WO1993021145A1 - Non-steroid progesterone receptor agonist and antagonist compounds and methods - Google Patents

Non-steroid progesterone receptor agonist and antagonist compounds and methods Download PDF

Info

Publication number
WO1993021145A1
WO1993021145A1 PCT/US1993/003909 US9303909W WO9321145A1 WO 1993021145 A1 WO1993021145 A1 WO 1993021145A1 US 9303909 W US9303909 W US 9303909W WO 9321145 A1 WO9321145 A1 WO 9321145A1
Authority
WO
WIPO (PCT)
Prior art keywords
cyclocymopol
compound
monomethyl ether
progesterone
methylidene
Prior art date
Application number
PCT/US1993/003909
Other languages
French (fr)
Inventor
I. Charles Pathirana
Tina S. Berger
Robert S. Stein
William Fenical
Todd K. Jones
Lawrence G. Hamann
Luc Farmer
Original Assignee
Ligand Pharmaceuticals Inc.
The Regents Of The University Of California
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 Ligand Pharmaceuticals Inc., The Regents Of The University Of California filed Critical Ligand Pharmaceuticals Inc.
Priority to EP93910811A priority Critical patent/EP0637296A4/en
Publication of WO1993021145A1 publication Critical patent/WO1993021145A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/72Receptors; Cell surface antigens; Cell surface determinants for hormones
    • C07K14/721Steroid/thyroid hormone superfamily, e.g. GR, EcR, androgen receptor, oestrogen receptor
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C205/00Compounds containing nitro groups bound to a carbon skeleton
    • C07C205/06Compounds containing nitro groups bound to a carbon skeleton having nitro groups bound to carbon atoms of six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C205/00Compounds containing nitro groups bound to a carbon skeleton
    • C07C205/45Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by at least one doubly—bound oxygen atom, not being part of a —CHO group
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C25/00Compounds containing at least one halogen atom bound to a six-membered aromatic ring
    • C07C25/24Halogenated aromatic hydrocarbons with unsaturated side chains
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C43/00Ethers; Compounds having groups, groups or groups
    • C07C43/02Ethers
    • C07C43/20Ethers having an ether-oxygen atom bound to a carbon atom of a six-membered aromatic ring
    • C07C43/23Ethers having an ether-oxygen atom bound to a carbon atom of a six-membered aromatic ring containing hydroxy or O-metal groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • C07C45/61Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups
    • C07C45/67Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by isomerisation; by change of size of the carbon skeleton
    • C07C45/673Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by isomerisation; by change of size of the carbon skeleton by change of size of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • C07C45/61Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups
    • C07C45/67Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by isomerisation; by change of size of the carbon skeleton
    • C07C45/68Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by isomerisation; by change of size of the carbon skeleton by increase in the number of carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • C07C45/61Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups
    • C07C45/67Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by isomerisation; by change of size of the carbon skeleton
    • C07C45/68Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by isomerisation; by change of size of the carbon skeleton by increase in the number of carbon atoms
    • C07C45/70Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by isomerisation; by change of size of the carbon skeleton by increase in the number of carbon atoms by reaction with functional groups containing oxygen only in singly bound form
    • C07C45/71Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by isomerisation; by change of size of the carbon skeleton by increase in the number of carbon atoms by reaction with functional groups containing oxygen only in singly bound form being hydroxy groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C47/00Compounds having —CHO groups
    • C07C47/52Compounds having —CHO groups bound to carbon atoms of six—membered aromatic rings
    • C07C47/575Compounds having —CHO groups bound to carbon atoms of six—membered aromatic rings containing ether groups, groups, groups, or groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/587Unsaturated compounds containing a keto groups being part of a ring
    • C07C49/603Unsaturated compounds containing a keto groups being part of a ring of a six-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/587Unsaturated compounds containing a keto groups being part of a ring
    • C07C49/753Unsaturated compounds containing a keto groups being part of a ring containing ether groups, groups, groups, or groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C69/00Esters of carboxylic acids; Esters of carbonic or haloformic acids
    • C07C69/017Esters of hydroxy compounds having the esterified hydroxy group bound to a carbon atom of a six-membered aromatic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/06Systems containing only non-condensed rings with a five-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/16Systems containing only non-condensed rings with a six-membered ring the ring being unsaturated

Definitions

  • This invention relates to intracellular receptors and ligands therefor. More specifically, this invention relates to compounds which are non-steroidal progesterone receptor antagonists or agonists, and methods for use of such compounds or ligands.
  • hormones certain small molecule, non-peptide hormones and similarly acting vitamins and vitamin metabolites (collectively hereinafter called “hormones”) modulate gene transcription by acting in concert with intracellular components, including intracellular receptors and discrete DNA promoter enhancer sequences known as hormone response elements (HREs).
  • hormone response elements HREs
  • ligands are synthesized in the body or may be taken in as a component of food. It has also been shown that compounds other than the natural ligands can act upon intracellular receptors to regulate hormone-responsive genes. For example, some natural product derivatives and synthetic compounds also function as ligands for these receptors.
  • Intracellular receptors form a class of structurally-related genetic regulators scientists have named "ligand dependent transcription factors.” Regulation of a gene by such factors requires both the intracellular receptor itself and a corresponding ligand which has the ability to selectively bind to the intracellular receptor in a way that affects gene activity. Until bound by a ligand, the intracellular receptor is unable to exert an effect on the gene. Hormone or other ligand molecules in the fluid surrounding a cell pass through the outer cell membrane by passive diffusion. Once inside the cell, the ligand binds to specific intracellular receptor proteins, creating a ligand/receptor complex. The binding of the ligand to its receptor induces a change in the shape of the intracellular receptor. This conformational change is believed to expose regions of the intracellular receptor that permit the intracellular receptor/1igand complex to bind to a specific subset of genes present in the cell' s DNA in the cell nucleus.
  • the blueprint to build specific proteins is encoded in the DNA sequence of each gene. This blueprint is copied in a process referred to as "transcription,” to give rise to the actual template for the production of specific proteins, messenger RNA or "mRNA". The mRNA then moves from the cell's nucleus into the cytoplasm and is translated, which results in the production of proteins encoded in the mRNA. Accordingly, a reduction in the transcription of mRNA reduces the production of the specific proteins.
  • the intracellular receptor/ligand complex binds to the specific site on the DNA, it alters the amount of the protein encoded by the gene that the cell is directed to produce, by altering the amount of mRNA transcribed by that gene.
  • a ligand which binds an intracellular receptor and mimics the effect of the natural ligand is referred to as an "agonist” ligand.
  • a ligand that inhibits the effect of the hormone is called an "antagonist.”
  • Intracellular receptors are referred to as "ligand-dependent transcription factors" because their activity is dependent upon the binding of their hormonal or other ligands, which are necessary to drive the intracellular receptor into its active conformation.
  • the intracellular receptors form a large family of proteins that are closely related in structure. They are important drug targets, and many drugs currently on the market are ligands for these receptors. Not surprisingly, the structural similarity of the receptors often results in cross-reactivity between a drug and receptors other than its target. It is apparent, therefore, that there is a need to find alternative ligands (agonists and antagonists) which are readily available for therapeutic administration, have added specificity for particular receptors, and have increased activity.
  • Ligands to the progesterone receptor are known to play an important role in gynecological medicine, cancer, and other health care problems of women. Its natural ligand, the female steroid progesterone, and synthetic analogues are, for example, used in birth control formulations. Antagonists to progesterone are useful in treating chronic disorders such as certain forms of hormone dependent cancer of the breast, ovaries, and endometrium (the lining of the uterus), and in treating uterine fibroids. Endometriosis, a leading cause of infertility in women, currently treated in early stage development by surgery, is also amenable to treatment with progesterone.
  • cymopol A group of prenylated bromohydroquinones, called collectively cymopols, has been isolated and identified by several investigators using as a starting material the green marine alga Cymopolia barbata (L.) Lamouroux (Dasycladaceae).
  • cymopol C 16 H 21 BrO 2
  • cymopol is a crystalline phenol which has a bromogeranyl-hydroquinone or brominated monoterpene-quinol structure.
  • Cyclocymopol [1-bromo-3-(4-bromo-2,5-dihydroxybenzyl)-2,2-dimethyl-4 methylene cyclohexane] and its monomethyl ether have also been obtained from C. barbata. See Högberg et al., supra. As described in McConnell et al., Phytochemistry, Vol. 21, No. 8, pp. 2139-41 (1982), C. barbata contains a mixture of optically active diastereomers of cyclocymopol, C 16 H 20 Br 2 O 2 , and cyclocymopol monomethyl ether, C 17 H 22 Br 2 O 2 , having the following structures:
  • Wall et al. J. Nat. Prod., Vol. 52, No. 5, pp. 1092-99 (1989), described additional diastereomeric cymopol compounds (cymobarbatol and 4-isocymobarbatol) which were determined to be highly active antimutagens.
  • Wall et al. reported obtaining pure cymobarbatol compounds, but were unable to obtain stable cyclocymopol fractions.
  • the forms of cyclocympol and cyclocymopol monomethyl ether obtained by Högberg et al., supra, were pure forms of formulae lb and 2b above.
  • the present invention is directed to compounds, compositions, and methods for modulating processes mediated by progesterone receptors. More particularly, the invention relates to non-steroidal compounds which are high affinity, high specificity ligands for progesterone receptors. These compounds exhibit progesterone receptor agonist or progesterone receptor antagonist activity, and modulate processes mediated by progesterone receptors. Accordingly, the invention also relates to methods for modulating processes mediated by progesterone receptors employing the compounds disclosed. Examples of compounds used in and forming part of the invention include cyclocymopol derivatives and purified diastereomers thereof, synthetic cyclocymopol analogs, and semisynthetic derivatives of natural cyclocymopols. Pharmaceutical compositions containing the compounds disclosed are also within the scope of this invention. Also included are methods for identifying or purifying progesterone receptors by use of the compounds of this invention.
  • Figure 1 presents the proton NMR spectrum for the individual pure 3R (panel a) and 3S (panel b) diastereomeric acetates of cyclocymopol monomethyl ether.
  • Figure 2 presents activation profiles for analysis of progesterone receptor activation by a cyclocymopol monomethyl ether diastereomeric mixture (compound SO-44), by a pure 3S diastereomeric acetate (compound SO-51), and by a pure 3R diastereomeric acetate (compound SO-52).
  • agonist dose response is shown in panel a
  • antagonist dose response in panel b.
  • Figure 3 presents activation profiles for analysis of progesterone receptor activation by (3R)-cyclocymopol monomethyl ether (compound SO-53).
  • 3R 3R-cyclocymopol monomethyl ether
  • agonist dose response is shown in panel a
  • antagonist dose response is shown in panel b.
  • Figure 4 presents activation profiles for analysis of progesterone receptor activation by (35)-cyclocymopol monomethyl ether (compound SO-54) and its acetate (compound SO-51).
  • agonist dose response is shown in panel a
  • antagonist dose response is shown in panel b.
  • Figure 5 presents activation profiles for analysis of progesterone receptor activation by (3R)-cyclocymopol monomethyl ether (compound SO-9).
  • 3R 3R-cyclocymopol monomethyl ether
  • FIG. 5 presents activation profiles for analysis of progesterone receptor activation by (3R)-cyclocymopol monomethyl ether (compound SO-9).
  • compound SO-9 3R-cyclocymopol monomethyl ether
  • Figure 6 presents activation profiles for analysis of glucocorticoid receptor activation by (3R)-cyclocymopol monomethyl ether (compound SO-09).
  • 3R glucocorticoid receptor activation by (3R)-cyclocymopol monomethyl ether
  • agonist dose response is shown in panel a
  • antagonist dose response is shown in panel b.
  • Figure 7 presents profiles of displacement of 3 H-labeled progesterone by cyclocymopol monomethyl ether diastereomers (panels a and b), and of the 3 H-labeled progesterone agonist R5020 by RU486 and by a (3R)-cyclocymopol monomethyl ether compound (SO-9).
  • Figure 8 presents profiles for analysis of progestrone binding for RU486 and (3R)-cyclocymopol monomethyl ether (compound SO-9).
  • Figure 9 presents profiles of the displacement of 3 H-labeled dexamethasone from glucocorticoid receptor for several compounds.
  • Figure 10 presents profiles showing the functional activities of cyclocymopol analogues in T47D cells.
  • Panel a shows ligand dependent induction of alkaline phosphatase in T47D cells by RTJ486 and cyclocymopol monomethyl ether diastereomers.
  • Inhibition by (3R)-cyclocymopol monomethyl ether (SO-53) of progesterone-stimulated induction of alkaline phosphatase is shown in panel b, and of R5020 stimulated induction in panel c.
