WO1997031531A1 - Antiandrogenic agents and methods of identification thereof - Google Patents

Antiandrogenic agents and methods of identification thereof Download PDF

Info

Publication number
WO1997031531A1
WO1997031531A1 PCT/US1997/002871 US9702871W WO9731531A1 WO 1997031531 A1 WO1997031531 A1 WO 1997031531A1 US 9702871 W US9702871 W US 9702871W WO 9731531 A1 WO9731531 A1 WO 9731531A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
compound
ddtl
growth
prostate
Prior art date
Application number
PCT/US1997/002871
Other languages
French (fr)
Inventor
Alex Elbrecht
Jeffrey H. Toney
Original Assignee
Merck & Co., Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck & Co., Inc. filed Critical Merck & Co., Inc.
Publication of WO1997031531A1 publication Critical patent/WO1997031531A1/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • A61K31/167Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide having the nitrogen of a carboxamide group directly attached to the aromatic ring, e.g. lidocaine, paracetamol
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C233/00Carboxylic acid amides
    • C07C233/57Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of rings other than six-membered aromatic rings
    • C07C233/59Carboxylic acid amides having carbon atoms of carboxamide groups bound to carbon atoms of rings other than six-membered aromatic rings having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a hydrocarbon radical substituted by halogen atoms or by nitro or nitroso groups
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5044Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics involving specific cell types
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • G01N33/743Steroid hormones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/02Systems containing only non-condensed rings with a three-membered ring

