EP1804806A2 - Procedes et compositions destines a ameliorer la degradation de facteurs de transcription de recepteur nucleaire et utilisations correspondantes - Google Patents

Procedes et compositions destines a ameliorer la degradation de facteurs de transcription de recepteur nucleaire et utilisations correspondantes

Info

Publication number
EP1804806A2
EP1804806A2 EP05806820A EP05806820A EP1804806A2 EP 1804806 A2 EP1804806 A2 EP 1804806A2 EP 05806820 A EP05806820 A EP 05806820A EP 05806820 A EP05806820 A EP 05806820A EP 1804806 A2 EP1804806 A2 EP 1804806A2
Authority
EP
European Patent Office
Prior art keywords
receptor
nuclear receptor
degradation
nuclear
pathway
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP05806820A
Other languages
German (de)
English (en)
Inventor
Charles C.Y. c/o ANDROSCIENCE CORPORATION SHIH
Ching-Yuan c/o ANDROSCIENCE CORPORATION SU
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
AndroScience Corp
Original Assignee
AndroScience Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US11/045,181 external-priority patent/US20050209205A1/en
Application filed by AndroScience Corp filed Critical AndroScience Corp
Publication of EP1804806A2 publication Critical patent/EP1804806A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/12Ketones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/16Masculine contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/18Feminine contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/10Anti-acne agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones
    • A61P5/28Antiandrogens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones
    • A61P5/32Antioestrogens