  • Figure 11 presents profiles showing the inhibition by RU486 of induction of alkaline phosphatase in T47D cells by (35)-cyclocymopol monomethyl ether (SO-54) in panel a and by its acetate (SO-51) in panel b.
  • Cyclocymopols useful in this invention are defined as those having the formulae:
  • X is carbon, oxygen, or nitrogen
  • R 1 is R 17 , -OR 17 , -N(R 17 ) (R 17 ,), -SR 17 , fluorine, chlorine, bromine, or -NO 2 ;
  • R 17 and (R 17 ,), each independently, are hydrogen, saturated or unsaturated C 1 -C 6 alkyl, C 3 -C 7 cycloalkyl, C 5 -C 7 aryl, or C 7 aralkyl, said alkyl groups being branched or straight-chain;
  • R 3 is R 17 or -OR 17 ;
  • R 4 is hydrogen, -OR 17 , -OC (O) R 17 , -OC (O) OR 17 ,
  • R 5 is hydrogen or OR 17 ;
  • R 6 is R 17 ;
  • R 7 and R 8 are R 18 , or R 7 and R 8 together are a carbocyclic 3-8 member ring;
  • R 13 and R 14 are -OR 17 or R 18 , except when R 13 is attached to an sp 2 carbon atom in the ring, then R 14 is not present and R 13 is -OR 17 or R 18 ;
  • R 15 and R 16 are R 18 or OR 17 , or R 15 and R 16 together are -CH 2 -O- forming an epoxide, or R 15 and
  • alkyl refers to straight-chain, branched-chain, cyclic structures, and combinations thereof.
  • aryl refers to aromatic groups which have at least one ring having a conjugated pi electron system and includes carbocyclic aryl, heterocyclic aryl and biaryl groups, all of which may be optionally substituted, being preferably phenyl or phenyl substituted by one to three substituents, such substituents being advantageously lower alkyl, hydroxy, lower alkoxy, lower acyloxy, halogen, cyano, trihalomethyl, lower alcylamino, or lower alkoxycarbonyl.
  • Carbocyclic aryl groups are groups wherein the ring atoms on the aromatic ring are carbon atoms.
  • Carbocyclic aryl groups include monocyclic carbocyclic aryl groups and optionally substituted naphthyl groups.
  • Heterocyclic aryl groups are groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring with the remainder of the ring atoms being carbon atoms. Suitable heteroatoms include oxygen, sulfur, and nitrogen, and suitable heterocyclic aryl groups include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolyl, pyrimidyl, pyrazinyl, imidazolyl, and the like, all optionally substituted.
  • aralkyl refers to an alkyl group substituted with an aryl group. Suitable aralkyl groups include benzyl and the like, and may be optionally substituted.
  • the cyclocymopol compounds of this invention bind selectively to the progesterone receptor.
  • the non-synthetic cyclocymopol compounds have agonist or antagonist activity depending on their stereoisomeric form.
  • the 3 ⁇ or 3R diastereomer of cyclocymopol monomethyl ether has progesterone receptor antagonist activity
  • the 3 ⁇ or 3Sdiastereomer of cyclocymopol monomethyl ether has progesterone receptor agonist activity.
  • other cyclocymopol analogs or derivatives have been found to predominently exhibit progesterone receptor antagonist activity regardless of their stereoisomeric form.
  • the co-transfection assay provides a method for identifying functional ligands (either agonists which mimic, or antagonists which inhibit, the effect of hormones) for ligand-responsive receptor proteins.
  • the co-transfection assay provides a mechanism to evaluate ability of a compound to function as an agonist or antagonist of the activity modulated by an intracellular receptor.
  • the co-transfection assay mimics an in vivo system in the laboratory.
  • a cloned gene for an intracellular receptor is introduced by transfection (a procedure to induce cells to take up foreign genes) into a background cell substantially devoid of endogenous intracellular receptors. This introduced gene directs the recipient cells to make the intracellular receptor protein.
  • a second gene is also introduced (co-transfected) into the same cells in conjunction with the intracellular receptor gene. This second gene functions as a reporter for the transcription-modulating activity of the target intracellular receptor.
  • the reporter acts as a surrogate for the products normally expressed by a gene under control of the target receptor and its natural hormone.
  • a preferred reporter gene is one which expresses the firefly enzyme luciferase.
  • the co-transfection assay can detect small molecule agonists or antagonists of target intracellular receptors. Exposing the cells to an agonist ligand increases reporter activity in the transfeeted cells that can be conveniently measured, reflecting ligand-dependent, intracellular receptor-mediated increases in reporter transcription. To detect antagonists, the co-transfection assay is carried out in the presence of a constant concentration of an agonist known to induce a defined reporter signal. Increasing concentrations of a test antagonist will decrease the reporter signal. The co-transfection assay is therefore useful to detect both agonists and antagonists of specific intracellular receptors. It determines not only whether a compound interacts with a particular intracellular receptor, but also whether this interaction mimics (agonizes) or blocks (antagonizes) the effects of the natural regulatory molecules on target gene expression.
  • Co-transfected cells are exposed to a medium to which is added the potential ligand that is being evaluated. If the candidate ligand diffuses into the cell and binds to the receptor and the resulting complex functions as an agonist, it binds to the co-transfected reporter gene and initiates transcription.
  • that gene is one that expresses, for example, luciferase
  • luciferase is produced which catalyzes a light-emitting reaction with its substrate luciferin.
  • the amount of light produced relative to the concentration of candidate ligand used in the assay provides a measure of the potency and efficacy of the compound tested.
  • Antagonist activity is evaluated by adding the candidate ligand and a known agonist to the co-transfected cells. Suppression of agonist-induced luciferase production by the candidate compound, and hence the amount of light produced, indicates the candidate ligand is an antagonist.
  • CV-1's Cultured monkey kidney cells
  • the receptor cDNA was introduced in a mammalian expression vector under the control of the Rous Sarcoma virus LTR. These vectors provide for the efficient production of the progesterone receptor in these cells, which do not normally express this receptor gene.
  • a reporter vector was also transfected, containing a firefly luciferase gene under the control of the hormone-responsive promoter. Addition of control hormone
  • the cyclocymopol compounds were tested at eight concentrations (10 -4 to 10 -11 M) for the generation of a full dose response curve, and were compared to the progesterone control hormone response. A total of three replicates per concentration point were tested for each compound, and the EC 50 was calculated for each positive response. Both agonist and antagonist activity for each test compound was determined in parallel. In the antagonist assay, 10 -8 M progesterone was added to the media immediately prior to the addition of the cyclocymopol test compounds at the eight concentrations.
  • the assays showed a 3R (3 ⁇ ) and 3S (3 ⁇ ) diastereomer mixture of cyclocymopol monomethyl ether (designated compound SO-44) to have progesterone receptor antagonist activity, as shown in Figure 2.
  • agonist dose response is shown in panel a, and antagonist dose response in panel b.
  • the corresponding purified 3R diastereomeric acetate designated SO-52
  • a purified 3S diastereomeric acetate designated SO-51
  • Compound SO-9 and the identical compound from a separate preparation, compound SO-53 exhibit progesterone receptor antagonist activity (see Figure 5), and were tested for cross reactivity with the other known intracellular receptor classes, e.g., glucocorticoid, mineralocorticoid, androgen, estrogen, and retinoic acid.
  • Compound SO-9 was also tested with orphan receptors (which are receptors whose natural ligand is unknown). The compound was found not to cross-react with any of the other receptors, which demonstrates that activity was limited to the progesterone receptor.
  • Figure 6 is illustrative, and shows that compound SO-9 demonstrated neither agonist nor antagonist activity with the glucocorticoid receptor, dexamethasone.
  • the 3R and 3S diastereomeric acetates of cyclocymopol monomethyl ether were also individually tested for cross reactivity with the other known intracellular receptor classes. This testing showed the 3R diastereomer to have slight agonist activity with the glucocorticoid receptor. No antagonist activity was detected with either compound.
  • a plasmid which expresses progesterone receptor was transfected into CV-1 cells by the method of calcium phosphate precipitation. After six hours, the cells were washed and incubated at 37°C with 95% O 2 /5% CO 2 for 40 hours prior to harvest.
  • Progesterone receptor antagonists such as RU486, can increase the specific binding of progesterone to its ligand binding site. This enigma was also observed upon performing binding studies with 3 H-progesterone in the presence of compound SO-9. As shown in Figure 8, this compound significantly increased the apparent Bmax of 3 H-progesterone.
  • T47D cells have proven to be a particularly useful model to investigate the molecular actions of sex steroids because they contain endogenous functional receptors for, and respond to, progestins, and their respective antagonists. Moreover, these cells contain exceptionally high titers of progesterone receptors and are exceptionally sensitive to the actions of progestins in a manner quite similar to their actions in normal and neoplastic mammary epithelial cells.
  • progestins induce de novo synthesis of a plasma-associated alkaline phosphatase, which has been reported to be similar, if not identical, to the alkaline phosphatase present in the normal breast and human milk.
  • T47D cells were cultured in RPMI 1640 medium fortified with 10% fetal bovine serum, 2 mM glutamine, 0.2 ug/ml bovine insulin and, 0.05 mg/ml gentamicin. Cells were plated in 100-mm plates in medium; 48 hours later they were changed to medium containing 2% charcoal-treated serum with or without test compounds in a final ethanol concentration of 0.1%. For routine induction of alkaline phosphatase, cells were treated for three days with two media changes and harvested as described below.
  • cytosol Cells were collected with a rubber policeman into phosphate-buffered saline, pelleted, and lysed with TPSG buffer (0.2% Triton X-100 containing 10 mM sodium phosphate pH-7.4, 0.1 M sucrose, and 10% glycerol) at 0° C for 30 min with vigorous vortex mixing every 5 min. Nuclei were sedimented at 2500 rpm, and the supernatant was saved as cytosol. The protein content of the cytosol was assayed by the method of Bradford.
  • (3R)-cyclocymopol monomethyl ether could function as a progesterone receptor antagonist, the effects of increasing concentrations of this compound on R5020-induced alkaline phosphatase activity were quantified in T47D cells.
  • 3R)-cyclocymopol monomethyl ether (compound SO-53) is an effective antagonist of the progesterone mimic.
  • 3R)-cyclocymopol monomethyl ether compound SO-53 attenuated this induction in a concentration-dependent manner.
  • progesterone agonist (35)-cyclocymopol monomethyl ether (compound SO-54), and its acetate (SO-51)
  • progesterone antagonist RU486, as shown in Figure 11.
  • 3R- cyclocymopol monomethyl ether (compound SO-53) functioned as a progesterone receptor antagonist and attenuated the effects of progesterone in a concentration- dependant manner.
  • Synthetic and semisynthetic cyclocymopol analogs have been prepared which also have activity for the intracellular receptor for progesterone.
  • Representative analogs of the present invention are prepared according to the following illustrative synthetic schemes and illustrative examples.
  • reaction mixture was allowed to stir at 0°C for 60 min, at which time TLC analysis indicated complete consumption of starting material, and the formation of a less polar product (Rf 0.81, 2:1 hexanes/ethyl acetate).
  • Hexane 100 mL was then added, and the contents of the flask were transferred to a separatory funnel containing 50 mL of saturated aqueous NH 4 Cl, rinsing with an additional 50 mL hexane and 10 mL water.
  • the layers were separated, and the organic phase was washed with 20 mL 10% Na 2 S 2 O 3 , dried over Na 2 SO 4 , and concentrated under diminished pressure.
  • the reaction mixture was then cooled to 0°C before the cautious addition of 50 mL water, and the contents of the flask were then poured into a 500 mL Erlenmeyer flask containing 150 mL ice-cold 10% H 2 SO 4 .
  • the mixture was then extracted with ether (2 ⁇ 200 mL), and the combined organics were washed successively with water (100 mL), and saturated aqueous NaHCO 3 (100 mL), dried over Na 2 SO 4 , and concentrated under diminished pressure to give 6.05 g (quantitative) of the dimethylenone (Rf 0.55, 2:1 hexane/ethyl acetate).
  • This compound was prepared from 3-ethoxy-6, 5,5-trimethylcyclohex-2-en-1-one (9) (1.60 g, 8.80 mmol) in the manner previously described for enone 8, yielding 1.03 g (85%) of the methylated enone as a colorless oil.
  • 1 H NMR 400 MHz, CDCl 3 ) ⁇ 0.90 and 1.07 (2s, 2 ⁇ 3H, geminal- CH 3 's), 1.10 (d, 3H, CHCH 3 ), 5.96 (dd, 1H, 2-H), 6.16 ppm (dd, 1H, 3-H).