Definitions

  • the present invention relates generally to a novel assay for identifying compounds with antiandrogenic activity as well as to antiandrogenic compounds identified from this assay.
  • the assay employs a hamster ductus deferens cell line (DDT1) ATCC CRL-1701 and ATCC CRL-12051 that is highly dependent on the addition of testosterone in synthetic serum-free media.
  • DDT1 hamster ductus deferens cell line
  • ⁇ > has been identified using the assay of the present invention as an antiandrogenic compound useful in the treatment and prevention of diseases of the prostate including prostatitis, benign prostatic hyperplasia (BPH) and prostatic carcinoma.
  • the present invention also provides for novel compositions employing compounds which exhibit antiandrogenic activity in the assay of the present invention.
  • the compounds which exhibit antiandrogenic activity in the assay of the present invention are useful in the systemic, including oral, and parenteral, including topical, treatment and prevention of diseases of the prostate including prostatic carcinoma, benign prostatic hyperplasia and prostatitis.
  • the compounds, including N-(4-chlorophenyl)-(Z,Z)-2,3- bis(cyclopropylmethylene)cyclopentane carboxamide, identified as androgen antagonists in the assay of the present invention may be used in combination with other active agents, for example, a 5 ⁇ -reductase inhibitor such as finasteride, epristeride, 17 ⁇ -N-(2,5- bis(trifluoromethyl))phenylcarbamoyl-4-aza-5ot-androst- 1 -en-3-one, or the compounds described in PCT publication WO 95/11254, or an ⁇ l - or ⁇ la-andrenergic receptor antagonist, or combinations of such other active agents with the androgen antagonist compound identified from the present assay, wherein such combinations would be useful in one or more of the above-mentioned methods of treatment or pharmaceutical compositions.
  • active agents for example, a 5 ⁇ -reductase inhibitor such as finasteride, epristeride
  • the DDT1 cell line has been reported to be an androgen- responsive cell line.
  • Norris et al. “Androgen Receptors in a Syrian Hamster Ductus Deferens Tumour Cell Line, " Nature 248:422-424 (1974).
  • Syms et al. "Glucocorticoid Effects on Growth, and Androgen Receptor Concentrations on DDTl MF-2 Cell Lines", J. Steroid Biochem., 28(2): 109-1 16 (1987) report that the DDTl -MF-2 smooth muscle tumor cell line contains receptors for and is differentially sensitive to androgens and glucocorticoids. They report that androgens stimulate growth, and glucocorticoids inhibit growth of the cell line.
  • the present invention provides for an assay to study compounds as potential anti- androgens in living cells.
  • Compounds identified as anti-androgens by the assay of the present invention are especially useful in the prevention and treatment of prostatic carcinoma, and they may also be useful in the treatment and prevention of other hyperandrogenic diseases such as acne vulgaris, seborrhea, female hirsutism, androgenetic alopecia, also called androgenic alopecia, which includes male and female pattern baldness, and benign prostatic hyperplasia.
  • hyperandrogenic diseases such as acne vulgaris, seborrhea, female hirsutism, androgenetic alopecia, also called androgenic alopecia, which includes male and female pattern baldness, and benign prostatic hyperplasia.
  • Benign prostatic hyperplasia and prostatic carcinoma are among the most common afflictions of aging men. Benign prostatic hyperplasia is often treated surgically with a procedure known as transurethral resection of the prostate (TURP). Other surgical procedures performed to release the obstruction of urine include incision or stents. Castration has also resulted in regression of prostatic enlargement.
  • Drug therapy for BPH has included alpha- 1 blockers which treat the symptoms of the disease by alleviating obstructive symptoms, but do not affect the underlying cause of the disease, the enlarged prostate gland.
  • Representative alpha- 1 blockers used in the treatment of BPH include: prazosin, terazosin, doxazosin, tamsulosin and alfuzosin. These drugs relax prostatic smooth muscle tone, decreasing intraurethral pressure without affecting bladder pressure. Common side effects of these agents are dizziness, headache and fatigue.
  • Finasteride (17 ⁇ -(N-tert-butylcarbamoyl)-4-aza-5 ⁇ - androst-l-ene-3-one), which is marketed by Merck & Co., Inc., under the tradename PROSCAR®, is an inhibitor of testosterone 5 ⁇ -reductase currently marketed for the treatment of benign prostatic hyperplasia.
  • a principal mediator of androgenic activity in the prostate is 5 ⁇ - dihydrotestosterone ("DHT"), formed locally in the prostate by the action of testosterone-5 ⁇ -reductase.
  • DHT dihydrotestosterone
  • Inhibitors of testosterone-5 ⁇ - reductase inhibit the conversion of testosterone (T) to DHT and serve to prevent or lessen symptoms of hyperandrogenic stimulation in the prostate. See especially United States Patent No.
  • Nonsteroidal antiandrogens such as flutamide and Casodex compete with DHT for androgen receptor sites in the prostate cells. These non-steroidal antiandrogens do not substantially change sexual potency and libido as the gonadotrophin releasing hormone agonists and progestogens do; however, these nonsteroidal antiandrogens often exhibit the undesirable tendency to feminize the male host (gynaecomastia) or initiate feed-back effects which would cause hyperstimulation of the testes.
  • GnRH Gonadotrophin-releasing hormone
  • LH leutinizing hormone
  • GnRH agonists are able to reduce the production of testosterone, induce shrinkage of prostate volume and reduce the severity of urinary symptoms of BPH.
  • these drugs have adverse effects such as impotence and flushing, which discourage a majority of patients from continuing with the drugs.
  • These androgen-suppressing agents are thus of inconsequential significance in BPH treatment, but are of major importance in the treatment of patients with advanced prostatic cancer.
  • Progestogens such as megestrol acetate, hydroxyprogesterone and medrogestone depress testosterone by inhibiting LH release and blocking androgen receptors, causing a reduction in prostatic volume.
  • Adverse effects such as decreased libido and impotence have kept progestogens from common use in BPH treatment.
  • WB2838 [3-Chloro-4-(2-amino-3- chlorophenyl)-pyrrole]: Non-steroidal androgen-receptor antagonist produced by A Pseduomonas" J. Antibiotics 46(9):1327-1333 (1993) describe the nonsteroidal androgen receptor antagonist labeled WB2838:
  • N-(4-chlorophenyl)-(Z,Z)-2,3-bis(cyclopropylmethylene) cyclopentane carboxamide is described by T. Wong "Synthesis nad Some Related Studies of Alkyl 2,3-bis(alkylidene)cyclopentance carboxylates", Ph.D. thesis, University of British Columbia, December, 1993. No use for this compound is described in the thesis.
  • Steroid hormones are involved in numerous aspects of cell growth and differentiation. In an effort to influence these processes chemists have developed analogs usually based on the four ring steroid nucleus. Nonsteroidal compounds interacting with steroid receptors are more rare and exhibit different pharmacological properties from their steroidal counterparts. We describe the use of androgen receptor binding assays and androgen-dependent DDT1 cells to identify and characterize novel antiandrogens, of particular interest are nonsteroidal antiandrogens.
  • FIGURE 1 is a graph of the results of two independent runs of the experiment described in Example 1. This graph shows the androgen dependent growth of DDT1 cells. Cells were plated in a 96 well plate at 2,000 cells/well in DFITS + 0.1 % fetal calf serum.
  • FIGURE 2 is a graph showing the effect of N-(4- chlorophenyl)-(Z,Z)-2,3-bis(cyclopropyl- methylene)cyclopentane carboxamide on the activation of the androgen receptor in CVl cells, as described in Example 4.
  • the filled triangles represent N-(4- chlorophenyl)-(Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentane carboxamide alone, the filled squares represent N-(4-chlorophenyl)- (Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentane carboxamide plus 10 nM dihydrotestosterone (DHT).
  • DHT dihydrotestosterone
  • the present invention relates to a method for determining the antiandrogenic action of a compound comprising:
  • DDT1 cells (c) measuring the growth of the DDT1 cells in the presence of the compound over a measured period of time.
  • the method for determining the antiandrogenic action of a compound comprises:
  • the DDT1 cells are grown in synthetic serum-free media.
  • the DDT1 cell line also referred to as the DDT1 MF-2 cell line, was an existing deposit with the American Type Culture Collection ATCC No. CCRL-1701 which has been re-deposited with the American Type Culture Collection (ATCC), 12301 Parklawn Drive, Rockville, Maryland, United States of America under the conditions of the Budapest Treaty as ATCC CRL-12051 on February 21, 1996.
  • the DDT1 smooth muscle cell line was cloned from cells derived from a leiomyosarcoma of the ductus deferens of a Syrian hamster (Mesocricetus auratus). It has maintained androgen and glucocorticoid receptors and alpha 1 and beta 2 adrenergic receptors.
  • the beta 2 adrenergic receptor is functionally coupled to adenylate cyclase.
  • Glucocorticoids regulate the expression of the c-sis proto- oncogene (inhibit). Growth is stimulated by androgens and inhibited by glucocorticoids.
  • the method of the present invention is carried out by comparing the rate of growth of DDT1 cells in the presence of the compound to the rate of growth of DDT1 cells in the absence of the compound.
  • a known volume of the solution containing the compound is added to the plated DDT1 cells.
  • the solution of the compound is of a known concentration.
  • the compound may be dissolved in a suitable solvent such as DMSO, ethanol, water or methylethyl ketone (MEK)>
  • a suitable solvent such as DMSO, ethanol, water or methylethyl ketone (MEK)>
  • MEK methylethyl ketone
  • the compound is dissolved in ethanol to form an ethanol solution.
  • a known volume of a known concentration of the ethanol solution is added.
  • the rate of growth of the DDT1 cells when a known volume of a known concentration of the compound in an ethanol solution cells is compared to the rate of growth of DDT1 cells when the same volume of ethanol is added.
  • the growth of the DDT1 cells is measured over the period of one hour.
  • the measurement of the growth of the DDT1 cells over the measured period of time is performed by counting the number of cells at the beginning and at the end of the period of time. This can be done by quantitating cells using a hemocytometer.
  • the measurement of the growth of the DDT1 cells over the measured period of time is performed by incubating the DDT1 cells with labeled thymidine and measuring the incorporation of the label as a measure of DNA synthesis.
  • the labeled thymidine is ⁇ H-thymidine
  • the incorporation of the tritium label is measured by incubating cells for a period of time, preferably overnight, and washing extensively to remove free ⁇ H-thymidine. Cells are then removed, preferably using trypsin, resuspended in media and the radioactivity quantitated, preferably by scintillation counting, for example using a Tri-Carb 2500TR Liquid Scintillation analyzer manufactured by Packard.
  • the measurement of the growth of DDT1 cells is measured by colorimetric analysis.
  • the growth may be measured by diluting 3-(4,5-dimethyl-thiazol-2-yl)-5-(3-carboxylmethoxy phenyl)-2- (4-sulfophenyl)-2H-tetrazolium into the medium and adding phenazine methosulfate, and measuring the absorbance at 490 nm.
  • the cells are plated in 96 well plates.
  • the assay is automated; i.e., the solutions are dispensed using a robotic device.
  • the tested compound is assayed to determine that the compound inhibits the binding of labeled dihydrotestosterone (DHT) to the androgen receptor (AR).
  • DHT labeled dihydrotestosterone
  • AR androgen receptor
  • the androgen receptor used in the assay can be obtained from tissue such as prostate or liver, preferably human tissue, or cloned from the known sequence, see Tilley et al., Proc. Nat'l. Acad. Sci. USA, 80:327-331 (1989).
  • the source of the human androgen receptor can be a cell line transfected with the human androgen receptor. Techniques for obtaining these transfected cell lines are well- known in the art and are preferred because of the uniformity of the receptor produced.
  • the androgen receptor is incubated with labeled DHT, particularly 3 H-DHT, and the compound to be tested or unlabelled DHT is added to the incubated receptor.
  • the unlabelled DHT may be run in a parallel experiment in order to quantitate the nonspecific binding.
  • the affinity for the androgen receptor is then calculated.
  • One method of calculating this affinity is to add an agent to remove free labeled DHT. This may be done by a charcoal technique by adding dextran-coated charcoal to the sample at 0.5 volume of the original assay volume, vortexing, and centrifuging at 3,000 x g for 15 minutes at 4°C. Bound labeled DHT (preferably 3H-DHT) is in the supernatant.
  • the bound androgen receptor- labeled DHT (preferably AR- ⁇ H-DHT) complexes can be precipitated using agents such as ammonium sulfate, and filtered through glass fiber membranes. After extensive washing, bound labeled DHT (preferably 3H-DHT) is measured. When ⁇ H-DHT is employed, the remaining label may be measured by scintillation counting of the glass fiber membranes, for example using a Tri-Carb 2500TR Liquid Scintillation analyzer manufactured by Packard. The compound may be tested for its ability to inhibit binding of DHT to the androgen receptor either before or after determining the antiandrogenic action of the compound according to the method of the present invention.
  • agents such as ammonium sulfate
  • bound labeled DHT preferably 3H-DHT
  • the remaining label may be measured by scintillation counting of the glass fiber membranes, for example using a Tri-Carb 2500TR Liquid Scintillation analyzer manufactured by Packard.
  • the compound
  • the present invention presents a convenient assay for monitoring the effect of compounds in a tissue culture system and demonstrates that the result of such an assay is the discovery of a nonsteroidal compound that acts as an antiandrogen.
  • the present invention relates to a method of treating or preventing diseases of the prostate comprising administering 0.001 to 200 mg per day of a compound capable of inhibiting andro gen- induced DDTl cell growth to a male human in need of such treatment.
  • the instant invention involves a method of treating and/or preventing BPH and prostatic carcinoma which comprises administering to a patient in need of such treatment a therapeutically effective amount of a compound which inhibits the testosterone induced growth of DDTl cells.
  • the compound which inhibits the testosterone-induced growth of DDTl cells is administered in a dosage amount between 0.001 to 200.0 mg/day.
  • the compound which inhibits the testosterone-induced growth of DDTl cells is administered in a dosage amount of from 0.01 to 50.0 mg/day, and in a sub-class of this embodiment, the compound which inhibits the testosterone-induced growth of DDTl cells is administered in a dosage amount of about 0.1 to 5.0 mg/day.
  • Compounds which inhibit the testosterone-induced growth of DDTl cells can be determined by employing the assay described in Example 1.
  • the methods of treating and preventing benign prostatic hyperplasia and prostatic carcinoma comprise administration to a patient in need of such treatment of a compound which inhibits the testosterone-induced growth of DDTl cells.
