsies were Obtained from Discarded tissue after debridement procedures on three consenting patients with venous reflux ulcers (approved protocol 01-0960(001) 03sux). A small portion of the specimens were fixed in formalin and processed for paraffin embedding, whereas the majority of the samples were stored in RNAlater (Ambion) for subsequent RNA isolation. Normal skin specimens were obtained as discarded tissue from voluntary surgery (approved protocol 25121).

Hybridization of Probes. Samples stored in RNAlater were homogenized and total RNA isolated using RNeasy Mini Kit (Qiagen, Valencia, Calif.) according to the manufacturer's protocol. Approximately 5 μg of total RNA was reverse transcribed and amplified as described (Stojadinovic et al., 2007, J Biol. Chem. 282:4021-4034). The cRNA probe was biotinylated using the Affymetrix ENZO BioArray HighYield RNA Transcript Labeling Kit, and labeled cRNA was hybridized to HG-U133A arrays (more than 22000 probe sets) (Affymetrix, Santa Clara, Calif.). The arrays were washed, stained with avidin-biotin-streptavidin-phycoerythin labeled antibody using an Affymetrix fluidics station and then scanned using an Agilent GeneArray Scanner system (Hewlett-Packard, Palo Alto, Calif.) as described by Affymetrix.

Data Analysis. Microarray Suite 5.0 (Affymetrix) was used for data extraction and for further analysis, data mining tool 3.0 (Affymetrix, Santa Clara, Calif.) and GeneSpring™ software 7.3.1 (Silicon Genetics, Redwood City, Calif.) were used for normalization, fold change and p-value calculations. Statistical comparisons of expression level, between test and control samples were performed using ANOVA test. Only genes with a p-value of less than 0.05 were considered to be statistically significant. Differential expressions of transcripts were determined by calculating the fold change. Genes were considered regulated if the expression levels differed more than 2-fold relative to normal skin.

Results. To determine which molecules play a role in pathogenesis of wound healing we utilized biopsies from patients with chronic ulcers and compared their expression profiles with those of normal human skin using microarray technology, as described above. Different samples were compared and a specific transcriptional profile was obtained. To determine statistically significant changes in gene expression levels between chronic ulcers and normal human skin, statistical analyses were performed using volcano plot software and only a subset of genes that demonstrate statistical significance (p value<0.05) selected. Using a two-fold difference in expression as a cut-off value, 557 genes were found to be differentially regulated between normal skin and non-healing edges from chronic wounds. Expression of ADAM12 was found to be approximately 5-fold greater in biopsy samples of chronic skin ulcers from all three patients, as compared to normal skin, with the average being 5.88.

Example 2 ADAM12 is Up-Regulated in Chronic Skin Ulcers

Protocol. Seven micrometer thick sections from the paraffin-embedded skin samples from three patients described in Example 1 (using three replicates from each patient) were cut and stained overnight with anti-ADAM12 antibody (rb122) at 1:100 dilution (generous gift of Dr. Wewer U. M.; for description, see Kveiborg et al., 2005, Cancer Res., 65(11):4754-4761) using Vectastain ABC Elite Universal Kit (Vector Labs) as previously described (Wang et al., 2002, J. Biol. Chem. 277:26573-26580). Unwounded skin from the same patients was used as a negative control. All negative controls were prepared by substitution of the primary antibody with PBS. Staining was analyzed using Nikon Eclipse E800 microscope and digital images were collected using SPOT-Camera Advanced program.

Results. Photographs of exemplary stained normal and chronic wound tissue are provided in FIG. 4. As expected, ADAM12 was observed in both the cytoplasm and membrane of the epidermis of normal skin (FIG. 4A). A robust increase in ADAM12 was observed in biopsies originating from the non-healing edge of chronic ulcers was observed, confirming the microarray data of Example 1 (FIG. 4B). Membranous staining was patchy and present throughout the epidermis in chronic wound samples, while in normal skin, membranous staining was continuous throughout the membrane, but present only on the membrane surrounding small number of keratinocytes.

These data, along with those of Example 1, confirm that expression of ADAM12 is induced in chronic wounds.

Example 3 ADAM12 Inhibits Migration of Keratinocytes in Chronic Skin Ulcers

Generation of ADAM12 knock-out mice (“ADAM12KO”) and wildtype (WT) littermates. Two male and two female ADAM12+/− mice (Kurisaki et al., 2003, Mol. Cell. Biol. 23; 55-61) were mated in two separate cages, producing litters of between 5 and 7 pups in each cage. ADAM12KO and WT littermate mice were maintained in an accredited animal facility (at the Hospital for Special Surgery) according to the guidelines of the American Veterinary association, and all experiments were approved by the HSS Institutional Animal Care and Use Committee. Each mating cage littered mice within 24-36 hours of each other, producing mice of similar age when used for experimental analysis. A total of four litters were produced. All littered mice were sacrificed and their tails used for genotype analysis by southern blotting to determine ADAM12+/+ (WT) and ADAM12KO. A total of 25 newborn pups were sacrificed with genotypes as follows: 7 ADAM12KO, 6 ADAM12+/+, 12 ADAM12+/−. Among these, the wild type and ADAM12KO mice were selected for further experimental analysis. Five ADAM12KO pups and five WT littermates were littered from cage 1 and two ADAM12KO and 1 WT were littered from cage 2.