  • This compound was prepared from 6-[2'-(tert-butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]methyl-5,5-dimethylcyclohex-2-en-1-one (11) (0.075 g, 0.166 mmol) in the manner previously described for olefin (13), with the following procedural changes necessitated by the incompatibility of structural features particular to this substrate and the typical synthetic methodology.
  • Tetra-n-butylammonium fluoride (0.20 mL of a 1.0 M solution in THF, 0.20 mmol, 1.20 equiv) was added, and the mixture was allowed to warm to room temperature. The contents of the flask were then poured into a separatory funnel containing 30 mL ethyl acetate and 10 mL 1.0 M NaHSO 4 , the layers were separated, and the organic phase was washed with 10 mL brine, dried over Na 2 SO 4 , and concentrated under diminished pressure.
  • the crude material thus obtained was immediately carried on to the next step by transferring to a 10 mL nalgene vial containing 2-3 mL THF, and 0.3 mL premade HF/pyridine complex was added. After stirring overnight at room temperature, the reaction mixture was worked up in the usual manner, and purification by flash column chromatography (silica gel, hexane/ethyl acetate, gradient elution) afforded 38.3 mg (64%) of the desired acetoxydiene as a colorless, oily solid.
  • This compound was prepared from 1-methylidene-6-(2'-acetoxy-4'-bromo-5'-methoxyphenyl)methyl-3,5,5-trimethylcyclohex-2-ene (17) (6.8 mg, 0.017 mmol) in the manner previously described for phenolic diene 15, affording 5.8 mg (96%) of the phenolic diene as a colorless, oily solid.
  • This compound was prepared from 4,5,5-trimethylcyclohex-2-en-1-one (10) (169 mg, 1.22 mmol) in the manner previously described for benzylated enone 11, affording 0.337 g (59%) of the less polar trans diastereomer as a colorless, oily solid, along with 22 mg (4%) of the more polar cis diastereomer as a colorless, oily solid, separable by flash column chromatography. The relative stereochemistry of each respective diastereomer was confirmed by nOe NMR experiments.
  • This compound was prepared from trans-6-[2'-(tert-butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]methyl-4,5,5-trimethylcyclohex-2-en-1-one (19) (150 mg, 0.321 mmol) in the manner previously described for benzylated ketone 12, affording 0.149 g (99%) of the trans ketone as a colorless, oily solid.
  • This compound was prepared from trans-6-[2'-(tert-butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]methyl-4,5,5-trimethylcyclohex-2-en-1-one (19) (70.0 mg, 0.15 mmol) in the manner previously described for acetoxy-diene 14, affording 46.8 mg (79%) of the acetoxy-diene as a colorless oil.
  • This compound was prepared from 5,5-dimethyleyelohex-2-en-1-one (8) (0.600 g, 4.83 mmol) and p-nitrobenzyl bromide (1.581 g, 7.32 mmol) in the manner described for the synthesis of enone 11, affording 627 mg (50%) of the nitro-enone as a pale yellow oil.
  • This compound was prepared from 6-(4'-nitrophenyl)methyl-5,5-dimethylcyclohex-2-en-1-one (24) (133 mg, 0.514 mmol) in the manner previously described for the synthesis of olefin 13, with the following procedural changes. Three equivalents of (trimethyl) silylmethyllithium were used, and the subsequent elimination step required 48 h to go to completion, affording 120 mg (86%) of the nitrodiene as a colorless oil.
  • This compound was prepared in three steps from cyclohexanone and 2-(tert-butyl)dimethylsilyloxy-4-bromo-5-methoxybenzyl bromide (6) as previously described for the synthesis of olefin 13, to give the desired olefin in three steps in 13.5% overall yield as a colorless oil.
  • This compound was prepared from (35)-2'-(tert-butyl)dimethylsilyloxycyclocymopol monomethyl ether (25.0 mg, 0.047 mmol) in the manner described for the synthesis of the cyclocymopol derivative 27, affording 5.5 mg (35%) of the debromophenol as a colorless oil, along with the remainder of the mass balance as deprotected starting material.
  • the 400 MHz 1 H NMR spectrum and TLC elution properties of this compound were identical to those reported for the racemic analog 13. (This compound is also designated Compound "G" below.)
  • N-methyl-N-(2-pyridyl) formamide (28.5 mg, 25 ⁇ L, 0.21 mmol, 2.1 equiv) was added as a solution in 1 mL THF, and the reaction mixture was allowed to stir for 30 min before quenching with 1 mL 1:4 acetic acid/methanol. The reaction mixture was then partitioned between hexane and 1.0 M NaHSO 4 , washed with pH 7 buffer, and the resultant organic phase was dried over Na 2 SO 4 and concentrated under diminished pressure.
  • This compound was prepared from (3R)-2'-(tert-butyl)dimethylsilyloxycyclocymopol monomethyl ether (15.2 mg, 0.029 mmol) and iodine (300 ⁇ L of a 0.25 M solution in benzene, 0.075 mmol, 2.6 equiv) in the manner previously described for cyclocymopol derivative 29, affording 9.2 mg (70%) of the iodocyclocymopol derivative as a colorless oily solid.
  • Efficacy is reported as the % maximal response observed for each compound relative to RU-486, a compound known to exhibit progesterone receptor antagonist activity. Also reported in Tables 1 and 2 for each compound is its potency or IC 50 (which is the concentration (nM), required to reduce the maximal response by 50%), and its binding activity for the progesterone receptor.
  • the synthetic cyclocymopol compounds were also individually tested for cross-reactivity with the other known intracellular receptor classes. This testing showed the compounds not to have activity with the glucocorticoid receptor, in contrast to RU-486 which shows significant activity for that receptor. Some derivative compounds were found to exhibit slight activity for the androgen receptor.
  • the co-transfection assay provides a functional assessment of the ligand being tested as either an agonist or antagonist of the specific genetic process sought to be affected, and mimics an in vivo system in the laboratory.
  • Ligands which do not react with other intracellular receptors, as determined by the co-transfection assay, can be expected to result in fewer pharmacological side effects. Because the co-transfection assay is conducted in living cells, the evaluation of a ligand provides an early indicator of the potential toxicity of the candidate ligand at concentrations where a therapeutic benefit would be expected.
  • non-steroid progesterone receptor antagonist and agonist compounds disclosed can be readily utilized in pharmacological applications where progesterone receptor antagonist or agonist activity is desired, and where it is desired to minimize cross reactivities with other related intracellular receptors.
  • In vivo applications of the invention include administration of the disclosed compounds to mammalian subjects, and in particular to humans.
  • the compounds of the present invention are small molecules which are relatively fat soluble or lipophilic and enter the cell by passive diffusion across the plasma membrane. Consequently, these ligands are well suited for administration orally as well as by injection. Upon administration, these ligands can selectively activate progesterone receptors and thereby modulate processes mediated by these receptors.
  • compositions of this invention are prepared in conventional dosage unit forms by incorporating an active compound of the invention, or a mixture of such compounds, with a nontoxic pharmaceutical carrier according to accepted procedures in a nontoxic amount sufficient to produce the desired pharmacodynamic activity in a mammalian and in particular a human subject.
  • the composition contains the active ingredient in an active, but nontoxic, amount selected from about 5 mg to about 500 mg of active ingredient per dosage unit. This quantity depends on the specific biological activity desired and the condition of the patient.
  • the pharmaceutical carrier or vehicle employed may be, for example, a solid or liquid.
  • a variety of pharmaceutical forms can be employed.
  • the preparation when using a solid carrier, can be plain milled micronized in oil, tableted, placed in a hard gelatin or enteric-coated capsule in micronized powder or pellet form, or in the form of a troche, lozenge, or suppository.
  • the preparation when using a liquid carrier, can be in the form of a liquid, such as an ampule, or as an aqueous or nonaqueous liquid suspension.
  • the following examples provide illustrative pharmacological composition formulations: Example 23
  • Hard gelatin capsules are prepared using the following ingredients:
  • the above ingredients are mixed and filled into hard gelatin capsules in 250 mg quantities.
  • a tablet is prepared using the ingredients below:
  • Tablets each containing 60 mg of active ingredient, are made as follows:
  • the active ingredient, starch, and cellulose are passed through a No. 45 mesh U.S. sieve and mixed thoroughly.
  • the solution of PVP is mixed with the resultant powders, which are then passed through a No. 14 mesh U.S. sieve.
  • the granules so produced are dried at
  • SCMS magnesium stearate
  • talc previously passed through a No. 60 mesh U.S. sieve, and then added to the granules which, after mixing, are compressed on a tablet machine to yield tablets each weighing 150 mg.
  • Suppositories each containing 225 mg of active ingredient, may be made as follows:
  • the active ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimum heat necessary. The mixture is then poured into a suppository mold of normal 2g capacity and allowed to cool.
  • An intravenous formulation may be prepared as follows:
  • the compound is dissolved in the glycerol and then the solution is slowly diluted with isotonic saline.
  • the solution of the above ingredients is then administered intravenously at a rate of 1 ml per minute to a patient.
  • the compounds of this invention also have utility when labeled as ligands for use in assays to determine the presence of progesterone receptors. They are particularly useful due to their ability to selectively activate progesterone receptors, and can therefore be used to determine the presence of such receptors in the presence of other related receptors.
  • these compounds can be used to purify samples of progesterone receptors in vitro .
  • Such purification can be carried out by mixing samples containing progesterone receptors with one or more of the cyclocymopol and derivative compounds disclosed so that the compound (ligand) binds to the receptor, and then separating out the bound ligand/receptor combination by separation techniques which are known to those of skill in the art. These techniques include column separation, filtration, centrifugation, tagging and physical separation, and antibody complexing, among others.

Abstract

Non-steroidal compounds which are high affinity, high specificity ligands for progesterone receptors are disclosed. The compounds include synthetic derivatives of cyclocymopol and its diastereomers, spectroscopically and chromatographically pure (3R)-cyclocymopol monomethyl ether, which functions as a progesterone receptor antagonist, and spectroscopically and chromatographically pure (3S)-cyclocymopol monomethyl ether, which functions as a progesterone receptor agonist. Also disclosed are methods for employing the disclosed compounds for modulating processes mediated by progesterone receptors and for treating patients requiring progesterone receptor agonist or antagonist therapy.

Description

DESCRIPTION
Non-Steroid Progesterone Receptor Agonist And
Antagonist Compounds And Methods
Related Application
This application is a continuation-in-part of application Serial No. 07/872,710 filed April 21, 1992, whose entire disclosure is incorporated herein by reference.
Field of the Invention
This invention relates to intracellular receptors and ligands therefor. More specifically, this invention relates to compounds which are non-steroidal progesterone receptor antagonists or agonists, and methods for use of such compounds or ligands.
Background of the Invention
A central problem in eukaryotic molecular biology continues to be the elucidation of molecules and mechanisms that mediate specific gene regulation in response to molecular inducers such as hormones. As part of the scientific attack on this problem, a great deal of work has been done in efforts to identify molecular inducers which are capable of mediating specific gene regulation.
Although much remains to be learned about the specifics of gene regulation, it is known that certain small molecule, non-peptide hormones and similarly acting vitamins and vitamin metabolites (collectively hereinafter called "hormones") modulate gene transcription by acting in concert with intracellular components, including intracellular receptors and discrete DNA promoter enhancer sequences known as hormone response elements (HREs).
These hormones, acting through, and as "ligands" for, their intracellular receptors, directly regulate hormone- responsive genes (and perhaps other important genes which are not directly hormone-responsive). Natural ligands for intracellular receptors are synthesized in the body or may be taken in as a component of food. It has also been shown that compounds other than the natural ligands can act upon intracellular receptors to regulate hormone-responsive genes. For example, some natural product derivatives and synthetic compounds also function as ligands for these receptors.
Intracellular receptors form a class of structurally-related genetic regulators scientists have named "ligand dependent transcription factors." Regulation of a gene by such factors requires both the intracellular receptor itself and a corresponding ligand which has the ability to selectively bind to the intracellular receptor in a way that affects gene activity. Until bound by a ligand, the intracellular receptor is unable to exert an effect on the gene. Hormone or other ligand molecules in the fluid surrounding a cell pass through the outer cell membrane by passive diffusion. Once inside the cell, the ligand binds to specific intracellular receptor proteins, creating a ligand/receptor complex. The binding of the ligand to its receptor induces a change in the shape of the intracellular receptor. This conformational change is believed to expose regions of the intracellular receptor that permit the intracellular receptor/1igand complex to bind to a specific subset of genes present in the cell' s DNA in the cell nucleus.