  • this compound is a non-steroidal compound.
  • Preferred compounds that may be employed in the present invention include N-(4-chIorophenyl)-(Z,Z)-2,3-bis(cyclopropyl- methylene)cyclopentane carboxamide of structural formula (I):
  • the nonsteroidal compound N-(4-chlorophenyl)-(Z,Z)-2,3- bis(cyclo ⁇ ropylmethylene)cyclopentane carboxamide which antagonizes the action of testosterone on DDTl cells but exhibits little or no effect on cell growth by itself.
  • This compound also blocks the binding of ⁇ H- DHT to the human androgen receptor expressed in stable transfectants of CHO cells at doses comparable to those that antagonize testosterone in DDTl cells.
  • This compound may thus represent a novel class of nonsteroidal antiandrogens which show promise in the treatment of prostate cancer.
  • the present invention has the objective of providing methods of treating and preventing diseases of the prostate including BPH and prostatic carcinoma by systemic, oral, parenteral or topical administration of a compound which inhibits the testosterone-induced growth of DDTl cells in a dosage amount between 0.001 to 200.0 mg/day, and more particularly, from about 0.01 to 50.0 mg/day, and most particularly 0.1 to 5.0 mg/day.
  • treating BPH is intended to include alleviating the obstructive symptoms of BPH, and slowing and/or reversing the growth of the prostate.
  • preventing BPH is intended to include preventing development of obstructive symptoms, and preventing the enlargement of the prostate.
  • the term “treating prostatic carcinoma” is intended to include slowing and/or stopping the growth of prostatic carcinoma.
  • the term “preventing prostatic carcinoma” is intended to include preventing the development of prostatic carcinoma in patients likely to develop prostatic carcinoma.
  • a compound which inhibits the testosterone- induced growth of DDTl cells may be co-administered with a 5 ⁇ - reductase 2 inhibitor, such as finasteride or epristeride; a 5 ⁇ -reductase 1 inhibitor such as 4,7 ⁇ -dimethyl-4-aza-5 ⁇ -cholestan-3-one, 3-oxo-4- aza-4,7 ⁇ -dimethy 1- 16 ⁇ -(4-chlorophenoxy)-5 ⁇ -androstane, and 3-oxo-4- aza-4,7 ⁇ -dimethyl- 16 ⁇ -(phenoxy)-5 ⁇ -androstane as disclosed in WO 93/23420 and WO 95/11254; dual inhibitors of 5 ⁇ -reductase 1 and 5 ⁇ - reductase 2 such as 3-oxo-4-aza-17 ⁇ -(2,5-trifluoromethylphenyl- carbamoyl)-5 ⁇ -androstane as disclosed in WO 95/07927; nonsteroidal antiandrog
  • the present invention also has the objective of providing suitable systemic, oral, parenteral and topical pharmaceutical formulations for use in the novel methods of treatment of the present invention.
  • compositions containing as an active ingredient a compound which inhibits the testosterone-induced growth of DDTl cells can be administered in a wide variety of therapeutic dosage forms in conventional vehicles for systemic administration.
  • the compounds can be administered in such oral dosage forms as tablets, capsules (each including timed release and sustained release formulations), pills, powders, granules, elixirs, tinctures, solutions, suspensions, syrups and emulsions.
  • compositions can be provided in the form of tablets containing 0.01 , 0.05, 0.1 , 0.2, 1.0, 2.0, 5.0, 10.0, 50.0 and 100.0 milligrams of the active ingredient for the adjustment of the dosage to the patient to be treated.
  • the compound which inhibits the testosterone-induced growth of DDTl cells may be administered in a pharmaceutical composition comprising the active compound in combination with a pharmaceutically acceptable carrier adapted for topical administration.
  • Topical pharmaceutical compositions may be, e.g., in the form of a solution, cream, ointment, gel, lotion, shampoo or aerosol formulation adapted for application to the skin.
  • Topical pharmaceutical compositions useful in the method of treatment of the present invention may include about 0.001 % to 0.1% of the active compound in admixture with a pharmaceutically acceptable carrier.
  • compounds of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three or four times daily.
  • the compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in that art.
  • the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.
  • the dosage regimen utilizing the compounds of the present invention is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound thereof employed.
  • a physician of ordinary skill can readily determine and prescribe the effective amount of the drug required to prevent, counter, arrest or reverse the progress of the condition.
  • Optimal precision in achieving concentration of drug within the range that yields efficacy without toxicity requires a regimen based on the kinetics of the drug's availability to target sites. This involves a consideration of the distribution, equilibrium, and elimination of the drug.
  • the compound which inhibits the testosterone-induced growth of DDTl cells herein described in detail can form the active ingredient, and are typically administered in admixture with suitable pharmaceutical diluents, excipients or carriers (collectively referred to herein as "carrier” materials) suitably selected with respect to the intended form of administration, that is, oral tablets, capsules, elixirs, syrups and the like, and consistent with conventional pharmaceutical practices.
  • carrier suitable pharmaceutical diluents, excipients or carriers
  • suitable pharmaceutical diluents, excipients or carriers suitably selected with respect to the intended form of administration, that is, oral tablets, capsules, elixirs, syrups and the like, and consistent with conventional pharmaceutical practices.
  • the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like.
  • Capsules containing the product of this invention can be prepared by mixing an active compound of the present invention with lactose and magnesium stearate, calcium stearate, starch, talc, or other carriers, and placing the mixture in a gelatin capsule. Tablets may be prepared by mixing the active ingredient with conventional tableting ingredients such as calcium phosphate, lactose, com starch or magnesium stearate. Moreover, when desired or necessary, suitable binders, lubricants, disintegrating agents and coloring agents can also be incorporated into the mixture.
  • Suitable binders include starch, gelatin, natural sugars such as glucose or beta- lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like.
  • Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.
  • Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like.
  • the liquid forms may be administered in suitably flavored suspending or dispersing agents such as the synthetic and natural gums, for example, tragacanth, acacia, methyl-cellulose and the like.
  • suspending or dispersing agents such as the synthetic and natural gums, for example, tragacanth, acacia, methyl-cellulose and the like.
  • Other dispersing agents which may be employed include glycerin and the like.
  • sterile suspensions and solutions are desired.
  • Isotonic preparations which generally contain suitable preservatives are employed when intravenous administration is desired.
  • Topical preparations containing the active drug component can be admixed with a variety of carrier materials well known in the art, such as, e.g., alcohols, aloe vera gel, aliantoin, glycerine, vitamin A and E oils, mineral oil, PPG2 myristyl propionate, and the like, to form, e.g., alcoholic solutions, topical cleansers, cleansing creams, skin gels, skin lotions, and shampoos in cream or gel formulations. See, e.g., EP 0 285 382.
  • the compounds of the present invention can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles.
  • Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.
  • Compounds of the present invention may also be delivered by the use of monoclonal antibodies as individual carriers to which the compound molecules are coupled.
  • the compounds of the present invention may also be coupled with soluble polymers as targetable drug carriers.
  • Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidephenol, polyhydroxy- ethylaspartamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues.
  • the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels.
  • the present invention also provides for a pharmaceutical composition for the treatment of diseases of the prostate comprising a compound capable of inhibiting androgen induced DDTl cell growth and a pharmaceutically acceptable carrier.
  • the compound capable of inhibiting androgen-induced DDTl cell growth is N-(4-chlorophenyl)-(Z,Z)-2,3- bis(cyclopropylmethylene)cyclopentane carboxamide of structural formula (I):
  • the present invention also provides for the use of a compound which inhibits the testosterone-induced growth of DDTl cells in the preparation of a medicament useful in the treatment of diseases of the prostate.
  • DDTl cells (ATCC Nos. CRL-1701 and CRL-12051) were maintained in Dulbecco's modified Eagle's medium (GLBCO BRL) (DMEM) + 2% bovine calf serum (Hyclone®, BCS defined, iron supplemented) and 10 nM testosterone. Media was charged every 48 hours.
  • DMEM Dulbecco's modified Eagle's medium
  • Bovine calf serum Hyclone®, BCS defined, iron supplemented
  • DMEM fetal calf serum
  • F12 GLBCO BRL
  • ITSTM insulin, transferrin, selenious acid
  • DFITS media 0.1% bovine calf serum + 0.1% bovine calf serum (Hyclon®, BCS defined, iron supplemented).
  • 3-(4,5- dimethylthiazol-2-yl)-5-(3-carboxyl methoxyphenyl)-2-(4-sulfophenyl)- 2H-tetrazolium (MTS) was diluted into media at a final concentration of 333 ⁇ g/mL and penazine methosulfate (PMS) was used at a final concentration of 25 ⁇ M with a total volume per well of 100 ⁇ L. Plates were then incubated for one hour at 37°C in a humidified 5% C ⁇ 2 atmosphere. A linear response between cell number and absorbance at 490 nm was found up to 20,000 cells/well.
  • the proliferation of DDTl cells in synthetic serum-free medium is very slow doubling time >5 days in the absence of added testosterone, but is robust in the presence of 10 nM testosterone as shown in FIGURE 1.
  • the addition of either N-(4-chlorophenyl)-(Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentane carboxamide or an ethanol mock control showed little or no effect on cell proliferation.
  • the addition of N-(4-chlorophenyl)- (Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentane carboxamide at 10 ⁇ M along with 10 nM testosterone reduced cell proliferation to levels close to that of the ethanol mock control.
  • N-(4-chlorophenyl)-(Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentane carboxamide inhibits testosterone-induced DDTl cell growth in a manner similar to that of hydroxyflutamide.
  • cytosol 100 ⁇ L of cytosol (100,000 x g supernatant) were incubated overnight at 4°C in the presence of 1 nM [ ⁇ H]-DHT with and without unlabelled DHT at 1 ⁇ M to assess non-specific binding with and without added compound to measure competition. All samples were prepared in duplicate. Compound stock solutions were typically 100 fold concentrated in 100% ethanol and equal volumes of ethanol were employed as a mock control. Free steroids were removed by addition of dextran-coated charcoal followed by centrifugation at 3,000 x g for fifteen minutes at 4°C. Receptor-bound [ ⁇ H]-DHT was then measured by Scintillation counting of the supernatants.
  • Binding to the human androgen receptor was measured essentially as described by Tilley et al., "Characterization and expression of cDNA encoding the human androgen receptor", Proc. Nat'l Acad. Sci. USA 86:327-331 (1989), and Summerfield et al., "Tissue-specific pharmacology of testosterone and 5oc- dihydrotestosterone analogues: characterization of a novel canine androgen-binding protein", Mol. Pharm. 47: 1080-1088 (1995) except that stable transfectants of CHO cells were employed (gift from Prof. Michael McPhaul Soiled Medical Center, Dallas, Texas) that typically express 30-80 fmoles receptor/mg soluble protein.
  • N-(4-chlorophenyl)-(Z,Z)-2,3-bis(cyclopropylmethylene) cyclopentane carboxamide exhibits an IC50 (fifty percent displacement (IC50) of 3H-5 ⁇ -DHT (1.0 nM) of 27.5 ⁇ 0.13 ⁇ M (two independent experiments). Hydroxyflutamide has an IC50 ⁇ 100 nM using the same binding assay.
  • Ligand dependent transcription assays were performed as described in Schmidt et al. "Identification of a new member of the steroid hormone receptor superfamily that is activated by a peroxisome proliferator and fatty acids," Mol. Endocrinol. 6: 1634-1641 (1992).
  • a luciferase cDNA expressed under the control of the promoter region of the probasin gene that confers androgen receptor dependent transcription was used as a reporter gene, see Rennie et al. "Characterization of two cis-acting DNA elements involved in the androgen regulation of the probasin gene" Mol Endocrinol. 7:23-36 (1993).
  • the promoter region, -426 to +40 of the rat probasin gene was amplified by polymerase chain reaction (PCR) and inserted upstream of the luciferase cDNA of pGL3 Basic plasmid (Promega).
  • the expression vector pSGAR that expressed the normal human AR was used as a source of receptor, see, Chang et al. "Molecular cloning of human and rat complementary DNA encoding androgen receptors," Science 240:324-326 (1988).
  • Transient transfection of CVl cells was performed by plating cells (1.5x10 ⁇ per mL) into 12 well dishes in phenol red-free medium supplemented with 10% fetal calf serum treated with activated charcoal to remove endogenous steroids.
  • Cells were then transfected the following day by addition of a calcium phosphate precipitate of plasmid DNA. Cells were then washed with fresh media after an overnight incubation and ligands were added. Cell extracts were prepared after forty eight hours and assayed for luciferase enzyme activity using the Luciferase Assay System (Promega). Each transfection was performed in triplicate and the fluorescence of each sample was measured using the AutoChemiluminometer (Berthold).
  • N-(4-chlorophenyl-(Z,Z)-2,3- bis(cyclopropyl-methylene)cyclopentanecarboxamide acts as an agonist or antagonist to the androgen receptor
  • the androgen receptor and the probasin luciferase reporter gene were co-transfected into CVl cells and N-(4-chlorophenyl-(Z,Z)- 2,3-bis(cyclopropyl-methylene)cyclopentane carboxamide was added to the cells without or with 10 nM DHT.
  • Treatment using N-(4- chlorophenyl-(Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentane carboxamide alone did not affect luciferase expression from the pPBluc reporter gene.
  • N-(4-chlorophenyI-(Z,Z)-2,3- bis(cyclopropylmethylene)cyclopentane carboxamide inhibited the transactivation mediated by DHT in a dose dependent manner. Fifty percent inhibition of transactivation occurred at about 2 ⁇ M and maximal inhibition occurred at 100 ⁇ M. The results are graphed in FIGURE 2.
  • CVl cells were co-transfected with the probasin reporter gene (pPBluc) and with either the androgen receptor (pSGAR) or control plasmid (pSV2neo) and treated with vehicle or with DHT at the indicated concentration as described above.
  • pPBluc probasin reporter gene
  • pSGAR androgen receptor
  • pSV2neo control plasmid
  • an oral composition of a compound of this invention 5 mg N-(4-chlorophenyl-(Z,Z)-2,3- bis(cyclopropylmethylene)cyclopentane carboxamide, is formulated with sufficient finely divided lactose to provide a total amount of 580 to 590 mg to fill a size 0 hard gelatin capsule.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • General Health & Medical Sciences (AREA)
  • Cell Biology (AREA)
  • Medicinal Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Toxicology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Organic Chemistry (AREA)
  • Endocrinology (AREA)
  • Pain & Pain Management (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