Definitions

  • the present invention relates generally to the field of medicine, and particularly to methods for degradation of transcription factors, especially steroid nuclear hormone receptors such as the androgen receptor (AR), progesterone receptor (PR), and estrogen receptors a and ⁇ (ER), and uses thereof.
  • steroid nuclear hormone receptors such as the androgen receptor (AR), progesterone receptor (PR), and estrogen receptors a and ⁇ (ER)
  • Androgen exerts its function by entering a target cell and binding to a specific androgen receptor (AR), leading to the activation of androgen-regulating genes.
  • the male circulating androgen hormone, testosterone is converted within cells of peripheral tissues by 5-alpha reductase enzymes to the main intracellular androgen, dihydrotestosterone (DHT).
  • DHT dihydrotestosterone
  • anti-androgen drugs that either inhibit the conversion of testosterone into DHT (for example, 5-alpha-reductase inhibitors) or that interfere with the binding between androgen and AR, but these anti-androgen drugs can cause side effects, such as impotence in some men receiving treatment.
  • the androgen-induced AR activation pathway is a multiple step process (see, for example, Lee and Chang (2003) J. Clin. Endocrinol. Metab., 88:4043-4054, which is incorporated by reference in its entirety herein) and is believed to not simply be limited to androgen and AR binding.
  • the AR activation pathway involves AR protein expression and AR protein degradation, and in addition, involves androgen-AR complex (A-AR complex) formation, A-AR complex phosphorylation, A-AR complex translocation from cytoplasm into nucleus, and A-AR complex and AR coregulator complex (A-AR-ARA) formation (see, for example, Lin et al.
  • the androgen-AR-ARA complex is believed to function as a transcription factor, binding to the androgen response element (ARE) on the promoter region of androgen-regulated gene(s) and recruiting other regulatory proteins involved in general transcription machinery that lead to the activation (expression or repression) of androgen-responsive target gene(s).
  • ARE androgen response element
  • Two iso forms (the full-length AR-B and the N- terminus truncated AR-A) of the androgen receptor are expressed in immunologically detectable forms in many fetal and adult human tissues (Wilson and McPhaul (1996)).
  • High AR levels are found in both male and female fetal reproductive tissues, and in varying levels in non-genital fetal tissues.
  • High AR levels are also found in adult reproductive tissues (prostate, endometrium, ovary, uterus, fallopian tube, testis, seminal vesicle, myometrium, and ejaculatory duct), and lower levels in adult breast, colon, lung and adrenal gland tissue.
  • the AR pathway is especially important in the development and proper function of the male reproductive organs as well as non- reproductive organs (including muscle, hair follicles, and the brain). It is involved in the pathology of several diseases or conditions, including prostate cancer and other cancers, male infertility, and Kennedy disease.
  • Prostate cancer is the most common malignancy in American men in terms of incidence and prevalence. It is the most frequently diagnosed neoplasm in the United States and the second leading cause of cancer-related death for American men (Boring et al., 1992). Prostate cancer strikes more than 180,000 men each year, about the same number as cases of breast cancer in women. It caused 31,900 deaths among American men in 1999, second only to lung cancer. The increase in incidence of prostate cancer each year correlates with the aging of American male population.
  • Prostate cancer cell growth is believed to rely upon androgen-induced activation of the androgen receptor (AR).
  • AR androgen receptor
  • Most prostate cancers are dependent on androgen when first diagnosed (Heinlein and Chang 2004), and thus can be treated with anti-androgens.
  • One effective treatment for metastatic prostate cancer is androgen blockage therapy, which employs either surgical or chemical castration, combined with anti-androgen treatment, to suppress the biological action of androgens (Crawford et al., 1989).
  • the median duration of cancer response to hormone depletion is only 18-36 months, and the cancer almost always relapses and becomes androgen non-responsive. In such cases, patients face less desirable therapies, such as chemotherapy.
  • alterations of anti-androgens delay the progression of recurrent prostate cancers (Dupont et al, 1993; Taplin et al., 1999), indicating that a prostate tumor or cancer which relapses on a specific anti-androgen therapy may respond to a different anti-androgen.
  • Kennedy Disease also known as Kennedy Disease, spinal and bulbar muscular atrophy or spinobulbar muscular atrophy (“SBMA”), or Kennedy Syndrome (see, for example, Paul E. Barkhaus (2003), "Kennedy Disease", electronic publication available on ⁇ line at http://www.emedicine.com/neuro/topic421.htm, accessed 15 April 2004) — is a rare, X- linked recessive genetic neuromuscular disease that is estimated to affect 1 in 40,000 individuals worldwide. It is progressive, and currently incurable and non-treatable. Both the spinal and bulbar neurons are affected, causing muscle weakness and atrophy throughout the body, most noticeably in the extremities and in the face and throat. Kennedy Disease causes speech and swallowing difficulties, major muscle cramps, as well as other symptoms.
  • SBMA spinobulbar muscular atrophy
  • Kennedy disease is believed to be caused by an androgen receptor mutation consisting of an abnormally long polyglutamine expansion in the N-terminus region of the AR gene.
  • one aspect of the present invention includes a method of inhibiting or reducing the effect of a nuclear receptor activation pathway is disclosed including providing a cell including a nuclear receptor activation pathway and introducing a compound capable of inducing, enhancing or increasing degradation of the desired nuclear receptor.
  • the nuclear receptor pathway may include one or more transcription factors, cofactors, coregulators, corepressors or signalling pathways. Each of which may be affected using the disclosed methods.
  • Degradation of the nuclear receptor may include interfering with phosporylation of a transcription factor or cofactor, interfering with dimerization (homo- or heterodimerization) of a transcription factor or cofactor, interfering with binding between a trascription factor and a cofactor, interfering with nuclear transfer of a transcription factor or cofactor and the like.
  • a method of inhibiting or reducing the effect of a STAT activation pathway including providing a cell including a STAT activation pathway and introducing a compound capable of inducing, enhancing or increasing the degradation of a STAT transcription factor protein.
  • pharmaceutical or cosmetic compositions are provided that are capable of modulating the effect of a nuclear receptor activation pathway or a STAT activation pathway by enhancing or increasing degradation of a transcription factor, a nuclear receptor or a STAT transcription factor protein by any one or more of a variety of mechanisms.
  • methods of treating a medical condition or a cause or symptom of a medical condition including administering a cosmetic composition or pharmaceutical composition capable of enhancing or inducing degradation of a transcription factor such as a nuclear receptor or a STAT transcription factor protein to an individual suffering from or at risk of developing a nuclear receptor related or STAT related medical condition.
  • a cosmetic composition or pharmaceutical composition capable of enhancing or inducing degradation of a transcription factor such as a nuclear receptor or a STAT transcription factor protein to an individual suffering from or at risk of developing a nuclear receptor related or STAT related medical condition.
  • Nonlimiting examples of conditions that may be treated with the disclosed pharmaceuticals or cosmetics include male infertility, Kennedy disease, prostate cancer, breast cancer, liver cancer, bladder cancer, benign prostate hyperplasia, acne, baldness, hirsutism, exposed wounds and unwanted preganancy.
  • FIGURE 1 depicts a graphical representation of the results of an androgen receptor transactivation assay as described in Example 2 using CV-I, green monkey kidney cells (FIGURE IA) and LNCaP, human prostate cancer cells (FIGURE IB).
  • FIGURE IA Non-androgen expressing CV-I cells were transfected with a wild type androgen receptor expression vector utilizing MMTV as a reporter gene and pRL as an internal transfection control.
  • DHT was added at InM and compound JC9 was provided at varying concentrations from 0 uM to 7.5 uM. Cell lysates were collected, enzyme activities assayed and the results were compared using percent (%) relative luciferase activity.
  • compound JC9 was capable of inhibiting DHT-induced androgen receptor transcriptional activation in a dose dependent manner.
  • LNCaP cells which express a mutant androgen receptor, were transfected with MMTV and prL.
  • DHT was provided at InM and compound JC9 was provided from 0 uM to 7.5 uM.
  • JC9 was provided from 0 uM to 7.5 uM.
  • FIGURE 2 depicts a graphical representation of cell growth (proliferation) and androgen receptor expression levels in JC9-treated LNCaP cells.
  • LNCaP cells were plated and incubate for two days.
  • JC9 and hydroxyflutamide (HF) were added, separately, to the medium at a final concentration of 5 uM with and without DHT.
  • FIGURE 2 A the results demonstrate that while DHT promote LNCaP cell growth in culture, JC9 significantly inhibits cell growth regardless in the presence or absence of DHT. On the other hand, HF only moderately inhibits cell growth in either condition.
  • FIGURE 2B depicts normalized androgen receptor signals for the JC9 sample as a percentage of baseline (Day 0) value.
  • Cell lysates collected from cell cultured with JC9 (FIGURE 2A) and AR expression was detected by Western Blot. Data indicated that the inhibition of AR expression in LNCaP cells, induced by JC-9, is in correlation with cell growth inhibition.
  • FIGURE 3 depicts a western blots analysis of LNCaP cell lysates that were cultured with JC 15 for 20 hr. in the presence or absence of DHT. Data demonstrated that JC 15 reduced AR, PR and PSA proteins expression regardless in the presence or absence of DHT, but did not affect expression of other proteins, such as GR, ER, PPAR, RXR, HSP and Actin.
  • FIGURE 4 depicts a western blot analysis of T47D (a human breast cancer) cell lysates demonstrating the specificity of JC9's ability to degrade the androgen receptor.
  • AR androgen receptor
  • PP AR ⁇ , PPAR/3 Peroxisome proliferator-activated receptors gamma and beta
  • RXR ⁇ retinoid X receptor alpha
  • ERa and Er/3 extracellular signal-related kinase
  • HSP70 heat shock protein 70
  • FIGURE 5 depicts a western blot analysis of LNCaP cell lysates upon exposure to compound JC9 and cyclohexamide, a protein synthesis inhibitor.
  • the reduction of androgen receptor over time in the presence of a protein synthesis inhibitor indicates JC9 enhances degradation of AR protein.
  • FIGURE 6 depicts fluorescence micrographs of monkey kidney COS-I cells transfected with the plasmid GFPARQ49 (which contained a green fluorescent protein as a reporter and the mutant androgen receptor Q49) as described in detail in Example 5.