Explant Wound Healing Assay. Skin explants were prepared from 1 day old pups as described (Mazzalupo et al., 2003, Dev. Dyn. 226:356-65). Briefly, one day old mice were sacrificed and washed in a 10% povidine/iodine solution and rinsed in phosphate buffered saline (PBS). The epidermis, dermis, and underlying adipose tissue and fascia were completely removed from the body of the animal by incision on the ventral aspect of the specimen and careful dissection over the dorsal aspect. The adipose tissue/fascia was removed in order to insure adherence of the dermis directly to the plate. The tissue specimen was once again washed in 10% povidine/iodine solution followed by phosphate-buttered saline (“PBS”). Six 3-mm full thickness biopsies were taken from the dorsal midline of each animal and plated individually in 24 well plates. Each sample was submerged in 200 μL of nutrient-rich media containing adenine (24.4 μg/ml), insulin (5 μg/mL), transferrin (5 μg/mL), hydrocortisone (0.4 mM), 10% fetal calf serum, cholera toxin (0.5 μg/ml), penicillin G (100 IU/mL), T3 (1 mM), gentamicin (25 μg/mL), 75% Dublecco's Modified Eagle's Medium, and 25% F-12 nutrient mixture (GIBCO™, Invitrogen cell culture, Carlsbad, Calif.). For additional stimulation of proliferation/migration, three of the six samples from each mouse were treated with epidermal growth factor (“EGF”) (25 μg/mL) and the other three were left untreated. Samples were incubated overnight at 37° C. and, the following day, media was replaced with 1.5 mL of fresh media and EGF samples were re-treated. Keratinocyte migration was observed over the course of 7 days, with changing of media on days 1, 3, and 5.

To document the keratinocyte migration front, pictures were taken on days 3, 5, and 7 using a Nikon Coolpix 35 mm camera with manual zoom mounted on a Nikon Eclipse TS100 inverted microscope. The objective was a Plan-UW 2x/0.06 (Nikon, Inc.), and the software used to process images was Picasa 2.0 (Microsoft).

Pictures taken on days 3, 5, and 7 were used for data analysis. Each experiment was performed in triplicate and migration, and each photograph was assessed and quantified by two different people, each measuring independently, and each blinded to the source of the photograph. The distance of keratinocyte migration was calculated using the image program, ImageJ (NIH). Briefly, the center of the punch biopsy was determined and all other measurements were made from that point, and all measurements were normalized to length of the radius of the biopsy. Using the line tool, an average of 10 line measurements per sample were taken from the center point to the periphery of the punch biopsy in the areas of furthest cell migration. An average of the measurement from the center point to the edge of the greatest confluent cell migration was calculated from the 12 line measurements taken from each sample. Measurements were carried out in this manner (i.e., center to punch biopsy edge and center to keratinocyte migration front) in order to account for the differences in manual zoom of the pictures, which would overestimate or underestimate the migration distance. The values measured from the center point to the edge of the punch biopsy were averaged to give the average radius of the biopsy. The values measured from the center point to the edge of confluent cell migration were averaged to give the average radial distance of cell migration. The values charted in FIG. 6 (described below) are migration distance calculated from the center of the biopsy to biopsy radius. A total of 11 wild type EGF treated, 10 wild type untreated, 16 ADAM12KO EGF treated, and 18 ADAM12KO untreated biopsies were included in statistical analysis using Student's t-test available at http://www.physics.csbsju.edu/. P values of less than 0.05 were considered statistically significant.

After 7 days of migration the punch biopsies were removed from the wells and the remaining adherent cells washed with PBS and fixed for 10 minutes in methanol at −4° C. Following fixation, cells were washed with PBS and permeabilized with 0.1% Triton X-100 in PBS for 10 minutes at room temperature. Cells were then washed in PBS and incubated in 5% BSA in PBS (1-2 ml) for 30 min. Cells were incubated overnight in 200 μl/well of rabbit polyclonal antibody detecting human K17 (1:1000) (a gift from Dr Coulombe) (McGowan & Coulombe, 1998, J. Cell. Biol. 143:469-86) diluted in 5% BSA in 1×PBS. Slides were then rinsed in PBS and incubated with secondary fluorescent-conjugated anti-rabbit IgG antibody (diluted as 1:160)(Sigma-Aldrich) for three hours at room temperature (50 μL). After a final wash in PBS the stained slides were examined under a Carl Zeiss microscope and digital images collected using Adobe Photoshop 4.0 TWAIN 32 program.