A novel assay for identifying compounds with antiandrogenic activity employing a hamster ductus deferens cell line (DDT1) ATCC CRL-1701 and ATCC CRL-12051 is disclosed, as well as antiandrogenic compounds identified from this assay, particularly the nonsteroidal compound N-(4-chlorophenyl)-(Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentane carboxamide. This compound is an antiandrogen useful in the treatment and prevention of diseases of the prostate including prostatitis, benign prostatic hyperplasia (BPH) and prostatic carcinoma. The present invention also provides novel compositions employing compounds which exhibit antiandrogenic activity in the assay of the present invention, as well as those described in previously described publications, and their use in combination in methods of treatment for diseases of the prostate or in pharmaceutical compositions.

Description

TITLE OF THE INVENTION
ANTIANDROGENIC AGENTS AND METHODS OF IDENTIFICATION THEREOF
SUMMARY OF THE INVENTION
The present invention relates generally to a novel assay for identifying compounds with antiandrogenic activity as well as to antiandrogenic compounds identified from this assay. The assay employs a hamster ductus deferens cell line (DDT1) ATCC CRL-1701 and ATCC CRL-12051 that is highly dependent on the addition of testosterone in synthetic serum-free media.
The nonsteroidal compound N-(4-chlorophenyI)-(Z,Z)-2,3- bis(cyclopropylmethylene)cyclopentane carboxamide of structural formula (I):
Figure imgf000003_0001
<■> has been identified using the assay of the present invention as an antiandrogenic compound useful in the treatment and prevention of diseases of the prostate including prostatitis, benign prostatic hyperplasia (BPH) and prostatic carcinoma. The present invention also provides for novel compositions employing compounds which exhibit antiandrogenic activity in the assay of the present invention. The compounds which exhibit antiandrogenic activity in the assay of the present invention are useful in the systemic, including oral, and parenteral, including topical, treatment and prevention of diseases of the prostate including prostatic carcinoma, benign prostatic hyperplasia and prostatitis.
The compounds, including N-(4-chlorophenyl)-(Z,Z)-2,3- bis(cyclopropylmethylene)cyclopentane carboxamide, identified as androgen antagonists in the assay of the present invention may be used in combination with other active agents, for example, a 5α-reductase inhibitor such as finasteride, epristeride, 17β-N-(2,5- bis(trifluoromethyl))phenylcarbamoyl-4-aza-5ot-androst- 1 -en-3-one, or the compounds described in PCT publication WO 95/11254, or an αl - or αla-andrenergic receptor antagonist, or combinations of such other active agents with the androgen antagonist compound identified from the present assay, wherein such combinations would be useful in one or more of the above-mentioned methods of treatment or pharmaceutical compositions.
BACKGROUND OF THE INVENTION
The DDT1 cell line has been reported to be an androgen- responsive cell line. Norris et al. "Androgen Receptors in a Syrian Hamster Ductus Deferens Tumour Cell Line, " Nature 248:422-424 (1974). Syms et al., "Glucocorticoid Effects on Growth, and Androgen Receptor Concentrations on DDTl MF-2 Cell Lines", J. Steroid Biochem., 28(2): 109-1 16 (1987) report that the DDTl -MF-2 smooth muscle tumor cell line contains receptors for and is differentially sensitive to androgens and glucocorticoids. They report that androgens stimulate growth, and glucocorticoids inhibit growth of the cell line. Harris et al., "Androgens and Glucocorticoids Modulate Heparin-Binding Growth Factor I mRNA Accumulation in DDT1 Cells as Analyzed by in situ Hybridization", Mol. Endo. 3(1 1 ):1839-1844 (1989), describe the ductus deferens smooth muscle tumor cell line (DDTi -MF-2) as steroid-sensitive. They report that treatment with
10 nM testosterone accelerated the growth of DDT1 cells in the absence of serum, and that glucocorticoids inhibit growth. They further identify the DDTl -MF-2 cell line as a useful model for steroid responsive tumor growth in vitro. There has been a need for tissue culture models that mimic the androgen responsiveness observed in androgen sensitive diseases, particularly that observed in human prostatic carcinomas. The present invention provides for an assay to study compounds as potential anti- androgens in living cells.
Lippman et al. in "The Effects of Androgens and Antiandrogens on Hormone-response Human Breast Cancer in Long- Term Tissue Culture" Cancer Research 30: 4610-4618 (1976), describe the MCF-7 androgen-responsive cell line. Unlike the cell line of the present invention, the MCF-7 cell line cannot be grown in synthetic media.
Similarly, Marugo et al. in "Effects of Dihydrotestosterone and Hydroxyflutamide on Androgen Receptors in Cultured Human Breast Cancer Cells (EVSA-T)" J. Steroid Biochem. Mole. Biol. 42(5):547-554 (1992), describe another androgen responsive cell line. However, like the cells of the MCF-7 cell line, these cells cannot be grown in synthetic media.
The human androgen receptor has been isolated and characterized. Tilley et al., "Characterization and Expression of a cDNA
Encoding the Human Androgen Receptor", Proc. Nat'l. Acad. Sci. USA, 80:327-331 (1989) report the isolation, characterization and expression of the cDNA encoding the human androgen receptor, which predicts a protein of 917 amino acids and a molecular weight of 98918. Rasmusson et al., "Therapeutic Control of Androgen
Action", Ann. Rep. Med. Chem. 29:225-234 (1994) present a review of the androgen receptor, chemical antagonists of the androgen receptor, and means of controlling androgen biosynthesis.
Compounds identified as anti-androgens by the assay of the present invention are especially useful in the prevention and treatment of prostatic carcinoma, and they may also be useful in the treatment and prevention of other hyperandrogenic diseases such as acne vulgaris, seborrhea, female hirsutism, androgenetic alopecia, also called androgenic alopecia, which includes male and female pattern baldness, and benign prostatic hyperplasia.
Benign prostatic hyperplasia (BPH) and prostatic carcinoma are among the most common afflictions of aging men. Benign prostatic hyperplasia is often treated surgically with a procedure known as transurethral resection of the prostate (TURP). Other surgical procedures performed to release the obstruction of urine include incision or stents. Castration has also resulted in regression of prostatic enlargement. Drug therapy for BPH has included alpha- 1 blockers which treat the symptoms of the disease by alleviating obstructive symptoms, but do not affect the underlying cause of the disease, the enlarged prostate gland. Representative alpha- 1 blockers used in the treatment of BPH include: prazosin, terazosin, doxazosin, tamsulosin and alfuzosin. These drugs relax prostatic smooth muscle tone, decreasing intraurethral pressure without affecting bladder pressure. Common side effects of these agents are dizziness, headache and fatigue.
Finasteride (17β-(N-tert-butylcarbamoyl)-4-aza-5α- androst-l-ene-3-one), which is marketed by Merck & Co., Inc., under the tradename PROSCAR®, is an inhibitor of testosterone 5α-reductase currently marketed for the treatment of benign prostatic hyperplasia. A principal mediator of androgenic activity in the prostate is 5α- dihydrotestosterone ("DHT"), formed locally in the prostate by the action of testosterone-5α-reductase. Inhibitors of testosterone-5α- reductase inhibit the conversion of testosterone (T) to DHT and serve to prevent or lessen symptoms of hyperandrogenic stimulation in the prostate. See especially United States Patent No. 4,377,584 assigned to Merck & Co., Inc., issued March 22, 1983. The utility of finasteride in the treatment of prostatic carcinoma is also disclosed in the following documents: EP 0 285,382, published 5 October 1988; EP 0 285 383, published 5 October 1988; Canadian Patent no. 1 ,302,277; and Canadian Patent no. 1 ,302,276.
Both prostatic carcinoma and BPH have been treated with antiandrogens. Nonsteroidal antiandrogens such as flutamide and Casodex compete with DHT for androgen receptor sites in the prostate cells. These non-steroidal antiandrogens do not substantially change sexual potency and libido as the gonadotrophin releasing hormone agonists and progestogens do; however, these nonsteroidal antiandrogens often exhibit the undesirable tendency to feminize the male host (gynaecomastia) or initiate feed-back effects which would cause hyperstimulation of the testes.
Gonadotrophin-releasing hormone (GnRH) agonists such as nafarelin, buserelin, goserelin and leuprorelin all reduce the release of leutinizing hormone (LH) by desensitizing the GnRH receptors in the anterior pituitary gland. GnRH agonists are able to reduce the production of testosterone, induce shrinkage of prostate volume and reduce the severity of urinary symptoms of BPH. Unfortunately, these drugs have adverse effects such as impotence and flushing, which discourage a majority of patients from continuing with the drugs. These androgen-suppressing agents are thus of inconsequential significance in BPH treatment, but are of major importance in the treatment of patients with advanced prostatic cancer. Progestogens, such as megestrol acetate, hydroxyprogesterone and medrogestone depress testosterone by inhibiting LH release and blocking androgen receptors, causing a reduction in prostatic volume. Adverse effects such as decreased libido and impotence have kept progestogens from common use in BPH treatment. Thus, there still remains a need for additional therapies for
BPH and prostatic carcinoma for individuals who cannot tolerate the side effects and/or do not experience adequate relief from presently available therapies.
There also remains a need for a compound for the treatment of diseases of the prostate that is a non-steroidal compound having different pharmacological properties from steroids. Hori et al. "WB2838 [3-Chloro-4-(2-amino-3- chlorophenyl)-pyrrole]: Non-steroidal androgen-receptor antagonist produced by A Pseduomonas" J. Antibiotics 46(9):1327-1333 (1993) describe the nonsteroidal androgen receptor antagonist labeled WB2838:
Figure imgf000008_0001
WB2838
N-(4-chlorophenyl)-(Z,Z)-2,3-bis(cyclopropylmethylene) cyclopentane carboxamide is described by T. Wong "Synthesis nad Some Related Studies of Alkyl 2,3-bis(alkylidene)cyclopentance carboxylates", Ph.D. thesis, University of British Columbia, December, 1993. No use for this compound is described in the thesis.