  • Transfected cells were treated with vehicle only (control) or with the test compound JC9.
  • Micrographs were taken under fluorescent imaging conditions for green fluorescent protein (GFP) and propidium iodide (PI). Control cells contained large amounts of fluorescent inclusions or aggregates. Cells that had been treated with JC9 contained substantially smaller amounts of fluorescent inclusions, suggesting that the expression of mutant Q49 androgen receptor was inhibited or degraded by JC9 treatment.
  • GFP green fluorescent protein
  • PI propidium iodide
  • FIGURE 7 depicts photographs demonstrating a representative visible improvement to skin condition resulting from topical application of the test compound JC15 (1 micromolar in a carrier base) to the forehead of an acne-affected male volunteer, as described in detail in Example 6.
  • FIGURE 7A depicts the volunteer's forehead prior to starting treatment with JC15.
  • FIGURE 7B depicts the same volunteer's forehead after one month of topical treatment with JC 15
  • FIGURE 8 depicts representative photographs of Fuzzy rats treated as described in detail in Example 7. Fuzzy rats were treated with topical creams containing vehicle only (left side animal) or JC9 (25 micromolar, right side animal) for the times indicated. The photographs show that bands of sebaceous glands were reduced within 4-5 weeks in the Fuzzy rats treated with JC9 (right side animal).
  • FIGURE 9 depicts representative photographs (FIGURES 9A-B) and graphical representations of duct and lobe size (FIGURES 9C-E) of sebaceous glands in Fuzzy rat skin. Skin tissue samples (split skin) were prepared and examined by microscopy.
  • FIGURE 9A-B are photographs depicting the duct and lobe of the sebaceous gland upon treatment with a vehicle control (9A) or compound JC9 (9B).
  • size of glandular lobes were measured by tracing the edges of the glandular lobes from areas of well-preserved glandular lobules, and then quantified with Image J software, and expressed as pixel counts contained within the traced areas. The data obtained showed that the size of the sebaceous glandular lobe is approximately twice in male Fuzzy rats as in females. Topical treatment with the vehicle only (control cream) did not produce a significant change in glandular lobe size. .
  • FIGURES 9D and 9E depict representative data showing that JC9 applied to skin in a vehicle cream significantly reduced the size of lobe and ducts of sebaceous glands in male Fuzzy rats.
  • FIGURE 1OA depicts results from studies of an animal model of alopecia (hair loss or baldness), as described in detail in EXAMPLE 8.
  • Six-week-old male C57BL/6J mice were shaved with an electric clipper, and then treated with a hair-removal cream.
  • One group of mice, represented by the two left-most animals (marked “vehicle #1” and “vehicle #2”) were shaved and treated only with ethanol.
  • a second group of mice, represented by the two right ⁇ most animals marked “testosterone #1" and "testosterone #2” were shaved and treated with a testosterone/ethanol solution in the morning and a control solution in the afternoon. The animals were photographed at the end of the 20-day treatment period.
  • FIGURE 1OB depicts further results from studies of an animal model of alopecia (hair loss or baldness), as described in detail in EXAMPLE 8.
  • Six-week-old male C57BL/6J mice were shaved with an electric clipper, and then treated with a hair-removal cream.
  • One group of mice, (represented by the animals marked "testosterone #1" and "testosterone #2”) were shaved and treated topically with testosterone in the morning and control solution in the afternoon for twenty days.
  • mice represented by the animals marked "JC9/testosterone #1" and "JC9/testosterone #2" were shaved and treated topically with testosterone in the morning and JC9 in the afternoon for twenty days.
  • Mice that received topical morning applications of testosterone and afternoon applications of the control solution only showed little or no re- growth of hair in the shaved areas after 20 days of treatment.
  • Mice that received topical morning applications of testosterone and afternoon applications of JC9 showed hair growth on day 8 and fully re-growth of hair in the shaved areas after 20 days of topical JC9 treatment.
  • FIGURE 11 depicts a photograph of nude mice inoculated with 2 million LNCaP cells.
  • the mice were given an intrapertioneal injection with either JC9 or a vehicle control three times per week for 7 weeks. After 7 weeks, the tumor was excised and weighed.
  • the JC9 treated nude mouse demonstrated a 75% reduction in tumor size compared to tumor excised from the mouse treated with vehicle control, indicated JC9 compound is capable of inhibits tumor cell growth in vivo.
  • the present invention makes use of a cell-based, functional assay system that measures reporter gene activity in order to identify novel compounds that inhibit a nuclear receptor pathway (or the androgen-induced androgen receptor (AR) activation pathway).
  • these novel compounds exert their anti-hormone activity through an inhibition mechanism different from that of the conventional 5-alpha-reductase inhibitors or androgen- AR binding inhibitors.
  • Active novel compounds have been identified, including, but not limited to, chemical derivatives and analogues of the natural compound, curcumin (diferuloylmethane), purified from the turmeric plant, Curcuma longa (see, for example, Ohtsu H. et al. (2002), J. Med.
  • curcumin derivatives include the JC series of compounds, such as JC9 (5 -hydroxy- l,7-bis(3, 4-dimethoxyphenyl)-l,4,6-heptatrien- 3-one) and JC 15 (7-(4-hydroxy-3-methoxyphenyl)-4-[3-(4-hydroxy-3- methoxyphenyl)acryloyl]-5-oxo-hept-6-enoic acid ethyl ester), whose structures and preparations are described in Ohtsu et al. (2002), J. Med. Chem., 45:5037-5042 and Ohtsu et al.
  • the present invention recognizes that specific degradation of a nuclear receptor is a novel and useful mechanism for modulating effects of a nuclear receptor-activated pathway.
  • the present invention answers the need for, and provides, new therapeutic approaches for preventing and treating disease conditions that are, at least in part, affected by the activity of a nuclear receptor, such as, but not limited to, steroid hormone receptors including the androgen receptor and the progesterone receptor.
  • the invention provides methods for treating or preventing such disease conditions in a subject.
  • the invention also provides methods for screening for compounds useful in the treatment or prevention of such disease conditions and disorders.
  • the invention further provides compositions useful in the treatment or prevention of such disease conditions.
  • the present invention includes several useful aspects, including: L A method of enhancing or inducing degradation of a nuclear receptor or a STAT transcription factor protein.
  • a pharmaceutical or cosmetic composition capable of enhancing or inducing degradation of a nuclear receptor or STAT transcription factor protein and thereby preventing or treating a cause or symptom of disease conditions that are, at least in part, affected by the activity of the nuclear receptor or STAT transcription factor protein.
  • a method to prevent or treat causes or symptoms of medical or disease conditions that are, at least in part, affected by the activity of a nuclear receptor or STAT transcription factor protein, by enhancing or inducing degradation of the nuclear receptor or STAT transcription factor protein.
  • the present invention includes a method of enhancing or increasing the degradation of a nuclear receptor (NR) or STAT transcription factor protein.
  • the methods of the present invention may therefore inhibit a nuclear receptor activation pathway or STAT activation pathway or both.
  • the methods may include providing a cell comprising a nuclear receptor pathway or STAT activation pathway and introducing a compound capable of enhancing degradation of a nuclear receptor or a STAT transcription factor protein.
  • Nuclear receptors are transcription factors that activate transcription of target genes in response to ligand. NRs are typically divided into type I, type II and type HI. Type I nuclear receptors include the classical steroid receptors (androgen receptor (AR), estrogen receptor a and ⁇ (ER), progesterone receptor (PR), glucocorticoid receptor (GR), and mineralocortocoid receptor (MR). Type II nuclear receptors dimerize with the 9-cis retinoic acid receptor (RXR) and include the receptors for vitamin D (VDR), thryroid hormone (TR), trans-retinoic acid receptor (RAR), and the peroxisome proliferator-activated receptors (PPAR). Type in includes orphan nuclear receptors (ONRs).
  • the method of the present invention may be applied to any one or more nuclear receptors or transcription factors of interest, where degradation of the nuclear receptor is desired.
  • Transcription factors of interest include the above-referenced type I, type II and type HI nuclear receptors, steroid hormone receptors, thyroid hormone receptor-like factors, STAT transcription factors, heat shock factors, and the like.
  • a list of suitable transcription factors is provided, for example, by the eukaryotic transcription factor database TRANSFAC® available at www.gene-regulation.com/pub/databases/transfac/cl.html (accessed 27 August 2004), which is incorporated by reference in its entirety herein.
  • Nuclear receptors of special interest include steroid hormone receptors, such as androgen receptors, progesterone receptors, estrogen receptors ( ⁇ and ⁇ ), glucocorticoid receptors, peroxisome proliferator- activated receptors (PPAR), 9-cis retinoic acid receptors (RXR), all trans-retinoic acid receptors (RAR) and orphan steroid hormone receptors.
  • steroid hormone receptors such as androgen receptors, progesterone receptors, estrogen receptors ( ⁇ and ⁇ ), glucocorticoid receptors, peroxisome proliferator- activated receptors (PPAR), 9-cis retinoic acid receptors (RXR), all trans-retinoic acid receptors (RAR) and orphan steroid hormone receptors.
  • RAR family members which include RAR ⁇ ,
  • RAR/3 and RAR ⁇ have a high affinity for all trans-retinoic acids and belong to the same class of nuclear transcription factors as thyroid hormone receptors, vitamin D3 receptor and ecdysone receptor.
  • the human RARa gene maps to chromosome 17.
  • the method may be applied to non-cell based screens, such as assays using cell-free preparations or isolated nuclear receptors, to isolated cells, such as cells grown in in vitro culture for assay purposes, or to intact, living subjects.
  • suitable subjects include mammals of research, agricultural, or economic interest, including rodents, lagomorphs, canids, felids, swine, bovids, and non-human primates.
  • Subjects can include human subjects of any age, sex, or physical condition.
  • Subjects of particular interest include human subjects diagnosed as having, or as being at risk for, a disease condition or conditions that are, at least in part, affected by the activity of a nuclear receptor such as a steroid hormone receptor (for example, an androgen receptor, a progesterone receptor, or an estrogen receptor).
  • a nuclear receptor such as a steroid hormone receptor (for example, an androgen receptor, a progesterone receptor, or an estrogen receptor).