The blueprint to build specific proteins is encoded in the DNA sequence of each gene. This blueprint is copied in a process referred to as "transcription," to give rise to the actual template for the production of specific proteins, messenger RNA or "mRNA". The mRNA then moves from the cell's nucleus into the cytoplasm and is translated, which results in the production of proteins encoded in the mRNA. Accordingly, a reduction in the transcription of mRNA reduces the production of the specific proteins.
Once the intracellular receptor/ligand complex binds to the specific site on the DNA, it alters the amount of the protein encoded by the gene that the cell is directed to produce, by altering the amount of mRNA transcribed by that gene. A ligand which binds an intracellular receptor and mimics the effect of the natural ligand is referred to as an "agonist" ligand. A ligand that inhibits the effect of the hormone is called an "antagonist." Intracellular receptors are referred to as "ligand-dependent transcription factors" because their activity is dependent upon the binding of their hormonal or other ligands, which are necessary to drive the intracellular receptor into its active conformation.
The intracellular receptors form a large family of proteins that are closely related in structure. They are important drug targets, and many drugs currently on the market are ligands for these receptors. Not surprisingly, the structural similarity of the receptors often results in cross-reactivity between a drug and receptors other than its target. It is apparent, therefore, that there is a need to find alternative ligands (agonists and antagonists) which are readily available for therapeutic administration, have added specificity for particular receptors, and have increased activity.
Ligands to the progesterone receptor are known to play an important role in gynecological medicine, cancer, and other health care problems of women. Its natural ligand, the female steroid progesterone, and synthetic analogues are, for example, used in birth control formulations. Antagonists to progesterone are useful in treating chronic disorders such as certain forms of hormone dependent cancer of the breast, ovaries, and endometrium (the lining of the uterus), and in treating uterine fibroids. Endometriosis, a leading cause of infertility in women, currently treated in early stage development by surgery, is also amenable to treatment with progesterone.
The identification of compounds which interact with progesterone receptors, and thereby affect transcription of genes which are responsive to progesterone, would be of significant value, e.g., for therapeutic applications such as treatment of hormonally-responsive gynecological and malignant disorders.
Further, the identification of compounds which have good specificity for the progesterone receptor, but which have less cross-reactivity for other intracellular receptors, would be of significant value since interaction of a ligand with other than the target intracellular receptors is known to result in significant undesirable pharmacological side effects. Accordingly, agonists and antagonists to the progesterone receptor which do not display cross-reactivity with other intracellular receptors will exhibit an improved therapeutic index.
A group of prenylated bromohydroquinones, called collectively cymopols, has been isolated and identified by several investigators using as a starting material the green marine alga Cymopolia barbata (L.) Lamouroux (Dasycladaceae). Among these, cymopol, C16H21BrO2, is a crystalline phenol which has a bromogeranyl-hydroquinone or brominated monoterpene-quinol structure. As described by Högberg et al., J.C.S. Perkin I, 1696-1701 (1976), cymopol [2-bromo-5-(3,7-dimethylocta-2,6-dienyl) hydroquinone] and its monomethyl ether, C17H23BrO2, have the following structures
Figure imgf000006_0001
Figure imgf000007_0001
Cymopol monomethyl ether
Cyclocymopol [1-bromo-3-(4-bromo-2,5-dihydroxybenzyl)-2,2-dimethyl-4 methylene cyclohexane] and its monomethyl ether have also been obtained from C. barbata. See Högberg et al., supra. As described in McConnell et al., Phytochemistry, Vol. 21, No. 8, pp. 2139-41 (1982), C. barbata contains a mixture of optically active diastereomers of cyclocymopol, C16H20Br2O2, and cyclocymopol monomethyl ether, C17H22Br2O2, having the following structures:
Figure imgf000007_0002
1a (R=H) : 2a (R=Me) : H (C-3)-pseudo-equatorial 1b (R=H) : 2b (R=Me) : H (C-3)-pseudo-axial (The above assumes the equatorial conformation for bromine at
C-1).
Through silica gel chromatography of an ether-soluble extract of (C. barbata. McConnell et al. were able to obtain a 1:1 mixture of α : β epimers of cyclocymopol. McConnell et al. also obtained a 3:1 mixture of α : β epimers of cyclocymopol monomethyl ether, which was enriched to a 4:1 mixture of the α: β epimers through purification techniques.
Wall et al., J. Nat. Prod., Vol. 52, No. 5, pp. 1092-99 (1989), described additional diastereomeric cymopol compounds (cymobarbatol and 4-isocymobarbatol) which were determined to be highly active antimutagens. Wall et al. reported obtaining pure cymobarbatol compounds, but were unable to obtain stable cyclocymopol fractions. Apparently, however, the forms of cyclocympol and cyclocymopol monomethyl ether obtained by Högberg et al., supra, were pure forms of formulae lb and 2b above.
The publications and references referred to above and hereafter in this specification are incorporated herein by reference.
Summary of the Invention
The present invention is directed to compounds, compositions, and methods for modulating processes mediated by progesterone receptors. More particularly, the invention relates to non-steroidal compounds which are high affinity, high specificity ligands for progesterone receptors. These compounds exhibit progesterone receptor agonist or progesterone receptor antagonist activity, and modulate processes mediated by progesterone receptors. Accordingly, the invention also relates to methods for modulating processes mediated by progesterone receptors employing the compounds disclosed. Examples of compounds used in and forming part of the invention include cyclocymopol derivatives and purified diastereomers thereof, synthetic cyclocymopol analogs, and semisynthetic derivatives of natural cyclocymopols. Pharmaceutical compositions containing the compounds disclosed are also within the scope of this invention. Also included are methods for identifying or purifying progesterone receptors by use of the compounds of this invention.
Brief Description of the Figures
The present invention may be better understood and its advantages appreciated by those skilled in the art by referring to the accompanying drawings wherein:
Figure 1 presents the proton NMR spectrum for the individual pure 3R (panel a) and 3S (panel b) diastereomeric acetates of cyclocymopol monomethyl ether.
Figure 2 presents activation profiles for analysis of progesterone receptor activation by a cyclocymopol monomethyl ether diastereomeric mixture (compound SO-44), by a pure 3S diastereomeric acetate (compound SO-51), and by a pure 3R diastereomeric acetate (compound SO-52). For these compounds and a progesterone control, agonist dose response is shown in panel a, and antagonist dose response in panel b.
Figure 3 presents activation profiles for analysis of progesterone receptor activation by (3R)-cyclocymopol monomethyl ether (compound SO-53). For this compound and a progesterone control, agonist dose response is shown in panel a, and antagonist dose response is shown in panel b.
Figure 4 presents activation profiles for analysis of progesterone receptor activation by (35)-cyclocymopol monomethyl ether (compound SO-54) and its acetate (compound SO-51). For these compounds and a progesterone control, agonist dose response is shown in panel a, and antagonist dose response is shown in panel b.
Figure 5 presents activation profiles for analysis of progesterone receptor activation by (3R)-cyclocymopol monomethyl ether (compound SO-9). For this compound and a progesterone control, against dose response is shown in panel a and antagonist dose response is shown in panel b.
Figure 6 presents activation profiles for analysis of glucocorticoid receptor activation by (3R)-cyclocymopol monomethyl ether (compound SO-09). For this compound and a dexamethasone control, agonist dose response is shown in panel a and antagonist dose response is shown in panel b.
Figure 7 presents profiles of displacement of 3H-labeled progesterone by cyclocymopol monomethyl ether diastereomers (panels a and b), and of the 3H-labeled progesterone agonist R5020 by RU486 and by a (3R)-cyclocymopol monomethyl ether compound (SO-9).
Figure 8 presents profiles for analysis of progestrone binding for RU486 and (3R)-cyclocymopol monomethyl ether (compound SO-9).
Figure 9 presents profiles of the displacement of 3H-labeled dexamethasone from glucocorticoid receptor for several compounds.
Figure 10 presents profiles showing the functional activities of cyclocymopol analogues in T47D cells. Panel a shows ligand dependent induction of alkaline phosphatase in T47D cells by RTJ486 and cyclocymopol monomethyl ether diastereomers. Inhibition by (3R)-cyclocymopol monomethyl ether (SO-53) of progesterone-stimulated induction of alkaline phosphatase is shown in panel b, and of R5020 stimulated induction in panel c.
Figure 11 presents profiles showing the inhibition by RU486 of induction of alkaline phosphatase in T47D cells by (35)-cyclocymopol monomethyl ether (SO-54) in panel a and by its acetate (SO-51) in panel b.
Detailed Description of the Preferred Embodiment
Cyclocymopols useful in this invention are defined as those having the formulae:
Figure imgf000011_0001
or
Figure imgf000011_0002
wherein:
the dotted lines in the structure depict optional double bonds;
X is carbon, oxygen, or nitrogen;
R1 is R17, -OR17, -N(R17) (R17,), -SR17, fluorine, chlorine, bromine, or -NO2;
R17 and (R17,), each independently, are hydrogen, saturated or unsaturated C1-C6 alkyl, C3-C7 cycloalkyl, C5-C7 aryl, or C7 aralkyl, said alkyl groups being branched or straight-chain;
R2 is -NO2, -N(OH)R17, fluorine, chlorine, bromine, iodine, R17, -N(R17) (R17.) , -SR17, -S(O)-R17, -S(O)2-R17, - CH2OH, -C(O)-H, -C(O)CH3, -C(O)-OCH3, -C=CH2, -C=CH-C(O)-OCH3, or R18; R18 and (R18,), each independently, are hydrogen, saturated or unsaturated C1-C6 alkyl, C3-C6 alkyl, C3-C7 cycloalkyl, C5-C7 aryl, or C7 aralkyl, said alkyl groups being branched or straight-chain which optionally may contain hydroxyl, aldehyde, ketone, nitrile, or ester groups;
R3 is R17 or -OR17;
R4 is hydrogen, -OR17 , -OC (O) R17, -OC (O) OR17,
-OC (O) N (R17) (R17, ) , -OS (O) 2R17, or -OS (O) -R17 ;
R5 is hydrogen or OR17;
R6 is R17 ;
R7 and R8, each independently, are R18, or R7 and R8 together are a carbocyclic 3-8 member ring;
R9 and R10, each independently, are chlorine, bromine, or R17, or R9 and R10 combined are =0, except when X=0, R9 and R10 are not present, and when X is N, then R10 is not present, or R9 and R10 together are joined in a carbocyclic
3-8 member ring;
R11 and R12, each independently, are -OR17, R18, are =0, or are =CH2, except when R11 is attached to an sp2 carbon atom in the ring, then R12 is not present and R11 is R18, or R11 and R13 together are joined in a carbocyclic 3-8 member ring or are -O- to form an epoxide;
R13 and R14, each independently, are -OR17 or R18, except when R13 is attached to an sp2 carbon atom in the ring, then R14 is not present and R13 is -OR17 or R18;
R15 and R16, each independently, are R18 or OR17, or R15 and R16 together are -CH2-O- forming an epoxide, or R15 and
R16 combined are =0 or =C(R18)(R18,), except when R15 is hydroxyl, then R16 is not hydroxyl, and when R15 is attached to an sp2 carbon atom in the ring, then Rιe is not present. Definitions
In accordance with the present invention and as used herein, the following terms are defined with the following meanings, unless explicitly stated otherwise.
The term alkyl refers to straight-chain, branched-chain, cyclic structures, and combinations thereof.
The term "aryl" refers to aromatic groups which have at least one ring having a conjugated pi electron system and includes carbocyclic aryl, heterocyclic aryl and biaryl groups, all of which may be optionally substituted, being preferably phenyl or phenyl substituted by one to three substituents, such substituents being advantageously lower alkyl, hydroxy, lower alkoxy, lower acyloxy, halogen, cyano, trihalomethyl, lower alcylamino, or lower alkoxycarbonyl.
Carbocyclic aryl groups are groups wherein the ring atoms on the aromatic ring are carbon atoms. Carbocyclic aryl groups include monocyclic carbocyclic aryl groups and optionally substituted naphthyl groups.
Heterocyclic aryl groups are groups having from 1 to 3 heteroatoms as ring atoms in the aromatic ring with the remainder of the ring atoms being carbon atoms. Suitable heteroatoms include oxygen, sulfur, and nitrogen, and suitable heterocyclic aryl groups include furanyl, thienyl, pyridyl, pyrrolyl, N-lower alkyl pyrrolyl, pyrimidyl, pyrazinyl, imidazolyl, and the like, all optionally substituted.
The term "aralkyl" refers to an alkyl group substituted with an aryl group. Suitable aralkyl groups include benzyl and the like, and may be optionally substituted.