Results. To functionally evaluate the role of ADAM12 in wound healing we used an established explant wound healing model in ADAM12 knockout mice (see Mahajan, et al., 2004, Mol. Cell. Biol. 24, 4994-5004). Ex vivo cultures of skin explants from wild type pups and their ADAM12−/− littermates were compared over a period of seven days. We measured and quantified keratinocyte migration for triplicate explants of all the pups from a total of four litters (from two cages) and found a significant increase in migratory capacity as well as response to EGF in the ADAM12KO mice when compared to WT controls. In all, we tested 78 explants from ADAM12KO mice in 5 separate trials. Thirty-four of these were tested on two separate trials in parallel with their wildtype littermates. On all occasions a significant increase in migration compared to wild type was observed. As expected, keratinocytes derived from WT mouse explants migrated out, and moved steadily during a seven day period (FIG. 5). When stimulated with EGF these keratinocytes responded with enhanced proliferation and migration. Interestingly, keratinocytes from explants derived from ADAM12KO pups showed a significant (p=0.0014) increase in migration compared to WT controls. Furthermore, additional stimulation by EGF also resulted in a significant increase in migration (p=0.026) in ADAM12KO skin explants, as compared to WT controls.

Lastly, we used a K17 specific antibody to test the origin of the migratory cells. Almost the entire cell population that migrated out of the explant was K17 positive (FIG. 5, panel labeled “K17Ab staining”), thus confirming that migratory cells are indeed keratinocytes.

To evaluate these findings in other members of the family of ADAMs we performed similar analyses using different mice backgrounds including ADAM 8−/−, ADAM 9−/−, ADAM15−/−, ADAM17−/−, ADAM19−/−, ADAM9−/− ADAM15−/− double knockout, ADAM9−/− ADAM12−/− ADAM15−/− triple knockout. ADAM9−/−, ADAM 17−/−, ADAM 19−/− showed no increase in migration compared to wildtype (data not shown). Interestingly, ADAM8−/− showed a dramatic reduction in keratinocyte outgrowth. These experiments were repeated on two separate occasions using the same method and analyses presented above. Taken together, we found that only ADAM12KO mice exhibit enhanced capacity of keratinocyte migration and better response to EGF stimulation, suggesting that inhibition of ADAM12 may promote epithelialization and wound healing by promoting the migration of keratinoctyes.

All publications, patents, patent applications and other documents cited in this application are hereby incorporated by reference in their entireties for all purposes to the same extent as if each individual publication, patent, patent application or other document were individually indicated to be incorporated by reference for all purposes.

While various specific embodiments have been illustrated and described, it will be appreciated that various changes can be made without departing from the spirit and scope of the invention(s).

Claims

1-15. (canceled)

16. A composition useful for promoting healing of a wound comprising an amount of an ADAM12 inhibitory compound effective to promote healing of wounded tissue.

17. In an article useful for dressing a wound, the improvement comprising including therein or thereon an amount of an ADAM12 inhibitory compound effective to promote healing of wounded tissue.

Patent History
Publication number: 20120052110
Type: Application
Filed: Aug 4, 2011
Publication Date: Mar 1, 2012
Applicant: Hospital for Special Surgery (New York, NY)
Inventors: Marjana TOMIC (Hillsdale, NJ), Carl Peter Blobel (Eastchester, NY), Asheesh Harsha (New York, NY)
Application Number: 13/197,833
Classifications
Current U.S. Class: Dressings (424/445); Peptides Of 3 To 100 Amino Acid Residues (530/300); Hormones, E.g., Prolactin, Thymosin, Growth Factors, Etc. (530/399); Polyclonal Antibody Or Immunogloblin Of Identified Binding Specificity (530/389.1); Dna Or Rna Fragments Or Modified Forms Thereof (e.g., Genes, Etc.) (536/23.1); The Acyclic Carbon Or Acyclic Carbon Chain Is Further Unsubstituted Or Alkyl Substituted Only (e.g., Tryptophane, Etc.) (548/496); Oxy In Acid Moiety (560/39); Phenylalanines (560/40); Monoclonal (530/388.1); Nucleic Acid Expression Inhibitors (536/24.5); Wound Healing Or Wound Repair Affecting (514/9.4); Hormone Or Derivative Affecting Or Utilizing (514/9.7); Immunoglobulin, Antiserum, Antibody, Or Antibody Fragment, Except Conjugate Or Complex Of The Same With Nonimmunoglobulin Material (424/130.1); Monoclonal Antibody Or Fragment Thereof (i.e., Produced By Any Cloning Technology) (424/141.1); 514/44.00R; 514/44.00A; C=x Bonded Directly Or Indirectly By An Acyclic Carbon Or Carbon Chain To Ring Carbon Of The Five-membered Hetero Ring (e.g., Tryptophan, Etc.) (x Is Chalcogen) (514/419); Compound Contains Two Or More C(=o)o Groups Indirectly Bonded Together By Only Conalent Bonds (514/533)
International Classification: A61L 15/20 (20060101); C07K 14/575 (20060101); C07K 16/18 (20060101); C07H 21/02 (20060101); C07D 209/20 (20060101); C07C 229/36 (20060101); A61K 38/02 (20060101); A61K 38/22 (20060101); A61K 39/395 (20060101); A61K 31/7105 (20060101); A61K 31/713 (20060101); A61K 31/405 (20060101); A61K 31/216 (20060101); A61P 17/02 (20060101); C07K 2/00 (20060101);