Steroid hormones are involved in numerous aspects of cell growth and differentiation. In an effort to influence these processes chemists have developed analogs usually based on the four ring steroid nucleus. Nonsteroidal compounds interacting with steroid receptors are more rare and exhibit different pharmacological properties from their steroidal counterparts. We describe the use of androgen receptor binding assays and androgen-dependent DDT1 cells to identify and characterize novel antiandrogens, of particular interest are nonsteroidal antiandrogens.
BRIEF DESCRIPTION OF THE DRAWINGS FIGURE 1 is a graph of the results of two independent runs of the experiment described in Example 1. This graph shows the androgen dependent growth of DDT1 cells. Cells were plated in a 96 well plate at 2,000 cells/well in DFITS + 0.1 % fetal calf serum. After 24 hours, the cells were washed with the same medium except without added serum and replaced with fresh serum free DFITS (day 0) with ethanol mock control (open squares), with 10 nM testosterone (open circles), with 10 μM N-(4-chlorophenyl)-(Z,Z)-2,3-bis(cyclopropyl- methylene)cyclopentane carboxamide alone (filled squares), or 10 μM N-(4-chlorophenyl)-(Z,Z)-2,3-bis(cyclopropyl- methylene)cyclopentane carboxamide in the presence of 10 nM testosterone (filled circles). Cell growth data were normalized to the greatest number of cells and error bars represent +/- one standard error of the mean (sem).
FIGURE 2 is a graph showing the effect of N-(4- chlorophenyl)-(Z,Z)-2,3-bis(cyclopropyl- methylene)cyclopentane carboxamide on the activation of the androgen receptor in CVl cells, as described in Example 4. The filled triangles represent N-(4- chlorophenyl)-(Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentane carboxamide alone, the filled squares represent N-(4-chlorophenyl)- (Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentane carboxamide plus 10 nM dihydrotestosterone (DHT).
DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to a method for determining the antiandrogenic action of a compound comprising:
(a) plating DDT1 cells (ATCC CRL-12051) in medium free from androgens; (b) adding a solution containing the compound to the plated
DDT1 cells; (c) measuring the growth of the DDT1 cells in the presence of the compound over a measured period of time.
In one embodiment of the present invention, the method for determining the antiandrogenic action of a compound comprises:
(a) plating DDT1 cells (ATCC CRL-12051) in medium free from androgens;
(b) adding a solution containing the compound to the plated DDT1 cells;
(c) measuring the growth of the DDT1 cells in the presence of the compound over a measured period of time and calculating a rate of growth of the DDT1 cells. In one aspect of the present invention, the DDT1 cells are grown in synthetic serum-free media. The DDT1 cell line, also referred to as the DDT1 MF-2 cell line, was an existing deposit with the American Type Culture Collection ATCC No. CCRL-1701 which has been re-deposited with the American Type Culture Collection (ATCC), 12301 Parklawn Drive, Rockville, Maryland, United States of America under the conditions of the Budapest Treaty as ATCC CRL-12051 on February 21, 1996. The DDT1 smooth muscle cell line was cloned from cells derived from a leiomyosarcoma of the ductus deferens of a Syrian hamster (Mesocricetus auratus). It has maintained androgen and glucocorticoid receptors and alpha 1 and beta 2 adrenergic receptors. The beta 2 adrenergic receptor is functionally coupled to adenylate cyclase. Glucocorticoids regulate the expression of the c-sis proto- oncogene (inhibit). Growth is stimulated by androgens and inhibited by glucocorticoids.
Preferably, the method of the present invention is carried out by comparing the rate of growth of DDT1 cells in the presence of the compound to the rate of growth of DDT1 cells in the absence of the compound. In one embodiment of the present invention, a known volume of the solution containing the compound is added to the plated DDT1 cells. In a further illustration of the present invention, the solution of the compound is of a known concentration. When a known volume of a known concentration of the compound is added and the resulting rate of growth of the DDT1 cells is compared to the rate of growth when a "blank" solution is added (a volume of the solvent used to dissolve the compound equal to the volume of the solution containing the compound in the comparative run), the IC50 of the compound may be computed. The compound may be dissolved in a suitable solvent such as DMSO, ethanol, water or methylethyl ketone (MEK)> Preferably, the compound is dissolved in ethanol to form an ethanol solution. In one aspect of the present invention, a known volume of a known concentration of the ethanol solution is added. In a further aspect of the present invention, the rate of growth of the DDT1 cells when a known volume of a known concentration of the compound in an ethanol solution cells is compared to the rate of growth of DDT1 cells when the same volume of ethanol is added.
In one embodiment of the present invention, the growth of the DDT1 cells is measured over the period of one hour. In one class of the present invention, the measurement of the growth of the DDT1 cells over the measured period of time is performed by counting the number of cells at the beginning and at the end of the period of time. This can be done by quantitating cells using a hemocytometer. In another class of the present invention, the measurement of the growth of the DDT1 cells over the measured period of time is performed by incubating the DDT1 cells with labeled thymidine and measuring the incorporation of the label as a measure of DNA synthesis. Preferably, the labeled thymidine is ^H-thymidine, and the incorporation of the tritium label is measured by incubating cells for a period of time, preferably overnight, and washing extensively to remove free ^H-thymidine. Cells are then removed, preferably using trypsin, resuspended in media and the radioactivity quantitated, preferably by scintillation counting, for example using a Tri-Carb 2500TR Liquid Scintillation analyzer manufactured by Packard. In still another class of the present invention , the measurement of the growth of DDT1 cells is measured by colorimetric analysis. In particular, the growth may be measured by diluting 3-(4,5-dimethyl-thiazol-2-yl)-5-(3-carboxylmethoxy phenyl)-2- (4-sulfophenyl)-2H-tetrazolium into the medium and adding phenazine methosulfate, and measuring the absorbance at 490 nm.
In one embodiment of the present invention, the cells are plated in 96 well plates. In a preferred class of this embodiment of the invention, the assay is automated; i.e., the solutions are dispensed using a robotic device.
In one embodiment of the method of the present invention, the tested compound is assayed to determine that the compound inhibits the binding of labeled dihydrotestosterone (DHT) to the androgen receptor (AR). The androgen receptor used in the assay can be obtained from tissue such as prostate or liver, preferably human tissue, or cloned from the known sequence, see Tilley et al., Proc. Nat'l. Acad. Sci. USA, 80:327-331 (1989). Alternatively, the source of the human androgen receptor can be a cell line transfected with the human androgen receptor. Techniques for obtaining these transfected cell lines are well- known in the art and are preferred because of the uniformity of the receptor produced. The androgen receptor is incubated with labeled DHT, particularly 3H-DHT, and the compound to be tested or unlabelled DHT is added to the incubated receptor. The unlabelled DHT may be run in a parallel experiment in order to quantitate the nonspecific binding. The affinity for the androgen receptor is then calculated. One method of calculating this affinity is to add an agent to remove free labeled DHT. This may be done by a charcoal technique by adding dextran-coated charcoal to the sample at 0.5 volume of the original assay volume, vortexing, and centrifuging at 3,000 x g for 15 minutes at 4°C. Bound labeled DHT (preferably 3H-DHT) is in the supernatant. In a filter binding assay, the bound androgen receptor- labeled DHT (preferably AR-^H-DHT) complexes can be precipitated using agents such as ammonium sulfate, and filtered through glass fiber membranes. After extensive washing, bound labeled DHT (preferably 3H-DHT) is measured. When ^H-DHT is employed, the remaining label may be measured by scintillation counting of the glass fiber membranes, for example using a Tri-Carb 2500TR Liquid Scintillation analyzer manufactured by Packard. The compound may be tested for its ability to inhibit binding of DHT to the androgen receptor either before or after determining the antiandrogenic action of the compound according to the method of the present invention.
The present invention presents a convenient assay for monitoring the effect of compounds in a tissue culture system and demonstrates that the result of such an assay is the discovery of a nonsteroidal compound that acts as an antiandrogen.
Further, the present invention relates to a method of treating or preventing diseases of the prostate comprising administering 0.001 to 200 mg per day of a compound capable of inhibiting andro gen- induced DDTl cell growth to a male human in need of such treatment. The instant invention involves a method of treating and/or preventing BPH and prostatic carcinoma which comprises administering to a patient in need of such treatment a therapeutically effective amount of a compound which inhibits the testosterone induced growth of DDTl cells. In one embodiment of this method, the compound which inhibits the testosterone-induced growth of DDTl cells is administered in a dosage amount between 0.001 to 200.0 mg/day. In one class of this embodiment, the compound which inhibits the testosterone-induced growth of DDTl cells is administered in a dosage amount of from 0.01 to 50.0 mg/day, and in a sub-class of this embodiment, the compound which inhibits the testosterone-induced growth of DDTl cells is administered in a dosage amount of about 0.1 to 5.0 mg/day. Compounds which inhibit the testosterone-induced growth of DDTl cells can be determined by employing the assay described in Example 1.
In a second embodiment of this invention, the methods of treating and preventing benign prostatic hyperplasia and prostatic carcinoma comprise administration to a patient in need of such treatment of a compound which inhibits the testosterone-induced growth of DDTl cells. Preferably, this compound is a non-steroidal compound.
Preferred compounds that may be employed in the present invention include N-(4-chIorophenyl)-(Z,Z)-2,3-bis(cyclopropyl- methylene)cyclopentane carboxamide of structural formula (I):