  • Such disease conditions include, but are not limited to, cancers (for example, prostate cancer, liver cancer, bladder cancer, and other cancers which involve the androgen receptor activation pathway, and breast cancer, which is affected by the androgen receptor and estrogen receptor), neurological and neuromuscular disorders (for example, Kennedy Disease, which is affected by the androgen receptor), skin disorders (for example, acne, which is caused by androgen- induced AR activation of sebaceous glands), hair disorders (for example, androgenetic alopecia or "male pattern baldness", where hair loss is caused in part by the androgen receptors in follicles and adjacent cells), and wound healing (where inflammation is affected by the androgen receptor in response to androgen).
  • Subjects may also include female animals in which pregnancy is not desired, or women of child-bearing age who do not wish to become pregnant, where degradation of the progesterone receptor may be useful in preventing conception or in inducing still birth.
  • the method may include administering one or more compounds able to enhance, induce or increase the rate of degradation of a nuclear receptor (NR), such as, but not limited to, a steroid hormone receptor.
  • NR nuclear receptor
  • the one or more compounds may act using one or more mechanisms of nuclear receptor degradation.
  • administering the one or more compounds results in the specific degradation of a targeted nuclear receptor, without substantially altering the levels or activity of other, non-targeted transcription factors or nuclear receptors.
  • the method may include administering the one or more compounds in a quantity sufficient to degrade the targeted nuclear receptor in a given tissue or cell type, thus lowering the targeted nuclear receptor to a desired level (for example, where the targeted nuclear receptor is the androgen receptor, the desired androgen receptor level may be a level that is substantially non-responsive to circulating androgen).
  • the method of the invention may make use of any one or more suitable mechanisms to induce, increase or enhance the degradation of a nuclear receptor.
  • suitable mechanisms include, but are not limited to, interfering with translocation of the nuclear receptor into the nucleus or retaining the nuclear receptor in the cytoplasm of a cell, exposing a motif within the nuclear receptor able to induce protease activity, increasing activity of a protease capable of degrading the nuclear receptor, inhibiting the stabilization of a nuclear receptor, reducing the solubility of the nuclear receptor, activating a pathway able to degrade the nuclear receptor, increasing ubiquination of the nuclear receptor, increasing phosphorylation of the nuclear receptor by an appropriate kinase (for example, in the case of the androgen receptor, by activating Akt kinase, which in at least some cases phosphorylates the androgen receptor, leading to the receptor's ubiquination and subsequent degradation by the proteosome; see, for example, Heinlein and Chang (2004) and Lin et al.
  • the method of the present invention induces or enhances degradation of a nuclear receptor by interfering with the translocation of the nuclear receptor into the nucleus.
  • androgen-bound androgen receptors are translocated from the cytoplasm to the nucleus where they regulate genes using a zinc finger motif. When blocked from translocation or when retained within the cytoplasm, the androgen receptor undergoes proteolysis, and is thus unable to affect the nuclear DNA.
  • the method of the present invention enhances or induces degradation of a nuclear receptor by exposing within the nuclear receptor a site or motif that is able to induce proteolysis in the presence of a protease.
  • Such exposure may occur by inducing a conformational change of a domain within the nuclear receptor, such as by binding a compound to the nuclear receptor or by phosphorylating a domain of the nuclear receptor.
  • a PEST proline-, glutamate-, serine-, and threonine-rich motif has been identified as able to induce ubiquitin-dependent protoeolysis in the presence of the E3 ligase Mdm2 and has been found in a hinge region of the androgen receptor (Lin et al. (2002) EMBOJ., 21 :4037-4048, which is incorporated by reference in its entirety herein).
  • a compound able to expose a motif such as PEST in the presence of a ligase such as Mdm2 may induce or enhance degradation of the androgen receptor and thus inhibit the androgen receptor-activated pathway.
  • increasing the activity of a suitable nuclear receptor-specific protease may induce or enhance the increase in nuclear receptor degradation.
  • the method of the present invention induces or enhances degradation of a nuclear receptor by preventing or reducing stabilization of the nuclear receptor.
  • Preventing or decreasing stabilization may occur by inhibiting interactions between two or more nuclear receptor or by preventing or decreasing the interaction between a nuclear receptor and a cofactor.
  • the androgen receptor is believed to dimerize with another androgen receptor, thereby increases the stability of the receptor. Dimerization may occur via interaction between the amino terminals of the receptors. Preventing dimerization, for example, by administering a compound able to reduce or eliminate dimerization by binding at or near the amino terminal domain, may induce or enhance the degradation of the androgen receptor and thus inhibit the androgen receptor activated pathway.
  • the interaction between a nuclear receptor and a cofactor may be disrupted, thereby inducing degradation of the nuclear receptor, hi one embodiment the present invention disrupts the interaction between a nuclear receptor and a STAT (signal transducer and activator of transcription) protein. hi another embodiment, the method of the present invention induces or enhances degradation of a nuclear receptor by inhibiting or reducing the binding of a stabilizing cofactor to the nuclear receptor.
  • steroid receptor coactivator 1 SRC-I
  • DBD DNA binding domain
  • a compound able to inhibit or reduce the binding of SRC-I may prevent or reduce SRC-I stabilization of the androgen receptor and thus result in increased degradation of the androgen receptor.
  • the method of the present invention induces or enhances degradation of a nuclear receptor by destabilization of a domain of the nuclear receptor.
  • a domain of the nuclear receptor For example, some studies suggest that the AF-2 domain of the androgen receptor stabilizes the receptor's overall structure, allowing the amino terminal domain to interact with coregulators.
  • a compound able to destabilize the AF-2 domain for example, by interacting or binding with the AF-2 domain or with a domain that itself interacts with the AF-2 domain, may reduce the stabilization of the amino terminal domain, reduce the interaction with coregulators, and increase the rate of degradation of the androgen receptor.
  • the method of the present invention induces or enhances degradation of a nuclear receptor by activating a pathway able to degrade the nuclear receptor.
  • the caspase-3 pathway has been suggested to induce degradation of the androgen receptor (Lee and Chang (2003)). Activation of the caspase-3 pathway may occur by the presence of a tumor suppressor, phosphatase and tensin homologue, PTEN, such as by an interaction between the AR DNA-binding domain and the PTEN phosphatase domain, believed to lead to AR retention in the cytoplasm. Therefore a compound able to induce or enhance the caspase-3 pathway may induce or enhance the degradation of the androgen receptor, hi another example, Akt kinase, which phosphorylates the androgen receptor, maybe activated, leading to ubiquination and subsequent degradation of the androgen receptor by the proteosome (Heinlein and Chang 2004).
  • interactions of STAT are reduced or inhibited.
  • the methods may include but are not limited to inhibiting the phosphorylation of STAT, reducing or inhibiting dimerization of STAT, reducing or inhibiting binding between STAT and a nuclear receptor and the like.
  • the present invention includes methods of targeting or reducing the STAT signalling Pathway.
  • STAT (signal transducer and activator of transcription) proteins are a family of latent cytoplasmic transcription factors involved in cytokine, hormone nuclear receptor, and growth factor signal transduction.
  • STAT transcription factor proteins mediate broadly diverse biologic processes, including cell growth, differentiation, apoptosis, fetal development, transformation, inflammation, and immune response. Seven members of the STAT family of transcription factors have been identified in mammalian cells: STATl, STAT2, STAT3, STAT4, STAT5a, STAT5b, and STAT6.
  • STAT transcription factor proteins are activated by the Janus superfamily tyrosine kinases (JAKs), which themselves are activated by interferons, IL-I, IL- 2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-11, IL-12, IL-13, growth hormone (GH), prolactin (PRL), erythropoietin (EPO), thrompopoietin, and tumor necrosis factor (TNF), granulocyte- macrophage colony-stimulating factor (GM-CSF).
  • JOKs Janus superfamily tyrosine kinases
  • STATs are believed to be phosphorylated on a single tyrosine residue in the carboxy terminal portion.
  • the modified STATs form homodimers or heterodimers through reciprocal interaction between the phosphotyrosine of one STAT and the SH2 domain of another.
  • STATs like nuclear receptors, are translocated to the nucleus and interact with specific regulatory elements to induce or enhance target gene transcription.
  • the methods of the present invention may reduce the effect of STAT or enhance or induce degradation of STAT.
  • STATl is believed to be involved in the TNFR1-TRADD signalling complex.
  • STATl acts as a TNFRl -signaling molecule to suppress NF-kB activation. It has also been shown that its activation slows growth and promotes apoptosis. After phosphorylation, STATl and STAT2 form heterodimers that function as more potent inducers of transcription than the STATl homodimer.
  • STATl is known to associate with the transcription factors p48, SpI, and p300.
  • Human STAT2 cDNA codes for a 851 amino acid protein with a predicted molecular weight of 113 kDa.
  • STAT2 associates with beta subunit of the type I IFN receptor in an interferon-dependent manner.
  • the unique acidic domain of the carboxy-terminal region of STAT2 may interact with cAMP-response-element binding protein.
  • STAT3 has been shown to be activated by IFN-alpha but not IFN-beta.
  • the transcription factors associated with STAT3 are c-Jun and cyclic AMP-responsive enhancer binding protein (CREB).
  • STAT4 is activated when cells are treated with interleukin-12, a key cytokine regulator of cell-mediated immunity. STAT4 is also activated by IFN alpha/beta.
  • STAT4 knock-out mice lymphocytes no longer proliferate in response to IL- 12, produce IFN gamma, or express natural killer cell cytotoxicity.
  • Both STAT5a and STAT5b regulate interleukin-7 induced B-cell precursor expansion.
  • STAT5b may also act as a transcriptional inhibitor as demonstrated by inhibition of NF-kB mediated signaling.
  • This STAT5b-mediated inhibitory effect on NF-kB signaling does not depend on STAT5b-DNA interactions but requires the carboxyl terminus of STAT5b as well as STAT5b nuclear translocation and/or accumulation, suggesting that STAT5b is competing for a nuclear factor(s) necessary for NF-kB-mediated activation of target promoters.