The term "lower" referred to herein in connection with organic radicals or compounds respectively defines such with up to and including 7, preferably up to and including 4 and advantageously one or two, carbon atoms. Such groups may be straight chain or branched. Representative compounds and derivatives according to the present invention include the following:
1-Methylidene-2- (2' -acetoxy-4' -bromo-5' -methoxyphenyl)methyl-3,3-dimethylcyclohexane;
(3S)-1-Debromocyclocymopol monomethyl ether,2'-acetate;
1-Methylidene-2- (2' -hydroxy-4' -bromo-5' -methoxyphenyl)methyl-3,3-dimethylcyclohexane;
(3S)-1-Debromocyclocymopol monomethyl ether,2'-methylcarbonate;
(3R,5R)-5-Hydroxycyclocymopol monomethyl ether; 2- (4'-Nitrophenyl)methylcyclohexanone;
(3S)-1-Debromocyclocymopol monomethyl ether;
1-Methylidene-6- (2' -acetoxy-4' -bromo-5' -methoxyphenyl)methyl-3,5,5-trimethyleyelohex-2-ene;
1-Methylidene-6-(4'-nitrophenyl)methyl-3,5,5-trimethylcyclohex-2-ene;
(3R)-1-Debromocyclocymopol monomethyl ether;
1-Methylidene-6- (2' -hydroxy-4' -bromo-5' -methoxyphenyl)methyl-5,5-dimethylcyclohex-2-ene;
1-Methylidene-6- (2' -acetoxy-4' -bromo-5' -methoxyphenyl)methyl-5,5-dimethylcyclohex-2-ene;
1-Methylidene-6-(3'-methyl-4'-nitrophenyl)methyl-5,5-dimethylcyclohex-2-ene;
trans-1-Methylidene-6-(2'-acetoxy-4'-bromo-5'-methoxyphenyl)methyl-4,5,5-trimethylcyclohex-2-ene;
1-Methylidene-2-(4' -bromophenyl)methyl-3,3-dimethylcyclohexane;
1-Methylidene-2- (2' -hydroxy-4' -bromo-5' -methoxyphenyl)methyl-3,3-dimethylcyclopentane;
1-Methylidene-2-(4'-nitrophenyl)methylcyclohexane;
(3R)-1-Debromocyclocymopol monomethyl ether,2'-methylcarbonate;
1-Methylidene-2-(2'-hydroxy-4'-bromo-5'-methoxyphenyl)methylcyclohexane;
(3R)-Cyclocymopol monomethyl ether,2'-methylcarbonate; (3R)-1-Debromocyclocymopol monomethyl ether,2'-benzoate; and
(3R)-4'-Iodocyclocymopol monomethyl ether.
Compounds comprising the class of cyclocymopol compounds and derivatives disclosed herein can be obtained by routine chemical synthesis by those skilled in the art, e.g., by modification of the cyclocymopol compounds disclosed or by a total synthesis approach.
The cyclocymopol compounds of this invention bind selectively to the progesterone receptor. We have found that the non-synthetic cyclocymopol compounds have agonist or antagonist activity depending on their stereoisomeric form. For example, the 3α or 3R diastereomer of cyclocymopol monomethyl ether has progesterone receptor antagonist activity, and the 3β or 3Sdiastereomer of cyclocymopol monomethyl ether has progesterone receptor agonist activity. In contrast, other cyclocymopol analogs or derivatives have been found to predominently exhibit progesterone receptor antagonist activity regardless of their stereoisomeric form.
In the current invention, individual diastereomers of cyclocymopol monomethyl ether have been isolated from each other and purified in accordance with the following example. Example 1
The marine alga Cymopolia barbata (L.) Lamouroux
(Dasycladaceae) was collected and frozen. Frozen sample was lyophilized and extracted with 1:1 MeOH/CH2Cl2 three times, and the extract was concentrated in vacuo to obtain an aqueous suspension of organic components. The concentrate was re-extracted with CH2Cl2 until no color came into the organic phase, and the CH2Cl2 extract was then concentrated to obtain the crude extract as a dark, green oil . The crude extract was purified by column chromatography on Sephadex LH20 with 1:1 MeOH/CH2Cl2, or vacuum flash chromatography on silica using a gradient of ethyl acetate in hexane, and the fractions were examined by thin layer chromatography (TLC). The fractions that contained cymopols were pooled together and separated by reversed phase high performance liquid chromatography
(HPLC) using 80% MeOH/H2O to yield a mixture of cyclocymopol monomethyl ether diastereomers which were identified by nuclear magnetic resonance (NMR) spectroscopy as a mixture of the following as shown below:
Figure imgf000016_0001
Proton NMR spectrum of the cyclocymopol monomethyl ether mixture indicated the presence of the two diastereomers in the ratio 4:1. The two compounds could not be separated by reversed phase HPLC (ODS Column, 20%H2O/MeOH solvent) where both migrated with the same retention.
The two compounds behaved similarly on normal phase HPLC (silica column, 5% EtOAc/hexane solvent), and an attempt to collect fractions inside the HPLC peak also failed to yield any separation. This indicated that the diastereomers of the mixture had very similar chromatographic behavior, even though diastereomers normally have different chromatographic characteristics.
In order to separate the diastereomers, the cyclocymopol monomethyl ether diastereomer mixture was reacted with acetic anhydride (Ac2O) and pyridine at room temperature for about 10 hours. The resulting mixture of diastereomeric acetates was completely separated by normal phase HPLC (silica column, 5% EtOAc/hexane) to yield the individual chromatographically-pure diastereomers, as follows:
Figure imgf000017_0001
Proton NMR analysis confirmed that two separate spectroscopically-pure diastereomeric acetates were obtained, as shown in Figure 1. The spectrum for the 3R diastereometric acetate is shown in panel a, and the spectrum for the 3S diastereometric acetate is shown in panel b of Figure 1. The individual pure diastereomers were separately reacted with K2CO3 in MeOH to convert them back to the parent form, and the resultant products were purified by reversed phase HPLC using 80% MeOH/H2O to obtain the individual purified diastereomers of cyclocymopol monomethyl ether, as follows:
Figure imgf000017_0002
Proton NMR analysis of the resulting individual cyclocymopol monomethyl ether diastereomers also showed them to be spectroscopically pure. The 3R or 3α diastereomer was determined to have been the major component of the cyclocymopol monomethyl ether diastereomer mixture obtained from C. barbata. Progesterone Receptor Activity
Utilizing the "cis-trans" or "co-transfection" assay described by Evans et al., Science, 240:889-95 (May 13, 1988), the two cyclocymopol monomethyl ether diastereomers were tested and found to have activity specifically for the intracellular receptor for progesterone. This assay is described in further detail in U.S. Patent Nos. 4,981,784 and 5,071,773, which are incorporated herein by reference. The co-transfection assay provides a method for identifying functional ligands (either agonists which mimic, or antagonists which inhibit, the effect of hormones) for ligand-responsive receptor proteins.
The co-transfection assay provides a mechanism to evaluate ability of a compound to function as an agonist or antagonist of the activity modulated by an intracellular receptor. The co-transfection assay mimics an in vivo system in the laboratory. In the co-transfection assay, a cloned gene for an intracellular receptor is introduced by transfection (a procedure to induce cells to take up foreign genes) into a background cell substantially devoid of endogenous intracellular receptors. This introduced gene directs the recipient cells to make the intracellular receptor protein. A second gene is also introduced (co-transfected) into the same cells in conjunction with the intracellular receptor gene. This second gene functions as a reporter for the transcription-modulating activity of the target intracellular receptor. The reporter acts as a surrogate for the products normally expressed by a gene under control of the target receptor and its natural hormone.
A preferred reporter gene is one which expresses the firefly enzyme luciferase. The co-transfection assay can detect small molecule agonists or antagonists of target intracellular receptors. Exposing the cells to an agonist ligand increases reporter activity in the transfeeted cells that can be conveniently measured, reflecting ligand-dependent, intracellular receptor-mediated increases in reporter transcription. To detect antagonists, the co-transfection assay is carried out in the presence of a constant concentration of an agonist known to induce a defined reporter signal. Increasing concentrations of a test antagonist will decrease the reporter signal. The co-transfection assay is therefore useful to detect both agonists and antagonists of specific intracellular receptors. It determines not only whether a compound interacts with a particular intracellular receptor, but also whether this interaction mimics (agonizes) or blocks (antagonizes) the effects of the natural regulatory molecules on target gene expression.
Co-transfected cells are exposed to a medium to which is added the potential ligand that is being evaluated. If the candidate ligand diffuses into the cell and binds to the receptor and the resulting complex functions as an agonist, it binds to the co-transfected reporter gene and initiates transcription. When that gene is one that expresses, for example, luciferase, luciferase is produced which catalyzes a light-emitting reaction with its substrate luciferin. Thus, after cell lysis and the introduction of luciferin, the amount of light produced relative to the concentration of candidate ligand used in the assay provides a measure of the potency and efficacy of the compound tested. Antagonist activity is evaluated by adding the candidate ligand and a known agonist to the co-transfected cells. Suppression of agonist-induced luciferase production by the candidate compound, and hence the amount of light produced, indicates the candidate ligand is an antagonist.
The progesterone receptor activity of the cyclocymopol monomethyl ether disastereomer compounds were demonstrated according to the following illustrative example. Example 2
Cultured monkey kidney cells (CV-1's) were transfected with the human receptor cDNA for the progesterone receptor. The receptor cDNA was introduced in a mammalian expression vector under the control of the Rous Sarcoma virus LTR. These vectors provide for the efficient production of the progesterone receptor in these cells, which do not normally express this receptor gene. A reporter vector was also transfected, containing a firefly luciferase gene under the control of the hormone-responsive promoter. Addition of control hormone
(progesterone) or agonist analogues (cyclocymopol compounds) enhanced transcription of the luciferase gene, resulting in an accumulation of the reporter protein luciferase in the cells, whereas antagonists inhibited luciferase production. The level of luciferase activity was then measured in cell extracts. Light emission is directly proportional to the effectiveness of the hormone-receptor complex in activating gene expression.
The cyclocymopol compounds were tested at eight concentrations (10-4 to 10-11M) for the generation of a full dose response curve, and were compared to the progesterone control hormone response. A total of three replicates per concentration point were tested for each compound, and the EC50 was calculated for each positive response. Both agonist and antagonist activity for each test compound was determined in parallel. In the antagonist assay, 10-8M progesterone was added to the media immediately prior to the addition of the cyclocymopol test compounds at the eight concentrations.
The assays showed a 3R (3α) and 3S (3β) diastereomer mixture of cyclocymopol monomethyl ether (designated compound SO-44) to have progesterone receptor antagonist activity, as shown in Figure 2. (For Figure 2, and for Figures 3 through 6, agonist dose response is shown in panel a, and antagonist dose response in panel b). As also shown in Figure 2, the corresponding purified 3R diastereomeric acetate (designated SO-52) exhibited progesterone receptor antagonist activity, and a purified 3S diastereomeric acetate (designated SO-51) exhibited progesterone receptor agonist activity. These assays also showed that a 3R diastereomer of cyclocymopol monomethyl ether (designated SO-53) exhibited progesterone antagonist activity, as shown in Figure 3. In contrast, the 3S diastereomer of cyclocymopol monomethyl ether (SO-54), and its acetate (SO-51), display progesterone receptor agonist activity, as shown in Figure 4.
Compound SO-9 and the identical compound from a separate preparation, compound SO-53, exhibit progesterone receptor antagonist activity (see Figure 5), and were tested for cross reactivity with the other known intracellular receptor classes, e.g., glucocorticoid, mineralocorticoid, androgen, estrogen, and retinoic acid. Compound SO-9 was also tested with orphan receptors (which are receptors whose natural ligand is unknown). The compound was found not to cross-react with any of the other receptors, which demonstrates that activity was limited to the progesterone receptor. Figure 6 is illustrative, and shows that compound SO-9 demonstrated neither agonist nor antagonist activity with the glucocorticoid receptor, dexamethasone.
The 3R and 3S diastereomeric acetates of cyclocymopol monomethyl ether were also individually tested for cross reactivity with the other known intracellular receptor classes. This testing showed the 3R diastereomer to have slight agonist activity with the glucocorticoid receptor. No antagonist activity was detected with either compound.
To investigate the interaction of the cyclocymopol analogs with the human progesterone receptor, and to further demonstrate that they display ligand activity with the progesterone receptor, the analogs were tested for their ability to displace 3H-labeled progesterone from cell extracts containing the human progesterone receptor. These tests were conducted according to the following illustrative example.
Example 3
A plasmid which expresses progesterone receptor was transfected into CV-1 cells by the method of calcium phosphate precipitation. After six hours, the cells were washed and incubated at 37°C with 95% O2/5% CO2 for 40 hours prior to harvest.