Figure imgf000013_0001
(I) and pharmaceutically acceptable salts, esters and prodrugs thereof.
The nonsteroidal compound N-(4-chlorophenyl)-(Z,Z)-2,3- bis(cycloρropylmethylene)cyclopentane carboxamide which antagonizes the action of testosterone on DDTl cells but exhibits little or no effect on cell growth by itself. This compound also blocks the binding of ^H- DHT to the human androgen receptor expressed in stable transfectants of CHO cells at doses comparable to those that antagonize testosterone in DDTl cells. This compound may thus represent a novel class of nonsteroidal antiandrogens which show promise in the treatment of prostate cancer.
The present invention has the objective of providing methods of treating and preventing diseases of the prostate including BPH and prostatic carcinoma by systemic, oral, parenteral or topical administration of a compound which inhibits the testosterone-induced growth of DDTl cells in a dosage amount between 0.001 to 200.0 mg/day, and more particularly, from about 0.01 to 50.0 mg/day, and most particularly 0.1 to 5.0 mg/day. The term "treating BPH" is intended to include alleviating the obstructive symptoms of BPH, and slowing and/or reversing the growth of the prostate. The term
"preventing BPH" is intended to include preventing development of obstructive symptoms, and preventing the enlargement of the prostate. The term "treating prostatic carcinoma" is intended to include slowing and/or stopping the growth of prostatic carcinoma. The term "preventing prostatic carcinoma" is intended to include preventing the development of prostatic carcinoma in patients likely to develop prostatic carcinoma. Also, a compound which inhibits the testosterone- induced growth of DDTl cells may be co-administered with a 5α- reductase 2 inhibitor, such as finasteride or epristeride; a 5α-reductase 1 inhibitor such as 4,7β-dimethyl-4-aza-5α-cholestan-3-one, 3-oxo-4- aza-4,7β-dimethy 1- 16β-(4-chlorophenoxy)-5α-androstane, and 3-oxo-4- aza-4,7β-dimethyl- 16β-(phenoxy)-5α-androstane as disclosed in WO 93/23420 and WO 95/11254; dual inhibitors of 5α-reductase 1 and 5α- reductase 2 such as 3-oxo-4-aza-17β-(2,5-trifluoromethylphenyl- carbamoyl)-5α-androstane as disclosed in WO 95/07927; nonsteroidal antiandrogens such as flutamide and Casodex, and alpha- 1 blockers such as prazosin, terazosin, doxazosin, tamsulosin, and alfuzosin. The term "co-administered" includes concurrent administration as well as separate provision of the identified medicaments.
The present invention also has the objective of providing suitable systemic, oral, parenteral and topical pharmaceutical formulations for use in the novel methods of treatment of the present invention. The compositions containing as an active ingredient a compound which inhibits the testosterone-induced growth of DDTl cells can be administered in a wide variety of therapeutic dosage forms in conventional vehicles for systemic administration. For example, the compounds can be administered in such oral dosage forms as tablets, capsules (each including timed release and sustained release formulations), pills, powders, granules, elixirs, tinctures, solutions, suspensions, syrups and emulsions. Likewise, they may also be administered in intravenous (both bolus and infusion), intraperitoneal, subcutaneous, topical with or without occlusion, or intramuscular form, all using forms well known to those of ordinary skill in the pharmaceutical arts. For oral administration, for example, the compositions can be provided in the form of tablets containing 0.01 , 0.05, 0.1 , 0.2, 1.0, 2.0, 5.0, 10.0, 50.0 and 100.0 milligrams of the active ingredient for the adjustment of the dosage to the patient to be treated. The compound which inhibits the testosterone-induced growth of DDTl cells may be administered in a pharmaceutical composition comprising the active compound in combination with a pharmaceutically acceptable carrier adapted for topical administration. Topical pharmaceutical compositions may be, e.g., in the form of a solution, cream, ointment, gel, lotion, shampoo or aerosol formulation adapted for application to the skin. Topical pharmaceutical compositions useful in the method of treatment of the present invention may include about 0.001 % to 0.1% of the active compound in admixture with a pharmaceutically acceptable carrier. Advantageously, compounds of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three or four times daily. The compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal routes, using those forms of transdermal skin patches well known to those of ordinary skill in that art. To be administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen. The dosage regimen utilizing the compounds of the present invention is selected in accordance with a variety of factors including type, species, age, weight, sex and medical condition of the patient; the severity of the condition to be treated; the route of administration; the renal and hepatic function of the patient; and the particular compound thereof employed. A physician of ordinary skill can readily determine and prescribe the effective amount of the drug required to prevent, counter, arrest or reverse the progress of the condition. Optimal precision in achieving concentration of drug within the range that yields efficacy without toxicity requires a regimen based on the kinetics of the drug's availability to target sites. This involves a consideration of the distribution, equilibrium, and elimination of the drug.
In the methods of the present invention, the compound which inhibits the testosterone-induced growth of DDTl cells herein described in detail can form the active ingredient, and are typically administered in admixture with suitable pharmaceutical diluents, excipients or carriers (collectively referred to herein as "carrier" materials) suitably selected with respect to the intended form of administration, that is, oral tablets, capsules, elixirs, syrups and the like, and consistent with conventional pharmaceutical practices. For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like. Capsules containing the product of this invention can be prepared by mixing an active compound of the present invention with lactose and magnesium stearate, calcium stearate, starch, talc, or other carriers, and placing the mixture in a gelatin capsule. Tablets may be prepared by mixing the active ingredient with conventional tableting ingredients such as calcium phosphate, lactose, com starch or magnesium stearate. Moreover, when desired or necessary, suitable binders, lubricants, disintegrating agents and coloring agents can also be incorporated into the mixture. Suitable binders include starch, gelatin, natural sugars such as glucose or beta- lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes and the like. Lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like.
The liquid forms may be administered in suitably flavored suspending or dispersing agents such as the synthetic and natural gums, for example, tragacanth, acacia, methyl-cellulose and the like. Other dispersing agents which may be employed include glycerin and the like. For parenteral administration, sterile suspensions and solutions are desired. Isotonic preparations which generally contain suitable preservatives are employed when intravenous administration is desired. Topical preparations containing the active drug component can be admixed with a variety of carrier materials well known in the art, such as, e.g., alcohols, aloe vera gel, aliantoin, glycerine, vitamin A and E oils, mineral oil, PPG2 myristyl propionate, and the like, to form, e.g., alcoholic solutions, topical cleansers, cleansing creams, skin gels, skin lotions, and shampoos in cream or gel formulations. See, e.g., EP 0 285 382. The compounds of the present invention can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines. Compounds of the present invention may also be delivered by the use of monoclonal antibodies as individual carriers to which the compound molecules are coupled. The compounds of the present invention may also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidephenol, polyhydroxy- ethylaspartamidephenol, or polyethyleneoxidepolylysine substituted with palmitoyl residues. Furthermore, the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polylactic acid, polyepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels.
Thus, the present invention also provides for a pharmaceutical composition for the treatment of diseases of the prostate comprising a compound capable of inhibiting androgen induced DDTl cell growth and a pharmaceutically acceptable carrier. In one class of this embodiment of the invention, the compound capable of inhibiting androgen-induced DDTl cell growth is N-(4-chlorophenyl)-(Z,Z)-2,3- bis(cyclopropylmethylene)cyclopentane carboxamide of structural formula (I):
Figure imgf000018_0001
(I)
The present invention also provides for the use of a compound which inhibits the testosterone-induced growth of DDTl cells in the preparation of a medicament useful in the treatment of diseases of the prostate.
The following examples are not intended to be limitations on the scope of the instant invention in any way, and they should not be so construed. Furthermore, the compounds described in the following examples are not to be construed as forming the only genus that is considered as the invention, and any combination of the compounds or their moieties may itself form a genus. Those skilled in the art will readily understand that known variations of the conditions and processes of the following preparative procedures can be used to prepare these compounds.
All temperatures given in the following examples are in degrees Celsius.
EXAMPLE 1 DDTl Assay
DDTl cells (ATCC Nos. CRL-1701 and CRL-12051) were maintained in Dulbecco's modified Eagle's medium (GLBCO BRL) (DMEM) + 2% bovine calf serum (Hyclone®, BCS defined, iron supplemented) and 10 nM testosterone. Media was charged every 48 hours. Cells were plated at -2,000/well in 96-well plates or -6,000 cells/cm2 in DMEM: F12 (GLBCO BRL) nutrient mixture (1 :) + ITS™ (insulin, transferrin, selenious acid) (Collaborative Biomedical Products/Becton Dickinson Labware) (final concentrations 5μg/mL insulin, 5μ/mL transferrin, 5ng/mL selenious acid) (DFITS media) + 0.1% bovine calf serum (Hyclon®, BCS defined, iron supplemented).
After 24 hours, the cells were washed with the same media except without added serum and replaced with fresh serum-free DFITS medium (day 0).