  • STAT5 has been theorized to be the physiological substrate of the insulin receptor.
  • STAT5 has been shown to be the essential mediator of prolactin induced milk protein gene activation.
  • STAT6 has been shown to be activated by interleukin-4 (IL-4), IL- 13, and IL-3.
  • IL-4 interleukin-4
  • IL-3 interleukin-4
  • IL-4 interleukin-4
  • IL-3 interleukin-4
  • IL-3 interleukin-4
  • Inhibiting or reducing the effect of STAT may allow the selective inhibition of a variety of molecules involved in the immune response or nuclear receptor associated disorders.
  • the present invention may counter act in part the stimulatory effect of IL-I ( ⁇ and ⁇ ), IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-I l, IL-12, IL-13, growth hormone (GH), prolactin (PRL), erythropoietin (EPO), thrompopoietin, and granulocyte-macrophage colony-stimulating factor (GM-CSF), epidermal growth factor (EGF), NF-kB, tumor necrosis factore (TNF) and the like.
  • the present application will have particular utility in the prevention of inflammation which occurs via stimulation by cytokines or other stimuli such as but not limited to exposure to a microbe, microbial sample or antigen.
  • the present invention also includes a pharmaceutical or cosmetic composition capable of inducing degradation of a transcription factor, nuclear receptor or STAT transcription factor protein and thereby preventing or treating causes or symptoms of disease conditions that are, at least in part, affected by the activity of the transcription factor or nuclear receptor.
  • Transcription factors of interest include steroid hormone receptors, thyroid hormone receptor- like factors, STAT transcription factors, heat shock factors, and the like.
  • Transcription factors of especial interest include steroid hormone receptors, such as androgen receptors, progesterone receptors, estrogen receptors (a and ⁇ ), glucocorticoid receptors, peroxisome proliferator-activated receptor (PPAR), 9-cis retinoic acid receptors (RXR), trans-retinoic acid receptors (RAR), and orphan steroid hormone receptors.
  • Steroid hormone receptors of particular interest include, but are not limited to, the androgen receptor and the progesterone receptor.
  • Compositions of particular interest are those capable of enhancing or inducing specific degradation of a steroid hormone receptor, such as, but not limited to, the androgen receptor, estrogen receptor, or progesterone receptor.
  • compositions of the invention may affect their activity by means of any one or more suitable mechanisms to induce or enhance the degradation of a transcription factor.
  • suitable mechanisms and non-limiting embodiments illustrating these mechanisms, are described in detail above in the section headed, "I. METHOD FOR ENHANCING DEGRADATION OF A TRANSCRIPTION FACTOR".
  • compositions of the invention may include one or more active compounds or effective variations thereof in a suitable carrier.
  • Any suitable active compounds may be of use, if they are capable of enhancing or inducing degradation of the transcription factor of interest.
  • these compounds are effective at enhancing or inducing degradation of the transcription factor of interest at physiologically acceptable levels, such as at levels that do not cause undesirable side effects or toxicity.
  • the active compound can optionally include one or more elements that provide additional benefits, such as improved stability, solubility, or delivery specificity.
  • Such elements can include peptides, polypeptides, proteins, carbohydrates, nucleic acids, lipophilic moieties, hydrophilic moieties, particulates, matrices, or combinations thereof.
  • the active compound can be linked, covalently or non- covalently, to a hydrophilic moiety (such as a phosphate or sulphate group or a carbohydrate or a chelating molecule) to improve solubility in aqueous buffers or bodily fluids, hi another example, the active compound or active fragment thereof may be treated to reduce toxicity or cytotoxicity.
  • the active compound can be linked, covalently or non- covalently, to a peptide or other moiety that protects the active compound from premature degradation, or to an antibody or other specific binding agent that specifically targets a desired tissue or cell type and thus improves delivery of the active compound to that specific tissue or cell type.
  • the active compound can be encapsulated or embedded in a liposome, a particulate, a matrix, a gel, a polymer, or the like, to improve stability or to enhance delivery.
  • Nonlimiting examples of suitable compounds are curcumin derivatives or analogues thereof capable of enhancing degradation of the desired nuclear receptor or STAT transcription factor protein.
  • curcumin analogues have been identified in U.S. Patent No. 6,790,979, which is herein incorporated by reference in its entirety.
  • the curcumin analogues JC9 and JC 15 are effective at enhancing degradation of a nuclear receptor when provided at pharmaceutically acceptable levels.
  • Suitable carriers of use in the compositions of the invention include diluents, excipients, or carrier materials, selected according to the intended form of administration and consistent with conventional pharmaceutical or cosmetic practice.
  • suitable carriers include, but are not limited to, water, physiological saline, phosphate-buffered saline, a physiologically compatible buffer, saline buffered with a physiologically compatible salt, a water-in-oil emulsion, and an oil-in-water emulsion, an alcohol, dimethylsulfoxide, dextrose, mannitol, lactose, glycerin, propylene glycol, polyethylene glycol, polyvinylpyrrolidone, lecithin, albumin, sodium glutamate, cysteine hydrochloride, and the like, and mixtures thereof.
  • Suitable carriers can also include appropriate pharmaceutically acceptable antioxidants or reducing agents, preservatives, suspending agents, solubilizers, stabilizers, chelating agents, complexing agents, viscomodulators, disintegrating agents, binders, flavoring agents, coloring agents, odorants, opacifiers, wetting agents, pH buffering agents, and mixtures thereof, as is consistent with conventional pharmaceutical practice ("Remington: The Science and Practice of Pharmacy", 20th edition, Gennaro (ed.) and Gennaro, Lippincott, Williams & Wilkins, 2000).
  • compositions of the present invention can be formulated and provided as is convenient, hi non-limiting examples, compositions may be formulated as dissolvable solids, solutions, suspension, liposome preparations, and the like, and provided to the cells by manual or automated delivery (such as by pipette, syringe, pump, auto-injector, and the like).
  • compositions of the present invention can be formulated and provided in any formulation suitable to the intended form of administration and consistent with conventional pharmaceutical practice ("Remington: The Science and Practice of Pharmacy", 20 th edition, Gennaro (ed.) and Gennaro, Lippincott, Williams & Wilkins, 2000).
  • suitable formulations include tablets, capsules, syrups, elixirs, ointments, creams, lotions, sprays, aerosols, inhalants, solids, powders, particulates, gels, suppositories, concentrates, emulsions, liposomes, microspheres, dissolvable matrices, sterile solutions, suspensions, or injectables, and the like.
  • injectables can be prepared in conventional forms either as liquid solutions or suspensions, as concentrates or solid forms suitable for solution or suspension in liquid prior to injection, or as emulsions.
  • compositions of the present invention can be formulated and administered systemically or locally.
  • Techniques for formulation and administration can be found in "Remington: The Science and Practice of Pharmacy” (20 th edition, Gennaro (ed.) and Gennaro, Lippincott, Williams & Wilkins, 2000).
  • Suitable routes of administration can include oral, intestinal, parenteral, transmucosal, transdermal, intramuscular, subcutaneous, transdermal, rectal, intramedullary, intrathecal, intravenous, intraventricular, intraatrial, intraaortal, intraarterial, or intraperitoneal administration.
  • compositions of the present invention can be administered to the subject by a medical device, such as, but not limited to, implantable devices, biodegradable implants, patches, and pumps.
  • a medical device such as, but not limited to, implantable devices, biodegradable implants, patches, and pumps.
  • the compositions may be formulated to include a dissolvable or nondissolvable matrix or medium (for example, a coating, membrane, film, impregnated matrix, polymer, sponge, gel, or porous layer on or within the medical device) to permit the release of the active compound or compounds over a specified period of time.
  • the present invention also includes methods of treatment of a variety of transcription factor-associated diseases or conditions by inducing the degradation of a transcription factor such as a nuclear receptor.
  • Diseases or conditions of interest include any that are associated with a specific transcription factor or nuclear receptor, and that have causes or symptoms that may be prevented, reduced, or reversed by degradation of the transcription factor or nuclear receptor in question.
  • Transcription factors of interest include steroid hormone receptors, thyroid hormone receptor-like factors, STAT transcription factors, heat shock factors, and the like.
  • Transcription factors of especial interest include steroid hormone receptors, such as androgen receptors (AR), progesterone receptors (PR), estrogen receptors (ER ⁇ and ER ⁇ ), glucocorticoid receptors (GR), peroxisome proliferator-activated receptor (PPAR), retinoid X receptors (RXR), retinoid A receptors (RAR) and orphan steroid hormone receptors.
  • Steroid hormone receptors of particular interest include, but are not limited to, the androgen receptor and the progesterone receptor.
  • Subjects can include human subjects of any age, sex, or physical condition.
  • Subjects of particular interest include human subjects diagnosed as having, or as being at risk for, a disease condition or conditions that are, at least in part, affected by the activity of a nuclear receptor (such as, but not limited to, a steroid hormone receptor).
  • subjects may be humans having or at risk of developing a disease, disorder or medical condition associated with expression or activity of a nuclear receptor.
  • subjects may be humans having or at risk of developing a disease or disorder associated with the activity of a nuclear receptor such as the androgen receptor (AR).
  • AR androgen receptor
  • Examples of medical conditions or diseases include, but not limited to, male infertility, Kennedy disease, prostate cancer, breast cancer, liver cancer, bladder cancer, benign prostate hyperplasia, acne, baldness, and hirsutism.
  • the medical condition may result from increased activity or expression of the nuclear receptor pathway or may be due to a mutation in the nuclear receptor activation pathway.
  • Kennedy disease is caused by an abnormally elongated androgen receptor.
  • Modulation of AR function or activity by enhancing or inducing AR degradation by a method of the present invention may serve as a means to prevent the development and progression of prostate cancer and to remedy other androgen-related disorders.
  • Use of the method of the present invention to enhance or induce or induce degradation of the androgen receptor may also have utility in the treatment of wounds, such as by improving wound healing, especially in aging men or a diabetic patient.