After incubation, cells were harvested and washed in phosphate buffered saline. Whole cell receptor extract was prepared by homogenizing the harvested cells in Tris-HCl buffer, pH=7.4, containing 30% glycerol, 1 mM EDTA, 12 mM monothioglycerol, 1 mM PMSF, and 0.5 M potassium chloride. The homogenate was incubated at 4°C for 60 min with resuspension every 10 min. The suspension was centrifuged (105,000 X g, 60 min) and the supernatant was collected and flash frozen in liquid nitrogen and stored frozen at -70°C.
Aliquots of the whole cell extract containing transfected progesterone receptor was incubated at 4°C for 24 hours with a constant concentration (5 nM) of tritiated progesterone and increasing concentrations (0 - 2.5 × 10-5M) of either unlabeled cold progesterone or test compound. The concentration of bound tritiated progesterone was determined in each sample by the dextran-coated charcoal adsorption technique, as follows.
To a 500 1 final volume incubation mixture, 400 1 of 7.5% (w/v) dextran-coated charcoal suspension in gelatin phosphate buffer was added. The mixture was vortexed and incubated at 4°C for 10 min and then centrifuged at 3000 rpm for 10 min. The amount of bound tritiated hormone was determined by liquid scintillation spectrophotometry of an aliquot of the supernatant. As shown in Figure 7a, the purified 3R diastereomer of cyclocymopol monomethyl ether (compound SO-53) and its corresponding acetate (compound SO-52) displaced 3H-progesterone from its ligand binding site in a concentration-dependent manner. Similar binding isotherms were obtained with the purified 3S diastereomeric acetate of cyclocymopol monomethyl ether (compound SO-51), as shown in Figure 7b. These compounds were between 2 and 3 orders of magnitude less potent than the endogenous hormone, progesterone. Similar binding studies were also carried out using the radiolabeled progesterone-agonist 3H-R5020, compound SO-9 (which is identical to compound SO-53), and the progesterone antagonist, RU486. As shown in Figure 7c, RU486 was a competitive antagonist of R5020, whereas compound SO-9 did not compete for this binding site.
Progesterone receptor antagonists, such as RU486, can increase the specific binding of progesterone to its ligand binding site. This enigma was also observed upon performing binding studies with 3H-progesterone in the presence of compound SO-9. As shown in Figure 8, this compound significantly increased the apparent Bmax of 3H-progesterone.
Specificity of binding of progesterone agonists and antagonists has been studied by comparing the ability of various ligands to displace 3H-glucocorticoid from the human glucocorticoid receptor. The data in Figure 9a illustrate that each of the ligands studied has affinity for the glucocorticoid receptor. The affinity of RU486 for the glucocorticoid receptor exceeds that of dexamethasone, and is 300-fold greater than that of compound SO-09. In fact, the affinity of RU486 for the glucocorticoid receptor is comparable to its affinity for the progesterone receptor. The functional activities of the cyclocymopol analogues in the human breast cell line, T47D, were also investigated. T47D cells have proven to be a particularly useful model to investigate the molecular actions of sex steroids because they contain endogenous functional receptors for, and respond to, progestins, and their respective antagonists. Moreover, these cells contain exceptionally high titers of progesterone receptors and are exquisitely sensitive to the actions of progestins in a manner quite similar to their actions in normal and neoplastic mammary epithelial cells. In T47D cells, progestins induce de novo synthesis of a plasma-associated alkaline phosphatase, which has been reported to be similar, if not identical, to the alkaline phosphatase present in the normal breast and human milk. These tests were conducted according to the following illustrative example.
Example 4
T47D cells were cultured in RPMI 1640 medium fortified with 10% fetal bovine serum, 2 mM glutamine, 0.2 ug/ml bovine insulin and, 0.05 mg/ml gentamicin. Cells were plated in 100-mm plates in medium; 48 hours later they were changed to medium containing 2% charcoal-treated serum with or without test compounds in a final ethanol concentration of 0.1%. For routine induction of alkaline phosphatase, cells were treated for three days with two media changes and harvested as described below.
Cells were collected with a rubber policeman into phosphate-buffered saline, pelleted, and lysed with TPSG buffer (0.2% Triton X-100 containing 10 mM sodium phosphate pH-7.4, 0.1 M sucrose, and 10% glycerol) at 0° C for 30 min with vigorous vortex mixing every 5 min. Nuclei were sedimented at 2500 rpm, and the supernatant was saved as cytosol. The protein content of the cytosol was assayed by the method of Bradford. Alkaline phosphatase activity was determined by incubating one volume of cell extract with three volumes of 1 mg/ml p-nitrophenol phosphate prepared in DEAM (1 M diethanolamine, pH=9, containing 2 mM magnesium chloride) at room temperature for 20 min. At the end of the incubation time, the reaction was stopped with an equal volume of IN NaOH. Ten standards of p-nitrophenol were prepared in DEAM buffer and absorbance was read at 405 nm. Alkaline phosphatase activity is expressed as pmol p-nitrophenol formed/min/mg protein.
As shown in Figure 10a, (35)-cyclocymopol monomethyl ether (compound SO-54), and its corresponding acetate (compound SO-51), are functional agonists in this system, whereas (3R)-cyclocymopol monomethyl ether (compound SO-53), and its corresponding acetate (compound SO-52), appear to be very weak agonists. The 3S form compounds exhibit increased efficacy at higher concentrations, an effect which has been found to be reproducible.
To determine if (3R)-cyclocymopol monomethyl ether could function as a progesterone receptor antagonist, the effects of increasing concentrations of this compound on R5020-induced alkaline phosphatase activity were quantified in T47D cells. As shown in Figures 10b and 10c, (3R)-cyclocymopol monomethyl ether (compound SO-53) is an effective antagonist of the progesterone mimic. Similarly, when alkaline phosphatase activity was stimulated by 1.0 nM progesterone, (3R)-cyclocymopol monomethyl ether (compound SO-53) attenuated this induction in a concentration-dependent manner.
In comparison, the progesterone agonist, (35)-cyclocymopol monomethyl ether (compound SO-54), and its acetate (SO-51), increased the activity of intracellular alkaline phosphatase in a concentration-dependent manner, and this was blocked by the progesterone antagonist, RU486, as shown in Figure 11. In this system, (3R)- cyclocymopol monomethyl ether (compound SO-53) functioned as a progesterone receptor antagonist and attenuated the effects of progesterone in a concentration- dependant manner.
Synthetic and semisynthetic cyclocymopol analogs have been prepared which also have activity for the intracellular receptor for progesterone. Representative analogs of the present invention are prepared according to the following illustrative synthetic schemes and illustrative examples.
Example 5 - Synthesis of Aromatic Subunit
2-Acetoxy-5-methoxybenzaldehyde (1):
To a flame-dried 50 mL round-bottomed flask containing 10.00 g (65.7 mmol) 2-hydroxy-5-methoxybenzaldehyde in 10 mL dry pyridine at 0°C under nitrogen atmosphere was added 7 mL acetic anhydride. The reaction mixture was then allowed to warm to room temperature and continually stirred until TLC analysis indicated complete consumption of starting material (50 min). Ethyl acetate
(150 mL) was added, and the mixture was then transferred to a separatory funnel and successively washed with IN HCl (3 × 50 mL), saturated aqueous ΝaHCO3 (1 × 50 mL), and brine (1 × 50 mL), dried over Na2SO4, and concentrated under reduced pressure to give 12.41 g (97%) of the acetylated phenol as a white solid. The product thus obtained was homogenous by TLC (Rf 0.41, 2:1 hexane/ethyl acetate), and was carried on to the next step without further purification.
2-Acetoxy-4-bromo-5-methoxybenzaldehyde (2):
To a 500 mL round-bottomed flask containing a solution of 20.0 g (168.1 mmol, 3.26 equiv) potassium bromide and 3.21 mL (10.0 g, 62.6 mmol, 1.21 equiv) bromine in 200 mL water at room temperature was added 10.00 g (51.5 mmol, 1.0 equiv) 2-acetoxy-5-methoxybenzaldehyde (1) as a finely divided white powder, portionwise over a period of 35 min. After 18 h stirring at room temperature, the reaction mixture was filtered under vacuum using a Bύchner funnel to give 10.59 g (89%) of the aryl bromide as a pale yellow solid (Rf 0.45, 2:1 hexanes/ethyl acetate). The product thus obtained was of greater than 98% purity by 1H NMR, and homogenous by TLC, and was carried on to the next step without further purification. A portion of the crude product was recrystallized from 5:1 ether/hexanes to give white needles.
2-Hydroxy-4-bromo-5-methoxybenzaldehyde (3):
To a 200 mL round-bottomed flask containing 6.90 g (25.3 mmol) 2 acetoxy-4-bromo-5-methoxybenzaldehyde (2) in 100 mL of 1% aqueous methanol at room temperature was added 5 g K2CO3, and the mixture was allowed to stir at room temperature for 1 h, at which time TLC analysis indicated complete consumption of starting material and the presence of a slightly more polar compound as the only detectable product. The reaction mixture was then neutralized to pH 5 with the addition of 1 N HCl, and the solvent was subsequently removed under diminished pressure. The residue was then dissolved in ethyl acetate
(200 mL), washed successively with 1 N HCl (1 × 50 mL), saturated aqueous NaHCO3 (1 × 50 mL), and brine (1 × 50 mL), dried over Na2SO4, and concentrated under diminished pressure to give 4.97 g (85%) of the bromophenol as a brownish-red oily solid. A portion of this crude material was recrystallized from 2:1 hexanes/ethyl acetate to give white needles. The remainder of the material was carried on to the next step without further purification.
2-(tert-Butyl)dimethylsilyloxy-4-bromo-5-methoxybenzaldehyde (4):
To a flame-dried 100 mL round-bottomed flask containing 2.76 g (11.95 mmol) 2-hydroxy-4-bromo-5-methoxybenzaldehyde (3) in 50 mL anhydrous dichloromethane under nitrogen atmosphere at room temperature was added 2.03 g (29.88 mmol, 2.50 equiv) imidazole, 2.25 g (14.94 mmol, 1.25 equiv) (tert-Butyl)- dimethylchlorosilane, and DMAP (100 mg, catalytic). The mixture was allowed to stir at room temperature for 75 min, at which time TLC analysis indicated complete consumption of starting material, and the formation of a less polar product (Rf 0.75, 2:1 hexanes/ethyl acetate). The reaction mixture was then poured into a separatory funnel containing 50 mL dichloromethane and 50 mL saturated aqueous NH4Cl, the layers were separated, and the organic phase was washed with 50 mL brine, dried over Na2SO4, and concentrated under diminished pressure to give 4.13 g (quantitative) of the silylated bromophenol as an off-white solid, a portion of which was recrystallized from 3:1 hexanes/ether to give an amorphous white solid.
2-(tert-Butyl)dimethylsilyloxy-4-bromo-5-methoxybenzyl alcohol (5):
To a flame-dried 200 mL round-bottomed flask containing 4.10 g (13.14 mmol) 2-(ten-butyl)dimethylsilyloxy-4-bromo-5-methoxybenzaldehyde (4) in 100 mL anhydrous methanol at 0°C was added 0.50 g (13.15 mmol, 1.0 mol equiv) NaBH4 over a period of 3 min. After 20 min at 0°C, TLC analysis indicated complete consumption of starting material, and the formation of a more polar product (Rf 0.42, 2:1 hexanes/ethyl acetate). Water (75 mL) was added, and the methanol was removed by rotary evapora-tion. The resultant aqueous residue was extracted with ethyl acetate (2 × 100 mL), and the combined organic layers were dried over Na2SO4, and concentrated under diminished pressure. Purif-ication by flash column chromatography (silica gel, hexanes/ethyl acetate, 5:1) afforded 4.10 g (98%) of the benzylic alcohol as a white solid.
2- (tert-Butyl) dimethylsilyloxy-4-bromo-5-methoxybenzyl bromide (6):
To a flame-dried 100 mL round-bottomed flask containing triphenylphosphine (1.27 g, 4.84 mmol, 1.05 equiv) in 25 mL anhydrous DMF at 0°C under nitrogen atmosphere was added bromine (0.24 mL, 4.84 mL, 1.05 equiv, plus enough extra to cause a persistent reddish tint to the solution, 1 drop) through an additional funnel. To this reaction mixture was added 2-(tert-butyl) dimethylsilyloxy-4-bromo-5-methoxybenzyl alcohol (5) through the addition funnel at a steady rate over 30 min, as a solution in 10 mL DMF. The reaction mixture was allowed to stir at 0°C for 60 min, at which time TLC analysis indicated complete consumption of starting material, and the formation of a less polar product (Rf 0.81, 2:1 hexanes/ethyl acetate). Hexane (100 mL) was then added, and the contents of the flask were transferred to a separatory funnel containing 50 mL of saturated aqueous NH4Cl, rinsing with an additional 50 mL hexane and 10 mL water. The layers were separated, and the organic phase was washed with 20 mL 10% Na2S2O3, dried over Na2SO4, and concentrated under diminished pressure. Purification by trituration at 0°C (2 × 30 mL ea. hexane) to remove residual triphenylphosphine oxide, followed by flash column chromatography (silica gel, hexane/ethyl acetate, gradient elution) afforded 1.51 g (80%) of the benzylic bromide as a colorless, viscous oil. 1H NMR (400 MHz, CDCl3) δ 0.28 [s, 6H, Si(CH3)2], 1.05 [s, 9H,
SiC(CH3)3], 3.87 (s, 3H, OCH3), 4.48 (s, 2H CH2Br), 6.79 and 7.01 ppm (2s, 2 × 1H, Ar-H).