In a typical experiment, cells were plated in sets of 12 in the presence of either a mock ethanol control (0.1 - 0.2% total volume) or N-(4-chlorophenyl)-(Z,Z)-2,3-bis(cyclopropylmethylene) cyclopentane carboxamide dissolved in 100% ethanol as a 1000X concentrated stock solution. Cell growth was then measured using a CelITiter96™ Non-Radioactive Cell Proliferation Assay (Promega Corporation) according to the manufacturer's protocol using a Bio Kinetics EL 340 microplate reader (BioTek Instruments, Inc.). 3-(4,5- dimethylthiazol-2-yl)-5-(3-carboxyl methoxyphenyl)-2-(4-sulfophenyl)- 2H-tetrazolium (MTS) was diluted into media at a final concentration of 333 μg/mL and penazine methosulfate (PMS) was used at a final concentration of 25 μM with a total volume per well of 100 μL. Plates were then incubated for one hour at 37°C in a humidified 5% Cθ2 atmosphere. A linear response between cell number and absorbance at 490 nm was found up to 20,000 cells/well. The proliferation of DDTl cells in synthetic serum-free medium is very slow doubling time >5 days in the absence of added testosterone, but is robust in the presence of 10 nM testosterone as shown in FIGURE 1. The addition of either N-(4-chlorophenyl)-(Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentane carboxamide or an ethanol mock control showed little or no effect on cell proliferation. In contrast, the addition of N-(4-chlorophenyl)- (Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentane carboxamide at 10 μM along with 10 nM testosterone reduced cell proliferation to levels close to that of the ethanol mock control. The classical nonsteroidal antiandrogen flutamide at 1 μM had the same effect, within experimental error, of blocking testosterone in this system. Thus N-(4-chlorophenyl)-(Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentane carboxamide inhibits testosterone-induced DDTl cell growth in a manner similar to that of hydroxyflutamide.
EXAMPLE 2 Binding to the Androgen Receptor
100 μL of cytosol (100,000 x g supernatant) were incubated overnight at 4°C in the presence of 1 nM [^H]-DHT with and without unlabelled DHT at 1 μM to assess non-specific binding with and without added compound to measure competition. All samples were prepared in duplicate. Compound stock solutions were typically 100 fold concentrated in 100% ethanol and equal volumes of ethanol were employed as a mock control. Free steroids were removed by addition of dextran-coated charcoal followed by centrifugation at 3,000 x g for fifteen minutes at 4°C. Receptor-bound [^H]-DHT was then measured by Scintillation counting of the supernatants.
Binding to the human androgen receptor (hAR) was measured essentially as described by Tilley et al., "Characterization and expression of cDNA encoding the human androgen receptor", Proc. Nat'l Acad. Sci. USA 86:327-331 (1989), and Summerfield et al., "Tissue-specific pharmacology of testosterone and 5oc- dihydrotestosterone analogues: characterization of a novel canine androgen-binding protein", Mol. Pharm. 47: 1080-1088 (1995) except that stable transfectants of CHO cells were employed (gift from Prof. Michael McPhaul Southwestern Medical Center, Dallas, Texas) that typically express 30-80 fmoles receptor/mg soluble protein.
N-(4-chlorophenyl)-(Z,Z)-2,3-bis(cyclopropylmethylene) cyclopentane carboxamide exhibits an IC50 (fifty percent displacement (IC50) of 3H-5α-DHT (1.0 nM) of 27.5 ± 0.13 μM (two independent experiments). Hydroxyflutamide has an IC50 ~ 100 nM using the same binding assay.
EXAMPLE 3
Preparation of N-(4-chlorophenyl-(Z,Z)-2,3-bis(cyclopropyl- methylene)cyclopentanecarboxamide
To a stirred solution of 4-chloroaniline (66 mg, 0.52 mmol) in dry benzene (2.6 mL, argon atmosphere) was added a solution of trimethylaluminum in toluene (0.26 mL, 2 M, 0.52 mmol) and the mixture was stirred at room temperature for 20 minutes. A solution of methyl (Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentanecarboxylate (80 mg, 0.344 mmol) in dry benzene (2.0 mL) was added and the mixture was refluxed for 4 hours. Hydrochloric acid (2 M, 4 mL) was added and the phases were separated. The aqueous phase was extracted three times with diethyl ether. The combined organic extracts were washed (brine), dried (magnesium sulfate) and concentrated. Flash chromatography (20 g silica gel, 4:1 petroleum ether-diethyl ether) of the crude product and removal of traces of solvent (vacuum pump) from the acquired material produced 93.5 mg (83%) of the amide, N- (4-chlorophenyl)-(Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentane carboxamide. Recrystallization of this material from 1 :1 dichloromethane-ethanol gave the amide N-(4-chlorophenyl)-(Z,Z)-2,3- bis(cyclopropylmethylene)cyclopentane carboxamide as colorless needle-like crystals (mp 126-127°C) that showed IR (KBr): 3255, 1664, 1594, 1494, 1399, 1096, 824 cm-1; U NMR (CDCI3, 400 MHz): 0.42- 0.60 (m, 4H), 0.80-0.95 (m, 4H), 1.77-1.90 (m, 2H), 2.00-2.10 (m, IH), 2.10-2.22 (m, IH), 2.35-2.53 (m, 2H), 3.40 (dd, IH, J=9, 4 Hz), 4.87 (d, IH, J=10 Hz), 4.96 (d, IH, J=10 Hz, 7.26 (d, 2H, J=9 Hz), 7.44 (d, 2H, J=9 Hz), 7.56 (br s, IH, wl/2 = 8 Hz); in a decoupling experiment, irradiation at 6 1.82 simplified the multiplets at 0.42-0.60 and 0.80- 0.95, and simplified the two doublets at 4.87 and 4.96 to two broad singlets (wl/2= 5, 3 Hz, respectively); in a series of NOE difference experiments, irradiation at 5 3.40 caused enhancement of the signals at 2.10-2.22 (9%) and 4.96 (10%); irradiation at δ 4.87 caused enhancement of the signals at 1.77-1.90 (4%) and 2.35-2.53 (3%); irradiation at δ 4.96 caused an enhancement of the signal at 3.40 (4%); ! 3c NMR (CDCI3, 50.3 MHz): δ 6.9, 7.1 , 8.5, 8.9, 13.8, 14.1 , 26.9, 31.9, 53.3, 120.6 , 128.85, 128.9, 130.2, 133.6, 135.6, 136.2, 136.8, 173.3. Exact mass calcd. for C2θH2235ClNO: 327.1392; found: 327.1384. Anal, calcd. for C20H22CINO: C 73.27, H 6.76, N, 4.27; found: C 73.17, H 6.67, N, 4.40.
EXAMPLE 4
Ligand dependent transcription assays were performed as described in Schmidt et al. "Identification of a new member of the steroid hormone receptor superfamily that is activated by a peroxisome proliferator and fatty acids," Mol. Endocrinol. 6: 1634-1641 (1992). A luciferase cDNA expressed under the control of the promoter region of the probasin gene that confers androgen receptor dependent transcription was used as a reporter gene, see Rennie et al. "Characterization of two cis-acting DNA elements involved in the androgen regulation of the probasin gene" Mol Endocrinol. 7:23-36 (1993). The promoter region, -426 to +40 of the rat probasin gene was amplified by polymerase chain reaction (PCR) and inserted upstream of the luciferase cDNA of pGL3 Basic plasmid (Promega). The expression vector pSGAR that expressed the normal human AR was used as a source of receptor, see, Chang et al. "Molecular cloning of human and rat complementary DNA encoding androgen receptors," Science 240:324-326 (1988). Transient transfection of CVl cells was performed by plating cells (1.5x10^ per mL) into 12 well dishes in phenol red-free medium supplemented with 10% fetal calf serum treated with activated charcoal to remove endogenous steroids. Cells were then transfected the following day by addition of a calcium phosphate precipitate of plasmid DNA. Cells were then washed with fresh media after an overnight incubation and ligands were added. Cell extracts were prepared after forty eight hours and assayed for luciferase enzyme activity using the Luciferase Assay System (Promega). Each transfection was performed in triplicate and the fluorescence of each sample was measured using the AutoChemiluminometer (Berthold). Thus to test whether N-(4-chlorophenyl-(Z,Z)-2,3- bis(cyclopropyl-methylene)cyclopentanecarboxamide acts as an agonist or antagonist to the androgen receptor, the effect of N-(4-chlorophenyl- (Z,Z)-2,3-bis(cyclopropyl-methylene)cyclopentanecarboxamide on either the AR dependent transcription from the pPBluc reporter gene or the ability of N-(4-chlorophenyl-(Z,Z)-2,3-bis(cyclopropylmethylene) cyclopentanecarboxamide to block the androgen receptor transactivation by DHT. The androgen receptor and the probasin luciferase reporter gene were co-transfected into CVl cells and N-(4-chlorophenyl-(Z,Z)- 2,3-bis(cyclopropyl-methylene)cyclopentane carboxamide was added to the cells without or with 10 nM DHT. Treatment using N-(4- chlorophenyl-(Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentane carboxamide alone did not affect luciferase expression from the pPBluc reporter gene. In contrast, N-(4-chlorophenyI-(Z,Z)-2,3- bis(cyclopropylmethylene)cyclopentane carboxamide inhibited the transactivation mediated by DHT in a dose dependent manner. Fifty percent inhibition of transactivation occurred at about 2 μM and maximal inhibition occurred at 100 μM. The results are graphed in FIGURE 2.
Table 1 : Androgen Receptor (AR) and DHT dependent transcription from the promoter of the probasin gene
Light Units + std Fold Stimulation + std
DHT PBluc pPBluc+AR pPBluc pPBluc+ AR control 4200+ 600 46000+ 4000 1.00+0.13 1.00+0.08
10-9 M 3000+ 300 290000+40000 0.71+0.07 6.3+0.9
10-7 M 3300+ 1100 1 100000+70000 0.78+0.25 24+1
CVl cells were co-transfected with the probasin reporter gene (pPBluc) and with either the androgen receptor (pSGAR) or control plasmid (pSV2neo) and treated with vehicle or with DHT at the indicated concentration as described above.
EXAMPLE 5 Oral Composition
As a specific embodiment of an oral composition of a compound of this invention, 5 mg N-(4-chlorophenyl-(Z,Z)-2,3- bis(cyclopropylmethylene)cyclopentane carboxamide, is formulated with sufficient finely divided lactose to provide a total amount of 580 to 590 mg to fill a size 0 hard gelatin capsule.
While the invention has been described and illustrated with reference to certain particular embodiments thereof, those skilled in the art will appreciate that various changes, modifications and substitutions can be made therein without departing from the spirit and scope of the invention. For example, effective dosages other than the particular dosages as set forth herein above may be applicable as a consequence of variations in the responsiveness of the mammal being treated for any of the indications for the compounds of the invention indicated above. Likewise, the specific pharmacological responses observed may vary according to and depending upon the particular active compound selected or whether there are present pharmaceutical carriers, as well as the type of formulation and mode of administration employed, and such expected variations or differences in the results are contemplated in accordance with the objects and practices of the present invention. It is intended, therefore, that the invention be defined by the scope of the claims which follow and that such claims be interpreted as broadly as is reasonable.