  • the method of the invention can be used to enhance or induce degradation of the progesterone receptor to prevent conception in females (such as women of child-bearing age).
  • compositions, cosmetics, pharmaceuticals and methods of the present invention may be utilized to prevent or treat a variety of skin disorders or other medical conditions.
  • the present invention may be useful in the treatment or prevention of Acne, Acne keloidalis nuchae, Acne necrotica, Acne urticata, Acne Vulgaris, Actinic Keratoses, Acute Febrile Neutrophilic Dermatosis, Aging skin, Allergic contact dermatitis, Alopecia areata, Androgenetic alopecia, Angioedema, Anthralin, Atopic dermatitis, Bacterial skin infections, Boils, Botulinum toxin, Bowen's disease, Brown spots, Chronic superficial scaly dermatosis, Cysts, Dermatitis, Dermatitis herpetiformis, Dermatofibromas, Dermographism, Eczema, Eczema Vaccinatum, Erythrodermic Psoriasis, Facial Rejuvenation,
  • the nuclear receptor of interest can be a normal nuclear receptor or a mutant nuclear receptor, where the mutation may be associated with disease symptoms or causes.
  • Kennedy Disease is one of a group of hereditary neuropathies (including
  • polyglutamine-expanded AR is structurally altered so as to be resistant to proteolysis, and may give rise to a possibly post- translationally misprocessed, denaturation- and proteolysis-resistant fragment that accumulates in the nucleus of the affected cells and may cause the observed pathogenic effects
  • Short polyglutamine repeats have also been implicated in an increased risk of developing prostate cancer (Lee and Chang (2003)), and may be associated with an increased risk of infertility (defective spermatogenesis) and undermasculinization in at least some male populations (Yong et al. (2003) Hum. Reprod. Update, 9(1): 1-7).
  • Kennedy Disease is characterized by neuronal intranuclear inclusions in the affected neurons. These inclusions contain an N-terminus truncated form of the mutant nuclear receptor (the androgen receptor, AR), and are associated with ubiquitin and proteosome components, suggesting that the polyglutamine-expanded AR is not adequately degraded by the proteosome and accumulates in the inclusions as a result (Bailey et al. (2002) Human MoI. Genetics, 11 :515-523).
  • chaperone proteins hi a cell culture model of Kennedy disease, chaperone proteins have been shown to increase the solubility of the mutant AR and thus enhance its degradation by the proteosome (Bailey et al. (2002) Human MoI.
  • Enhancing or inducing degradation of the mutant nuclear receptor (the androgen receptor) by a method of the present invention may be useful in preventing or reducing the pathogenic effects associated with the mutant nuclear receptor.
  • Such degradation can be accomplished by any suitable mechanism, such as, but not limited to, repair of the mutant AR to allow correct degradation, molecular chaperone enhancement of mutant AR solubilization and subsequent degradation, and prevention of overexpressed AR from forming aggregates.
  • This approach may have therapeutic value in other polyglutamine diseases characterized by the accumulation of neuronal intranuclear inclusions. It has been reported that, among the elderly, males heal acute wounds more slowly that do females.
  • Any suitable mechanism may be used to induce or enhance the degradation of a nuclear receptor. Suitable mechanisms, and non-limiting embodiments illustrating these mechanisms, are described in detail above in the section headed, "I. METHOD FOR ENHANCING DEGRADATION OF A TRANSCRIPTION FACTOR".
  • EXAMPLE 1 OVERVIEW OF ENHANCING DEGRADATION OF A TRANSCRIPTION FACTOR
  • any mechanism that enhances degradation of the nuclear receptor of interest can be used, including, but not limited to, interfering with translocation of the nuclear receptor into the nucleus or retaining the nuclear receptor in the cytoplasm of a cell, exposing a motif within the nuclear receptor able to induce protease activity, increasing activity of a protease capable of specifically degrading the nuclear receptor, inhibiting the stabilization of a nuclear receptor, reducing the solubility of the nuclear receptor, activating a pathway able to degrade the nuclear receptor, increasing ubiquination of the nuclear receptor, increasing phosphorylation of the nuclear receptor by an appropriate kinase, inducing apoptosis, or reducing an interaction between a nuclear receptor and a cofactor able to stabilize the nuclear receptor.
  • the nuclear receptor of interest is the steroid hormone receptor, the androgen receptor.
  • assays can be used to detect down regulation of nuclear receptor transcriptional activity and therefore degradation of the nuclear receptor of interest, or to detect the downstream effects of such degradation.
  • assays used to detect the down regulation of the androgen receptor may be used at least in part to detect degradation of the androgen receptor.
  • Non-limiting examples of such methods and assays, as applicable to the androgen receptor, are described in general below.
  • proteins are transferred from the gel to a nitrocellulose membrane following standard Western blot procedures.
  • the membrane is blocked with a suitable blocking agent (such as 10% non-fat milk in phosphate-buffered saline supplemented with 0.1% Tween-20 (PBST)) overnight.
  • PBST phosphate-buffered saline supplemented with 0.1% Tween-20
  • the membrane is incubated with a suitable primary antibody specific for human AR (for example, anti-human AR from BD-PharMingen) at 4 degrees Celsius overnight or at room temperature for 2 hours.
  • the membrane is rinsed with PBST three times, 10 minutes each time, and then incubated with an appropriate secondary antibody (for example, an enzyme- labelled secondary antibody, such as horseradish peroxidase-conjugated secondary antibody) for 1 hour at room temperature.
  • an enzyme- labelled secondary antibody such as horseradish peroxidase-conjugated secondary antibody
  • the membrane is rinsed with PBST, and a suitable visualization procedure is used to detect the secondary antibody (for example, horseradish peroxidase can be detected with a colorimetric substrate or by a chemiluminescent substrate such as that provided by the enhanced chemiluminescence (ECL Plus) kit from Amersham).
  • the secondary antibody signal as a measure of the amount of androgen receptor protein on the blot, can be normalized to the total amount of protein loaded for each sample by stripping the membrane following the manufacture's recommendations and re-incubating the membrane with an appropriate antibody (such as an antibody to beta-actin, Sigma). Quantification of the protein signals can be carried out by densitometry, using appropriate software (ImageJ software from the National Institutes of Health).
  • Non-limiting examples of compounds that were tested for their ability to degrade AR include curcumin derivatives and analogues, whose structures and preparations are described in Ohtsu et al. (2002), J. Med. Chem., 45:5037-5042 and Ohtsu et al. (2003) Bioorg. Med. Chem., 11 :5083-5090, which are incorporated by reference in their entirety herein.
  • the compounds JC 15 and JC9 were tested on cultured cells.
  • JC 15 is a synthetic curcumin derivative bearing an ethoxycarbonylethyl moiety on carbon 4 of the conjugated beta-diketone system and has the structure 7-(4-hydroxy-3-methoxyphenyl)-4-[3- (4-hydroxy-3-methoxyphenyl)acryloyl]-5-oxo-hept-6-enoic acid ethyl ester.
  • JC9 is another synthetic curcumin derivative (trivial name, dimethylcurcumin), and has the structure 5- hydroxy-1, 7-bis(3 ,4-dimethoxyphenyl)-l,4,6-heptatrien-3 -one; it is obtainable, for example, by permethylation of natural curcumin with diazomethane.
  • EXAMPLE 2 ENHANCING DEGRADATION OF A NUCLEAR RECEPTOR
  • a compound known to enhance degradation of the nuclear receptor, androgen receptor (AR) was examined for its effects on AR activity and on cell proliferation.
  • AR nuclear receptor
  • AR plays a significant role in stimulating prostate cancer cell proliferation, and thus, modulation of AR activity by AR degradation could serve as a useful means to delay or control prostate cancer progression.
  • Prostate cancer cells, non-prostate tumor cells, and normal cells can be used in this transient transfection assay, which measures transactivation of the androgen receptor (AR) and can be used to detect a reduction of AR activity caused by AR degradation.
  • AR androgen receptor
  • CV-I green monkey kidney cells lacking an androgen receptor
  • LNCaP cells prostate cancer cells containing an endogenous mutant AR
  • ARE androgen response element
  • CV-I and LNCaP cells obtained from ATCC, Manassas, VA were maintained in Richter's Improved MEM Insulin (RPMI) medium supplemented with penicillin (25 units/millilter), streptomycin (25 micrograms/milliliter), and 10% heat- inactivated fetal bovine serum (FBS).
  • RPMI Richter's Improved MEM Insulin
  • FBS heat- inactivated fetal bovine serum
  • the cells were co-transfected with the SuperFect Transfection Reagent (Qiagen) and DNA mixtures consisting of an MMTV- luciferase reporter plasmid containing an AR-binding element.
  • the plasmid pRL-TK was used as an internal control for transfection efficiency.
  • the medium was changed to RPMI medium supplemented with 10% charcoal-stripped serum.
  • the cells were treated with the test compound at varying concentrations (from 0 uM to 7.5 uM), in the presence or absence of 1 nanomolar dihydrotestosterone (DHT), and incubated for a further 20 hours.
  • DHT nanomolar dihydrotestosterone
  • JC9 was capable of inhibiting DHT-induced androgen receptor transcriptional activation in a does dependent manner.
  • LNCaP cells express an endogenous mutant AR that is found in prostate cancer patients.
  • This clinically relevant cell model was used to study JC9's effect in suppressing prostate cancer cell growth.
  • Cells were plated at a density of 6.3 x 10 4 cells/well in 6-well tissue culture dishes. Two days later, the complete medium was aspired and 10% charcoal/dextran-treated (hormone depleted) serum-containing medium was added. Test compounds, JC9 and hydroxyfiutamide (HF) were then added to the medium at a final concentration of 5 ⁇ M with or without 1 nM of DHT. For vehicle control, the same amount of DMSO was added.
  • the medium was aspired once per day, and fresh medium containing test compounds and/or DHT was added. At designated times, a portion of cells was harvested by trypsinization, and cell count was performed using a hemacytometer.
  • JC9's effect on prostate cancer cell growth was further assessed in the presence of a prostatic androgen, DHT.
  • DHT a prostatic androgen
  • JC9 still displayed a good potency to decrease LNCaP cell growth; the magnitude of cell growth decrease in the presence or absence of DHT was actually comparable.
  • HF at the same test concentration, exerted a moderate activity to repress DHT-stimulated cell growth; this finding is in line with the previously reports that HF is a weak antiandrogen against LNCaP mutant AR.
  • JC9 can effectively nullify prostate cancer cell growth in the presence or absence of male hormones. Since JC9 appears to be more potent than HF, JC9 may have potential to be developed into a drug candidate for prostate cancer disease management.
  • AR is the key factor in regulating prostate cancer cells' response to androgens.