Figure imgf000030_0001
Example 6 - Synthesis of Aliphatic Subunits
3-Ethoxy-5,5-dimethylcyclohex-2-en-1-one (7):
To a flame-dried 1 L round-bottomed flask containing 15.0 g (107 mmol) 5, 5-dimethylcyclohexane-1, 3-dione and 120 mL absolute ethanol in 300 mL anhydrous benzene under nitrogen atmosphere was added 750 mg p-toluenesulphonic acid monohydrate (catalytic). The flask was fitted with a Dean Stark trap for removal of water, and a reflux condenser, and the mixture was heated to reflux for 9 h. Upon cooling to room temperature, the solvent was removed by rotary evaporation, and the residue was dissolved in 300 mL ethyl acetate. The organic solution was then washed successively with 10% aqueous NaOH (2 × 100 mL), water (1 × 100 mL), and brine (1 × 100 mL), dried over Na2SO4, ahd concentrated under diminished pressure to give 16.5 g (92%) of the keto-enol ether as a pale yellow oil (Rf 0.24, 2:1 hexanes/ethyl acetate) of greater than 98% purity by 1H NMR.
5.5-Dimethylcyclohex-2-en-1-one (8):
To a flame-dried 100 mL round-bottomed flask containing lithium aluminum hydride (0.95 g, 24.4 mmol, 0.5 mol equiv) in 35 mL anhydrous ether under nitrogen atmosphere at 0°C was added 8.20 g (48.7 mmol) 3-ethoxy-5,5-dimethylcyclohex-2-en-1-one (7) portionwise through a syringe as a solution in 10 mL anhydrous ether. The reaction mixture was allowed to warm to room temperature, and after 4h, TLC analysis indicated complete consumption of starting material. The reaction mixture was then cooled to 0°C before the cautious addition of 50 mL water, and the contents of the flask were then poured into a 500 mL Erlenmeyer flask containing 150 mL ice-cold 10% H2SO4. The mixture was then extracted with ether (2 × 200 mL), and the combined organics were washed successively with water (100 mL), and saturated aqueous NaHCO3 (100 mL), dried over Na2SO4, and concentrated under diminished pressure to give 6.05 g (quantitative) of the dimethylenone (Rf 0.55, 2:1 hexane/ethyl acetate). 1H NMR (400 MHz, CDCl3) δ 1.05 (s, 6H, geminal-CH3's), 2.23 (dd, 2H, CHCH2) , 2.28 (s, 2H, COCH2), 6.03 (ddd, 1H, 2-H), 6.87 ppm (ddd, 1H, 3-H).
3-Ethoxy-6,5,5-trimethylcyclohex-2-en-1-one (9):
To a flame-dried 300 mL round-bottomed flask containing diisopropylamine (1.83 mL, 13.06 mmol, 1.1 equiv) in 50 mL anhydrous THF at -78°C under nitrogen atmosphere was added n-butyllithium (5.70 mL of a 2.2 M solution in hexane, 12.50 mmol, 1.05 equiv). After 20 min at -78°C, 3-ethyoxy-5,5-dimethylcyclohex-2-en-1-one (7) was added as a solution in 3 mL THF, and the reaction mixture was allowed to stir at that temperature for 15 min before gradual warming to 0°C, and subsequent addition of iodomethane (3.5 mL, 59.5 mmol, 5 equiv). The reaction mixture was then allowed to warm to room temperature, and after 3h, was quenched which saturated aqueous NH4Cl. The contents of the flask were then extracted with 100 mL ethyl acetate, and the organic phase was washed successively with water (50 mL) and brine (50 mL), dried over Na2SO4, and concentrated under diminished pressure. Purification by flash column chromatography (silica gel, hexane/ethyl acetate, gradient elution) afforded 1.94 g (90%) of the methylated keto-enol ether (Rf 0.40, 2:1 hexanes/ethyl acetate) as a colorless oil.
4,5,5-Trimethylcyclohex-2-en-1-one (10):
This compound was prepared from 3-ethoxy-6, 5,5-trimethylcyclohex-2-en-1-one (9) (1.60 g, 8.80 mmol) in the manner previously described for enone 8, yielding 1.03 g (85%) of the methylated enone as a colorless oil. 1H NMR (400 MHz, CDCl3) δ 0.90 and 1.07 (2s, 2 × 3H, geminal- CH3's), 1.10 (d, 3H, CHCH3), 5.96 (dd, 1H, 2-H), 6.16 ppm (dd, 1H, 3-H).
Figure imgf000033_0001
Synthesis of Cyclocymopol Analogs
Example 7
6-[2 '-(tert-Butyl)dimethylsilyloxy-4'-bromo- 5'methoxyphenyl] methyl-5,5-dimethylcyclohex-2-en-1-one (11):
To a flame-dried 50 mL round-bottomed flask containing diisopropylamine (0.188 mL, 1.34 mmol, 1.1 equiv) in 10 mL anhydrous THF at -78°C under nitrogen atmosphere was added n-butyllithium (0.58 mL of a 2.2 M solution in hexane, 1.28 mmol, 1.05 equiv). After 20 min at -78°C, 5,5-dimethylcyclohex-2-en-one (8) (0.151 g, 1.22 mmol) was added as a solution in 1 mL of THF, and the reaction mixture was allowed to stir at that temperature for 15 min, at which time the cooling bath was removed. When the temperature of the reaction mixture (monitored using a thermocouple probe) reached -5°C, 2-(tert-butyl)dimethylsilyloxy-4-bromo-5-methoxybenzylbromide (6)
(1.00 g, 2.44 mmol, 2.0 equiv) was added all at once as a solution in 2 mL THF. The reaction mixture was then allowed to warm to room temperature, and after 3 h, TLC analysis indicated complete consumption of the enone starting material, and the formation of a product of intermediate polarity with respect to the two starting components (Rf 0.59, 2:1 hexanes/ethyl acetate), and the reaction was quenched by the addition of 5 mL saturated aqueous NH4Cl. The contents of the flask were transferred to a separatory funnel, and extracted with 60 mL ethyl acetate, and the resultant organic phase was washed with 30 mL brine, dried over Na2SO4, and concentrated under diminished pressure. Purification by flash column chromatography (silica gel, hexane/ethyl acetate, gradient elution) afforded 0.459 g (83%) of the benzylated enone as a colorless, viscous oil, which solidified on standing.
2-[2 '-(tert-Butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]methyl-3,3-dimethylcyclohexanone (12):
To a flame-dried 100 mL round-bottomed flask containing 6-[2'-(tert-butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]methyl-5,5-dimethylcyclohex-2-en-1-one (11) (0.154 g, 0.454 mmol) in 22 mL ethyl acetate (which had been pre-dried over K2CO3) at room temperature was added 20 mg 5% palladium on carbon, and after flushing/evacuating the vessel 3 times with nitrogen, a hydrogen atmosphere was introduced and maintained by use of a balloon. After
24 h, the flask was again flushed several times with nitrogen, and the contents of the flask were filtered, rinsing with an additional 100 mL ethyl acetate. Rotary evaporation of the solvent afforded 0.156 g (quantitative) of the saturated benzylic ketone as a colorless, viscous oil (Rf 0.67, 2:1 hexane/ethyl acetate).
1-Methylidene-2-(2'-hydroxy-4'-bromo-5'-methoxyphenyl)methyl-3,3-dimethylcyclohexane (racemic 1-desbromocyclocvmopol monomethyl ether) (13):
To a flame-dried 50 mL round-bottomed flask containing 2-[2'-(tert-butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]methyl-3,3-dimethylcyclohexanone (12) (0.130 g, 0.28 mmol) in 5 mL anhydrous THF at -78°C under nitrogen atmosphere was added (trimethyl)silymethyllithium (0.420 mL of a 1.0 M solution in pentane, 0.42 mmol, 1.50 equiv). An immediate change from colorless to a yellow reaction solution was observed, and TLC analysis at that time indicated complete consumption of starting material, and the formation of a less polar product (Rf 0.79, 2:1 hexanes/ethyl acetate), and the reaction was subsequently quenched with 4 mL saturated aqueous NH4Cl. Ethyl acetate
(30 mL) extraction of the reaction mixture, drying over
Na2SO4, and concentration under diminished pressure gave
0.152 g (quantitative) of a crude product, which appeared to be a single diastereomer of the ketone addition product by 1H NMR analysis. A portion of this crude intermediate
(0.015 g, 0.028 mmol) was placed in a 10 mL nalgene vial containing 2 mL THF, 0.2 mL of a pre-made HF/pyridine complex was added, and the mixture was allowed to stir at room temperature for 32 h, at which time TLC analysis indicated complete consumption of starting material, and formation of a more polar product, having passed through a most polar intermediate (confirmed as the desilylated phenol by 1H NMR). The contents of the reaction vessel were transferred to a separatory funnel containing 20 mL ethyl acetate and 10 mL 1.0 M NaHSO4. The layers were separated, and the resultant organic phase was washed with 10 mL brine, dried over Na2SO4, and concentrated under diminished pressure. Purification by flash column chromatography (silica gel, hexane/ethyl acetate, gradient elution) afforded 8.7 mg (92%) of the phenolic olefin as a colorless oil. 1H NMR (400 MHz, CDCl3) δ 0.98 and 1.00
(2s, 2 × 3H, geminal-CH3's), 2.64 and 2.80 (d of ABq, 2H, benzylic-CH2), 3.80.(s, 3H, OCH3) 4.36 and 4.63 (2s, 2 × 1H, olefinic-CH2), 6.58 and 6.95 ppm (2s, 2 × 1H, Ar-H).
(This compound is also referred to as Compound "C" below.)
Figure imgf000037_0001
Example 8
1-Methylidene-6- (2' -acetoxγ-4' -broao-5'-methoxyphenyl)methyl-5,5-dimethylcyclohex-2-ene (14):
This compound was prepared from 6-[2'-(tert-butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]methyl-5,5-dimethylcyclohex-2-en-1-one (11) (0.075 g, 0.166 mmol) in the manner previously described for olefin (13), with the following procedural changes necessitated by the incompatibility of structural features particular to this substrate and the typical synthetic methodology. Upon formation of the initial (trimethyl) silylmethyllithium addition adduct to enone 11, the phenolic protecting group was exchanged prior to effecting elimination, using the following protocol adhered to for all cyclocymopol analogs possessing a 1,3-diene moiety as an extension of the methylidene olefin. The crude addition product (0.090 g, 0.166 mmol) was dissolved in 5 mL anhydrous THF containing 0.20 mL acetic anhydride (large excess), and cooled to 0°C under nitrogen atmosphere. Tetra-n-butylammonium fluoride (0.20 mL of a 1.0 M solution in THF, 0.20 mmol, 1.20 equiv) was added, and the mixture was allowed to warm to room temperature. The contents of the flask were then poured into a separatory funnel containing 30 mL ethyl acetate and 10 mL 1.0 M NaHSO4, the layers were separated, and the organic phase was washed with 10 mL brine, dried over Na2SO4, and concentrated under diminished pressure. The crude material thus obtained was immediately carried on to the next step by transferring to a 10 mL nalgene vial containing 2-3 mL THF, and 0.3 mL premade HF/pyridine complex was added. After stirring overnight at room temperature, the reaction mixture was worked up in the usual manner, and purification by flash column chromatography (silica gel, hexane/ethyl acetate, gradient elution) afforded 38.3 mg (64%) of the desired acetoxydiene as a colorless, oily solid. 1H NMR (400 MHz, CDCl3) δ 0.73 and 1.11 (2s, 2 × 3H, geminal-CH3's), 2.27 (s, 3H, acetate-CH3), 3.83 (s, 3H, OCH3), 4.13 and 4.68 (2s, 2 × 1H, C=CH2), 5.70 and 6.04 (2dd, 2 × 1H, 2-H, 3-H), 6.52 and 7.19 ppm (2s , 2 × 1H, Ar-H).