Claims

WHAT IS CLAIMED IS:
1. A method for determining the antiandrogenic action of a compound comprising: (a) plating DDTl cells (ATCC CRL-12051) in medium free from androgens;
(b) adding a solution containing the compound to the plated DDTl cells;
(c) measuring the growth of the DDTl cells in the presence of the compound over a measured period of time.
2. The method for determining the antiandrogenic action of a compound of Claim 1 comprising:
(a) plating DDTl cells (ATCC CRL-12051) in medium free from androgens;
(b) adding a solution containing the compound to the plated DDTl cells;
(c) measuring the growth of the DDTl cells in the presence of the compound over a measured period of time and calculating a rate of growth of the DDTl cells.
3. The method of Claim 2 wherein the rate of growth of DDTl cells in the presence of the compound is compared to the rate of growth of DDTl cells in the absence of the compound.
4. The method of Claim 2 wherein the medium is synthetic serum-free medium.
5. The method of Claim 4 wherein a known volume of the solution containing the compound is added to the plated DDTl cells.
6. The method of Claim 5 wherein the solution contains a known concentration of the compound.
7. The method of Claim 2 wherein the solution of the compound is an ethanol solution.
8. The method of Claim 2 wherein the growth of the DDTl cells is measured over the period of one hour.
9. The method of Claim 1 wherein the measurement of the growth of the DDTl cells over the measured period of time is performed by counting the number of cells at the beginning and at the end of the period of time.
10. The method of Claim 2 wherein the measurement of the growth of the DDTl cells over the measured period of time is performed by incubating the DDTl cells with labeled thymidine and measuring the incorporation of the label as a measure of DNA synthesis.
11. The method of Claim 10 wherein the labeled thymidine is ^H-thymidine.
12. The method of Claim 2 wherein the measurement of the growth of the DDTl cells over the measured period of time is performed by colorimetric analysis.
13. The method of Claim 12 wherein the growth of the DDTl cells is measured by diluting 3-(4,5-dimethyl-thiazol-2-yl)-5-(3- carboxylmethoxy phenyl)-2-(4-sulfophenyl)-2H-tetrazolium into the medium and adding phenazine methosulfate, and measuring the absorbance at 490 nm.
14. The method of Claim 1 wherein the cells are plated in 96 well plates.
15. The method of Claim 1 wherein the compound tested inhibits binding of labeled dihydrotestosterone to the androgen receptor.
16. A method of treating or preventing diseases of the prostate comprising administering 0.001 to 200 mg per day of a compound capable of inhibiting androgen-induced DDTl cell growth to a male human in need of such treatment.
17. The method of Claim 16 wherein the disease of the prostate is prostatic cancer.
18. The method of Claim 16 wherein the disease of the prostate is benign prostatic hyperplasia.
19. The method of Claim 16 wherein the compound is N- (4-chlorophenyl)-(Z,Z)-2,3-bis(cyclopropylmethylene)cyclopentane carboxamide of structural formula (I):
Figure imgf000028_0001
0
20. A pharmaceutical composition for the treatment of diseases of the prostate comprising a compound capable of inhibiting androgen induced DDTl cell growth in the method of Claim 1 and a pharmaceutically acceptable carrier.
21. The pharmaceutical composition of Claim 20 wherein the compound is N-(4-chlorophenyl)-(Z,Z)-2,3- bis(cyclopropylmethylene)cyclopentane carboxamide of structural formula (I):
Figure imgf000029_0001
(I)
PCT/US1997/002871 1996-03-01 1997-02-25 Antiandrogenic agents and methods of identification thereof WO1997031531A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US60943596A 1996-03-01 1996-03-01
US1263896P 1996-03-01 1996-03-01
US08/609,435 1996-03-01
US60/012,638 1996-03-01

Publications (1)

Publication Number Publication Date
WO1997031531A1 true WO1997031531A1 (en) 1997-09-04

Family

ID=26683825

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1997/002871 WO1997031531A1 (en) 1996-03-01 1997-02-25 Antiandrogenic agents and methods of identification thereof

Country Status (1)

Country Link
WO (1) WO1997031531A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000050454A1 (en) * 1999-02-24 2000-08-31 Trustees Of Tufts College Androgen-induced suppressor of cell proliferation and uses thereof
US6994992B1 (en) 1999-02-24 2006-02-07 Trustees Of Tufts College Androgen-induced suppressor of cell proliferation and uses thereof

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5130462A (en) * 1988-03-30 1992-07-14 E. R. Squibb & Sons, Inc. Cyclobutane derivatives

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5130462A (en) * 1988-03-30 1992-07-14 E. R. Squibb & Sons, Inc. Cyclobutane derivatives

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
ANTICANCER RES., 1989, Vol. 9, NERI et al., "Complete Androgen Blockade as Treatment for Advanced Prostate Cancer: Clinical Response and Side Effects", pages 13-16. *
J. STEROID BIOCHEM., 1984, Vol. 20, No. 1, SMITH et al., "Differential Effects of Androgens and Glucocorticoids on Regulation of Androgen Receptor Concentrations and Cell Growth", pages 277-281. *
J. STEROID. BIOCHEM., 1986, Vol. 24, No. 5, SMITH et al., "Steroid Regulation of Receptor Concentration and Oncogene Expression", pages 51-55. *
J. STEROID. BIOCHEM., 1988, Vol. 29, No. 1, TURCOTTE et al., "Androgen Binding as Evidenced by a Whole Cell Assay System Using Cultured Canine Prostatic Epithelial Cells", pages 69-76. *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000050454A1 (en) * 1999-02-24 2000-08-31 Trustees Of Tufts College Androgen-induced suppressor of cell proliferation and uses thereof
US6994992B1 (en) 1999-02-24 2006-02-07 Trustees Of Tufts College Androgen-induced suppressor of cell proliferation and uses thereof

Similar Documents

Publication Publication Date Title
US5872150A (en) Treatment of prostate disease with a nonsteroidal anti-androgenic compound
JP5934670B2 (en) Diarylthiohydantoin compounds
JP2862376B2 (en) 16-substituted-4-aza-androstane-5α-reductase isozyme 1 inhibitors
JP2005507860A (en) Hedgehog signaling pathway mediators, related compositions and uses
US5510485A (en) 17-ester, amide, and ketone derivatives of 3-oxo-4-azasteroids as 5A-reductase inhibitors
MXPA06008520A (en) Enhancement of anti-androgenic activity by a combination of inhibitors targeting different steps of a steroid-dependent gene activation pathway and uses thereof.
JP2014094952A (en) Local active &#34;soft&#34; anti-androgen agent
JP3820349B2 (en) Steroid 3-O-sulfamate derivatives as inhibitors of estrone sulfatase
JPH09504553A (en) 7-Substituted-4-aza-steroid derivatives as 5α-reductase inhibitors
EP0756481B1 (en) 17$g(b)-ARYL-4-AZA-STEROID DERIVATIVES
WO1997031531A1 (en) Antiandrogenic agents and methods of identification thereof
JP5144543B2 (en) Steroidal compounds as steroid sulfatase inhibitors
EP0711164B1 (en) 4-aza-pregnane 5-alpha-reductase isozyme 1 inhibitors
AU707324B2 (en) 4-azasteroids for treatment of hyperandrogenic conditions
EP0862556B1 (en) 17-alkyl-7-substituted-4-aza steroid derivatives
US6001844A (en) 4-Azasteroids for treatment of hyperandrogenic conditions
Li et al. Synthesis and in Vitro Evaluation of 4-Substituted N-(1, 1-Dimethylethyl)-3-oxo-4-androstene-17. beta.-carboxamides as 5. alpha.-Reductase Inhibitors and Antiandrogens
AU696320B2 (en) 4-aza-19-norandrostane derivatives
CA2169750A1 (en) Di- and tetra-hydrobenzo[f]quinolin-3-ones
HRP950100A2 (en) 16-substituted-4-aza-androstane 5-alpha-reductase isozyme 1 inhibitors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA JP US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 97531052

Format of ref document f/p: F

NENP Non-entry into the national phase

Ref country code: CA

122 Ep: pct application non-entry in european phase