  • JC9 influences the steady-state level of AR.
  • LNCaP cells were treated with 5 ⁇ M of JC9 as mentioned above.
  • cell lysate was prepared for Western blot analysis following the previously described conditions.
  • a color detection method was subsequently employed to examine AR and actin protein signals in the membranes, and the resultant protein signals were quantitated by densitometry.
  • FIGURE 2B normalized AR signals are reported, which were expressed as a percentage of baseline (Day 0) value.
  • LNCap human prostate tumor cells are xenografted by subcutaneous injection (2 x 10 6 per site) into nude mice. Mice are subsequently treated by intraperitoneal injection of either the vehicle solution alone as a control or the test compounds (JC9 )) at a dose of 100 milligrams per kilogram body weight, three times a week for 7 weeks. Tumor volume is measured twice a week over the next 7 weeks. Treatment with compounds such as JC9 or JC 15 for a sufficient period of time (for example, from between 2 weeks to a few months) is expected to result in a significantly reduced rate of tumor growth.
  • EXAMPLE 3 SPECIFICITY OF STEROID HORMONE RECEPTOR DEGRADATION IN DIFFERENT CELL LINES
  • Cells were incubated with JC 15 for 24 or 48 hours, and were lysed in 250 microliters of Ix SDS/PAGE loading buffer. Approximately 40 micrograms of total cellular protein was loaded in each lane of a pre-cast gel (NuPAGE, Invitrogen). Protein separation and transfer were performed following the manufacturer's instructions.
  • PSA levels were also observed to be decreased by JC 15 treatment.
  • PSA expression is regulated primarily by the androgen receptor, which induces PSA expression through androgen response element-containing enhancer elements in the PSA promoter (Heinlein & Chang (2004)), and the observed reduction in PSA levels is consistent with the observed decrease in AR levels.
  • the Western blots of T47D cell lysates are shown in FIGURE 4. Incubating T47D cells with JC9 (5 or 10 micromolar) in the presence or absenec of estrodial (E2) decreased the cellular concentrations of AR but not other receptor proteins.
  • EXAMPLE 4 ENHANCING DEGRADATION OF A TRANSCRIPTION FACTOR IN THE PRESENCE OF A PROTEIN SYNTHESIS INHIBITOR
  • FIGURE 5 A representative western blot from an experiment is depicted in FIGURE 5.
  • a reduction of endogenous AR concentration in the control cells was detected within about 2-3 hours of treatment with cycloheximide, suggesting that de novo AR synthesis contributes to the steady- state level of this receptor.
  • the observed reduction of existing AR protein indicates that the test compound (JC9) enhanced or increased the degradation of existing AR protein (and thus decreased AR activity) within 4 hours or less.
  • EXAMPLE 5 DEGRADATION OF A MUTANT ANDROGEN RECEPTOR
  • SMBA spinobulbar muscular atrophy
  • Monkey kidney COS-I cells were plated at a density of 3 x 10 4 cells per 0.5-milliliter volume onto alcohol-cleaned and sterilized cover slips placed in 35 -millimeter suspension culture dishes containing Dulbecco's modified Eagle's (DME) medium containing 10% FBS.
  • DME Dulbecco's modified Eagle's
  • the cells were transfected with plasmids containing either the Ql 9 (plasmid GFPARQ 19) or Q49 (plasmid GFPARQ49) mutant androgen receptor and green fluorescent protein (GFP) as a reporter.
  • the medium was changed again to fresh CD/DME medium (with or without 1.5 nanomolar DHT), and either vehicle or JC9 added (final concentration 5 micromolar).
  • the medium was removed and the cells fixed with 1% formaldehyde in phosphate-buffered saline (PBS) for 1 hour at room temperature. The formaldehyde was removed and the fixed cells washed with PBS three times, and the coverslips then allowed to dry. Coverslips were marked to indicate the treatment scheme and hydrophobic circles made with a wax pencil around the cells.
  • PBS phosphate-buffered saline
  • Each coverslip was stained with 200 microliters of propidium iodide (0.7 micrograms per milliliter in water) for 5 minutes at room temperature, then rinsed three times with PBS.
  • the coverslips were air-dried, mounted on slides with a glycerol-based mounting agent, and stored at 4 degrees Celsius if necessary prior to observation with fluorescence microscopy. Representative micrographs showing COS-I cells transfected with the
  • GFPARQ49 plasmid are depicted in FIGURE 6. As shown in the micrographs, transfected cells expressed the plasmid as shown by the fluorescent reporter protein GFP. Control cells contained large amounts of fluorescent inclusions or aggregates. Cells that had been treated with JC9 contained substantially smaller amounts of fluorescent inclusions, suggesting that the expressed mutant Q49 androgen receptor was degraded by JC9 treatment.
  • EXAMPLE 6 TREATMENT OF AN ANDROGEN-RELATED DISORDER IN HUMAN SUBJECTS BY DEGRADATION OF THE ANDROGEN RECEPTOR
  • This example describes the treatment of a nuclear receptor-related disorder (acne vulgaris) in a subject by degradation of the nuclear receptor (the androgen receptor).
  • Acne vulgaris commonly known simply as acne, is a red skin rash that typically affects the face, chest, and back of teenaged and young adult humans of either sex, though it can occur at any age and on other body areas (see, for example, J. C. Harper and J. Fulton, Jr. (2003), "Acne Vulgaris", electronically available at www.emedicine.com/derm/topic2.htm, accessed 23 April 2004).
  • Acne affects nearly all people at some point in their life, and can cause permanent scarring and emotional distress and low self-esteem, as well as potentially leading to more severe health problems, such as skin infections.
  • the androgen receptor which is expressed in the basal cells and glandular cells of sebaceous glands, has a skin distribution that is similar between males and females (Blauer et al. (1991) J. Investig. Dermatol, 97:264- 268). In the skin, the androgen receptor stimulates terminal sebocyte differentiation and the production of sebum.
  • Common treatments for acne often have undesirable side effects. For example, topical retinoids can lead to sun sensitivity, antibiotics may result in antibiotic resistance, and benzoyl peroxide can cause contact dermatitis.
  • topical retinoids can lead to sun sensitivity
  • antibiotics may result in antibiotic resistance
  • benzoyl peroxide can cause contact dermatitis.
  • a basic carrier formulation was prepared by mixing two solutions: (1) a water-based solution containing aristoflex avc, Osmocide, Tween 20, and water; and (2) an oil-based solution containing isopropyl myristate, coconut detergent, ethylparaben, isobutylparaben, methylparaben, and propylparaben.
  • the test compounds (JC 15 or JC9) were added to the cream to a final concentration of 1 to 2.5 micromolar, as needed.
  • EXAMPLE 7 REDUCTION OF SEBACEOUS GLANDS IN RATS BY DEGRADATION OF THE ANDROGEN RECEPTOR
  • test compounds JC 15 and JC9 which were shown in the previous examples to ameliorate the effects of the androgen receptor-activated pathway, specifically by inducing degradation of the androgen receptor, and to be effective in treating acne in human subjects, were used to reduce sebaceous glandular lobe size in an animal model. Effective reduction of sebaceous glands by topical treatment may be useful in treating skin conditions such as acne. Fuzzy rats were used in this animal model as described in Ye et al. (1997) Skin Pharmacol, 10:10288-10297, which is incorporated by reference in its entirety herein.
  • Topical creams were prepared as described in EXAMPLE 6.
  • the test creams contained JC9 (25 micromolar) or JC 15 (1 micromolar); a control cream with only vehicle added was also prepared.
  • the test or control creams were applied using a cotton swab to the dorsal skin of the animal, once daily, over a period of 8 weeks. Animals were then sacrificed and skin samples collected for microscopic examination. Commercial hair remover was applied to the dorsal surface of the euthanized animals. After 5 minutes, the hair remover and hairs were removed with a tissue. The area was thoroughly cleaned with 75% isopropyl alcohol.
  • FIGURE 9A-E the size of the sebaceous glandular lobe is approximately twice in male Fuzzy rats as in females.
  • Topical treatment with the vehicle only (control cream) did not produce a significant change in male rats.
  • Topical treatment of male rats with the test compounds JC 15 and JC9 resulted in a reduction in the size of the sebaceous glandular lobe and duct, with the decrease caused by JCl 5 approximately equivalent to the decrease caused by castration.
  • EXAMPLE 8 TREATMENT OF ANDROGEN-INDUCED ALOPECIA IN AN ANIMAL MODEL BY DEGRADATION OF THE ANDROGEN RECEPTOR
  • This example describes the treatment of a nuclear receptor-related disorder in a subject by degradation of the nuclear receptor, hi this example, the nuclear receptor is the steroid hormone receptor, the androgen receptor.
  • the nuclear receptor-related disorder is alopecia (hair loss or baldness), which is known to be affected by the androgen receptor, hi this example, the test compound JC9, which was shown in the previous examples to ameliorate the effects of the androgen receptor-activated pathway, specifically by inducing degradation of the androgen receptor, is used to treat hair loss in an animal model.
  • mice C57BL/6J mice were used in this animal model for hair loss and regrowth (Uno et al. (1990) J. Cutaneous Aging & Cosm. Derm., 1 :193, which is incorporated by reference in its entirety herein).
  • Six-week-old male mice (6 to 7 animals per group) were shaved with an electric clipper, and then treated with a hair-removal cream for 1 to 2 minutes. Animals that were found to have a dark skin color after shaving, indicating that they were in anagen phase where there is active growth of hair follicles, were excluded from the study.
  • mice One day after hair removal, a first group of animals each received 100 microliters of a 1% testosterone solution in ethanol, applied topically to the shaved area, once each morning for twenty consecutive days. A second group of animals each received 100 microliters of vehicle (ethanol) alone, applied topically to the shaved area, once each morning for twenty consecutive days. The first group of mice (testosterone-treated) were divided further into a control group and a treatment group.
  • each mouse in the control group received 100 microliters of a control solution (60% ethanol, 20% propylene glycol, and 20% water) and each mouse in the treatment group received 100 microliters of the test compound, JC9 (0.02% in the same 60% ethanol, 20% propylene glycol, and 20% water solution), applied topically to the shaved area, once each afternoon for twenty consecutive days. Hair re-growth in the shaved areas was observed and photographed at 12, 15, 18, 21 , and 24 days after beginning of the topical treatments.
  • a control solution 50% ethanol, 20% propylene glycol, and 20% water
  • JC9 0.02% in the same 60% ethanol, 20% propylene glycol, and 20% water solution
  • mice that were shaved and then received topical morning applications of testosterone and afternoon applications of the control solution only showed little or no re-growth of hair in the shaved areas after 20 days of treatment (FIGURE 10A).
  • Mice that were shaved and then received topical morning applications of testosterone and afternoon applications of JC9 showed rapid re-growth of hair in the shaved areas from day 10 to day 20 (FIGURE 10B).

Landscapes

  • Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Endocrinology (AREA)
  • Reproductive Health (AREA)
  • Dermatology (AREA)
  • Diabetes (AREA)
  • Epidemiology (AREA)
  • Gynecology & Obstetrics (AREA)
  • Urology & Nephrology (AREA)
  • Pregnancy & Childbirth (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Cosmetics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

L'invention concerne des procédés, des compositions, des compositions pharmaceutiques et cosmétiques destinées à améliorer la dégradation d'un récepteur nucléaire (NR) ou d'une protéine de facteur de transcription (STAT). Ces procédés, ces compositions et ces produits cosmétiques et pharmaceutiques peuvent être utilisés afin de prévenir ou de traiter des troubles ou des états médicaux qui sont au moins en partie affectés par un trajet d'activation de récepteur nucléaire ou un trajet d'activation de STAT.
EP05806820A 2004-09-02 2005-09-02 Procedes et compositions destines a ameliorer la degradation de facteurs de transcription de recepteur nucleaire et utilisations correspondantes Withdrawn EP1804806A2 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US60667804P 2004-09-02 2004-09-02
US11/045,181 US20050209205A1 (en) 2004-01-28 2005-01-27 Enhancement of anti-androgenic activity by a combination of inhibitors targeting different steps of a steroid-dependent gene activation pathway and uses thereof
PCT/US2005/031447 WO2006029040A2 (fr) 2004-09-02 2005-09-02 Procedes et compositions destines a ameliorer la degradation de facteurs de transcription de recepteur nucleaire et utilisations correspondantes

Publications (1)

Publication Number Publication Date
EP1804806A2 true EP1804806A2 (fr) 2007-07-11

Family

ID=36036907

Family Applications (1)

Application Number Title Priority Date Filing Date
EP05806820A Withdrawn EP1804806A2 (fr) 2004-09-02 2005-09-02 Procedes et compositions destines a ameliorer la degradation de facteurs de transcription de recepteur nucleaire et utilisations correspondantes

Country Status (6)

Country Link
EP (1) EP1804806A2 (fr)
JP (1) JP2008540327A (fr)
AU (1) AU2005282624A1 (fr)
CA (1) CA2578159A1 (fr)
MX (1) MX2007002583A (fr)
WO (1) WO2006029040A2 (fr)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP5292569B2 (ja) * 2006-11-10 2013-09-18 国立大学法人名古屋大学 抗老化転写因子活性化剤及びその利用
US9000222B2 (en) 2007-01-08 2015-04-07 Androscience Corporation Compounds with (1E, 6E)-1,7-bis-(3,4-dimethoxyphenyl)-4,4-disubstituted-hepta-1,6-diene-3,5-dione structural scaffold, their biological activity, and uses thereof
US8236852B2 (en) 2007-01-08 2012-08-07 Androscience Corporation Compounds with (substituted phenyl)-propenal moiety, their derivatives, biological activity, and use thereof
US8710272B2 (en) 2007-01-08 2014-04-29 Androscience Corporation Compounds with (1 E, 6E)-1,7-bis-(3,4-dimethoxyphenyl)-4,4-disubstituted-hepta-1,6-diene-3,5-dione structural scaffold, their biological activity, and uses thereof
KR20100051808A (ko) 2007-07-31 2010-05-18 앤드로사이언스 코포레이션 안드로겐 수용체 분해 강화제를 포함한 조성물 및 탈모와 피부질환의 예방 또는 치료 방법

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2770842B1 (fr) * 1997-11-13 1999-12-17 Oreal Nouveaux composes derives de n-aryl 2-hydroxy alkylamides
US6790979B2 (en) * 2002-04-17 2004-09-14 University Of North Carolina At Chapel Hill Curcumin analogues and uses thereof
BRPI0407354A (pt) * 2003-02-11 2006-01-10 Warner Lambert Co Derivados de uréia e tiouréia

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2006029040A2 *

Also Published As

Publication number Publication date
MX2007002583A (es) 2007-09-14
AU2005282624A1 (en) 2006-03-16
WO2006029040A3 (fr) 2008-12-04
WO2006029040A2 (fr) 2006-03-16
CA2578159A1 (fr) 2006-03-16
JP2008540327A (ja) 2008-11-20

Similar Documents

Publication Publication Date Title
US20060084704A1 (en) Methods and compositions for enhancing degradation of nuclear receptor transcription factors and uses thereof
US9446127B2 (en) Compositions including androgen receptor degradation (ARD) enhancers and methods of prophylactic or therapeutic treatment of skin disorders and hair loss
Wang et al. Genistein suppresses psoriasis-related inflammation through a STAT3–NF-κB-dependent mechanism in keratinocytes
JP5424923B2 (ja) 乾癬及び他の皮膚疾患治療におけるシクロパミンの使用
CA2481499C (fr) Utilisation de cyclopamine pour traiter le psoriasis et autres troubles cutanes
DE69731302T2 (de) Verwendung von substanzen, die die östrogen wirkung fördern, zur behandlung von wunden
US20050209205A1 (en) Enhancement of anti-androgenic activity by a combination of inhibitors targeting different steps of a steroid-dependent gene activation pathway and uses thereof
WO2006029040A2 (fr) Procedes et compositions destines a ameliorer la degradation de facteurs de transcription de recepteur nucleaire et utilisations correspondantes
Ishikawa et al. Histamine induces human β-defensin-3 production in human keratinocytes
Karnam et al. Pharmacological blockade of HDAC3 accelerates diabetic wound healing by regulating macrophage activation
Guo et al. Magnolol may contribute to barrier function improvement on imiquimod‑induced psoriasis‑like dermatitis animal model via the downregulation of interleukin‑23
MX2008009442A (es) Uso de la minosina o un derivado de la misma, en el tratamiento de las manifestaciones cutaneas de la psoriasis y de los desordenes cutaneos relacionados con esta y una composicion cosmetica o farmaceutica que la comprende.
Meng et al. Apigenin ameliorates imiquimod-induced psoriasis in C57BL/6J mice by inactivating STAT3 and NF-κB
JP5697099B2 (ja) 掻痒抑制剤
He et al. Amygdalin ameliorates alopecia areata on C3H/HeJ mice by inhibiting inflammation through JAK2/STAT3 pathway
KR20160013226A (ko) 크리신을 포함하는 피부 보습용 또는 피부 염증 개선용 조성물
Schoepe Investigations of in vitro Test Systems for the Detection of Glucocorticoid-induced Skin Atrophy as a Tool in Drug Discovery

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20070326

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA HR MK YU

DAX Request for extension of the european patent (deleted)
R17D Deferred search report published (corrected)

Effective date: 20081204

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 31/343 20060101ALI20090202BHEP

Ipc: A61K 31/444 20060101ALI20090202BHEP

Ipc: A61K 31/4741 20060101AFI20090202BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20110401