(This compound is also designated Compound "L" below.) Example 9
1-Methylidene-6-(2'-hydroxy-4'-bromo-5'-methoxyphenyl)methyl-5,5-dimethylcvclohex-2-ene (15):
In a 10 mL test tube was combined 1-methylidene-6- (2'-acetoxy-4'-bromo-5'-methoxyphenyl)methyl-5,5-dimethylcyclohex-2-ene (14) (10.0 mg, 0.026 mmol) and 2.0 mL of 5% methanolic K2CO3. After 10 min at room temperature, the methanol was removed by rotary evaporation, and the resultant residue was dissolved in 20 mL ethyl acetate. The organic solution was then washed with saturated aqueous NH4Cl, dried over Na2SO4, and concentrated under diminished pressure. Purification by flash column chromatography (silica gel deactivated with triethylamine, hexane/ethyl acetate, gradient elution) afforded 7.1 mg (81%) of the phenolic diene as a colorless, oily solid. 1H NMR (400 MHz, CDCl3) δ 0.73 and 1.16 (2s, 2 × 3H, geminal-CH3's), 3.82 (s, 3H, OCH3), 4.23 and 4.72 (2s, 2 × 1H, C=CH2), 5.78 and 6.08 (2dd, 2 × 1H, 2-H, 3-H), 6.51 and 6.99 ppm (2s, 2 × 1H, Ar-H).
(This compound is also designated Compound "K" below.)
Example 10
6- [ 2 ' - (ten-Butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]methyl-3,5,5-trimethyl-cvclohex-2-en-1-one (16):
This compound was prepared from isophorone (0.168 g, 1.22 mmol) in the manner previously described for enone 11, affording 0.342 g (60%) of the alkylation product (Rf 0.40, 2:1 hexane/ethyl acetate) as a colorless, oily solid. 1-Methylidene-6-(2'-acetoxy-4'-bromo-5'-methoxyphenyl)methyl-3,5,5-trimethylcvclohex-2-ene (17):
This compound was prepared from 6-[2'-(tert-butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]methyl-3,5,5-trimethylcyclohex-2-en-1-one (16) (42.0 mg, 0.090 mmol) in the manner previously described for acetoxy diene 14, affording 18.4 mg (52%) of the acetoxy-diene as a colorless oil. 1H NMR (400 MHz, CDCl3) δ 0.87 and 1.11
(2s, 2 × 3H, geminal-CH3's), 1.78 (s, 3H, olefinic-CH3), 2.26 (s, 3H, acetate-CH3), 3.83 (s, 3H, OCH3), 4.03 and
4.58 (2s, 2 × 11, C=CH2), 5.82 (s, 1H, 2-H), 6.52 and 7.19 ppm (2s, 2 × 1H, Ar-H).
(This compound is also designated Compound "H" below.) Example 11
1 -Methylidene-6-(2'-hvdroxy-4'-bromo-5'-methoxyphenyl)methyl-3,5,5-trimethylcyclohex-2-ene (18):
This compound was prepared from 1-methylidene-6-(2'-acetoxy-4'-bromo-5'-methoxyphenyl)methyl-3,5,5-trimethylcyclohex-2-ene (17) (6.8 mg, 0.017 mmol) in the manner previously described for phenolic diene 15, affording 5.8 mg (96%) of the phenolic diene as a colorless, oily solid. 1H NMR (400 MHz, CDC13) δ 0.74 and 1.15 (2s, 2 × 3H, geminal-CH3's), 1.79 (s, 3H, olefinic-CH3), 2.26 (s, 3H, acetate-CH3), 3.81 (s, 3H, OCH3), 4.16 and 4.62 (2s, 2 × 1H, C=CH2), 5.87 (s, 1H, 2-H), 6.52 and 6.99 ppm (2s, 2 × 1H, Ar-H).
Figure imgf000041_0001
Example 12
trans-6- [2 ' -(tert-Butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]methyl-4,5,5-trimethylcyclohex-2-en-1-one (19):
This compound was prepared from 4,5,5-trimethylcyclohex-2-en-1-one (10) (169 mg, 1.22 mmol) in the manner previously described for benzylated enone 11, affording 0.337 g (59%) of the less polar trans diastereomer as a colorless, oily solid, along with 22 mg (4%) of the more polar cis diastereomer as a colorless, oily solid, separable by flash column chromatography. The relative stereochemistry of each respective diastereomer was confirmed by nOe NMR experiments.
trans-2- [2'-(tert-Butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]methyl-3,3,4-trimethyleyelohexanone (20):
This compound was prepared from trans-6-[2'-(tert-butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]methyl-4,5,5-trimethylcyclohex-2-en-1-one (19) (150 mg, 0.321 mmol) in the manner previously described for benzylated ketone 12, affording 0.149 g (99%) of the trans ketone as a colorless, oily solid.
trans-1-Methylidene-6-(2 '-acetoxy-4'-bromo-5'-methoxyphenyl)methyl-4,5,5-trimethylcyclohex-2-ene (21):
This compound was prepared from trans-6-[2'-(tert-butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]methyl-4,5,5-trimethylcyclohex-2-en-1-one (19) (70.0 mg, 0.15 mmol) in the manner previously described for acetoxy-diene 14, affording 46.8 mg (79%) of the acetoxy-diene as a colorless oil. 1H NMR (400 MHz, CDCl3) δ 0.76 and 1.14 (2s, 2 × 3H, geminal-CH3's), 1.00 (d, 3H, CHCH3), 2.30 (s, 3H, acetate-CH3), 3.85 (s, 3H, OCH3), 4.12 and 4.67 (2s, 2 × 1H, C=CH2), 5.46 (d, 1H, 2-H), 6.01 (dd, 1H, 3-H), 6.53 and 7.21 ppm (2s, 2 × 1H, Ar-H).
(This compound is also designated Compound "N" below.) Example 13
trans-1-Methylidene-6-(2'-hydroxy-4'-bromo-5'-methoxyphenyl)methyl-4,5,5-trimethylcyclohex-2-ene (22) :
This compound was prepared from trans-1-methylidene-6- (2'-acetoxy-4'-bromo-5'-methoxyphenyl)methyl-4,5,5-trimethylcyclohex-2-ene (21) (5.5 mg, 0.014 mmol) in the manner previously described for phenolic diene 15, affording 4.1 mg (84%) of the phenolic diene as a colorless, oily solid. 1H NMR (400 MHz, CDCl3) δ 0.74 and 1.18 (2s, 2 × 3H, geminal-CH3's), 1.00 (d, 3H, CHCH3) , 3.80
(s, 3H, OCH3) , 4.22 and 4.70 (2s, 2 × 1H, C=CH2) , 5.52
(d, 1H, 2-H) , 6.04 (dd, 1H, 3-H), 6.50 and 6.99 ppm (2s, 2
× 1H, Ar-H) .
Figure imgf000044_0001
Example 14
trans-1-Methylidene-2 - (2 ' -hydroxy-4 ' -bromo-5 , -methoxyphenyl)methyl-3 , 3 , 4 -trimethylcyclohexane (23) :
This compound was prepared from trans-2 -[2'-(tert-butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]methyl-3,3,4-trimethylcyclohexanone (20) (47.0 mg, 0.089 mmol) in the manner previously described for phenolic olefin 13 to afford 22.6 mg (72%) of the trans phenolic olefin as a colorless, oily solid. 1H NMR (400 MHz, CDCl3) δ 0.79 and 1.08 (2s, 2 × 3H, geminal-CH3's), 0.85 (d, 3H, CHCH3), 3.80
(s, 3H, OCH3), 4.36 and 4.58 (2s, 2 × 1H, C=CH2), 6.50 and
6.99 ppm (2s, 2 × 1H, Ar-H).
Example 15
6-(4'-Nitrophenyl)methyl-5,5-dimethylcyclohex-2-en-1-one (24):
This compound was prepared from 5,5-dimethyleyelohex-2-en-1-one (8) (0.600 g, 4.83 mmol) and p-nitrobenzyl bromide (1.581 g, 7.32 mmol) in the manner described for the synthesis of enone 11, affording 627 mg (50%) of the nitro-enone as a pale yellow oil. 1H NMR (400 MHz, CDCl3) δ 1.02 and 1.18 (2s, 2 × 3H, geminal-CH3's), 2.83 and 3.10 (d of ABq, 2H, benzylic-H's), 6.00 (ddd, 1H, 2-H), 6.81 (ddd, 1H, 3-H), 7.38 and 8.10 ppm (2d, 2 × 2H, Ar-H).
Example 16
1-Methylidene-6-(3'-methyl-4'-nitrophenyl)methyl-5,5-dimethylcyclohex-2-ene (25):
This compound was prepared from 6-(4'-nitrophenyl)methyl-5,5-dimethylcyclohex-2-en-1-one (24) (133 mg, 0.514 mmol) in the manner previously described for the synthesis of olefin 13, with the following procedural changes. Three equivalents of (trimethyl) silylmethyllithium were used, and the subsequent elimination step required 48 h to go to completion, affording 120 mg (86%) of the nitrodiene as a colorless oil. 1H NMR (400 MHz, CDCl3) δ 0.92 and 1.12 (2s, 2 × 3H, geminal-CH3's), 2.58 (s, 3H, Ar-CH3), 4.05 and 4.64 (2s, 2 × 1H, methylidene-CH2), 5.70 (ddd, 1H, 3-H), 6.03 (dd, 1H, 2-H), 6.98 (s, 1H, Ar-H), 6.99 and 7.88 ppm (2d, 2 × 1H, Ar-H).
(This compound is also designated Compound "M" below.)
Example 17
1-Methylidene-2-(2 '-hydroxy-4'-bromo-5'-methoxyphenyl)methylcyclohexane (26):
This compound was prepared in three steps from cyclohexanone and 2-(tert-butyl)dimethylsilyloxy-4-bromo-5-methoxybenzyl bromide (6) as previously described for the synthesis of olefin 13, to give the desired olefin in three steps in 13.5% overall yield as a colorless oil. 1H NMR (400 MHz, CDCl3) δ 2.54 and 2.92 (d of ABq, 2H, benzylic-H's), 3.84 (s, 3H, OCH3), 4.46 (s, 1H, OH), 4.62 and 4.71 (2s, 2 × 1H, methylidene-CH2), 6.65 and 6.98 ppm (2s, 2 × 1H, Ar-H).
Figure imgf000047_0001
Semisynthetic Derivatives of Natural Cyclocymopols
Example 18
(3R)-1-Debromocyclocymopol monomethyl ether (27):
To a flame-dried 10 mL round-bottomed flask containing (3R)-2'-(tert-butyl)dimethylsilyloxycyclocymopol monomethyl ether (19.5 mg, 0.036 mmol) in 1 mL anhydrous benzene with 1-2 mg AIBN at room temperature was added n-Bu3SnH (39 μL, 0.144 mmol, 4.0 equiv). After 90 min, TLC analysis indicated virtually complete consumption of starting material, and formation of a slightly less polar product. Carbon tetrachloride (200 μL) was added, and after 1 h at room temperature followed by 1.5 h at 0°C, 2 mL THF and 200 μL 1.0 M tetrabutylammonium fluoride solution in THF were added. After 10 min at 0°C, pH 7 buffer was added, and the reaction mixture was extracted with hexane. The resultant organic solution was dried over Na2SO4, and concentrated under diminished pressure. Purification by flash column chromatography (silica gel, 10% ethyl acetate in hexane) afforded 7.5 mg (61%) of the debromophenol as a colorless oil. The 400 MHz 1H NMR spectrum and TLC elution properties of this compound were identical to those reported for the racemic analog 13.
(This compound is also designated Compound "J" below.) Example 19
(35)-1-Debromocyclocymopol monomethyl ether (28):
This compound was prepared from (35)-2'-(tert-butyl)dimethylsilyloxycyclocymopol monomethyl ether (25.0 mg, 0.047 mmol) in the manner described for the synthesis of the cyclocymopol derivative 27, affording 5.5 mg (35%) of the debromophenol as a colorless oil, along with the remainder of the mass balance as deprotected starting material. The 400 MHz 1H NMR spectrum and TLC elution properties of this compound were identical to those reported for the racemic analog 13. (This compound is also designated Compound "G" below.)
Example 20
(3R)-4'-Formylcyclocymopol monomethyl ether (29):
To a flame-dried 10 mL round-bottomed flask containing (3R)-2'-tert-butyl)dimethylsilyloxycyclocymopol monomethyl ether (53.4 mg, 0.10 mmol) in 2 mL anhydrous THF at -78 °C was added n-butyllithium ( 70 μL of a 2 . 15 M solution in hexane, 0.15 mmol, 1.50 equiv) all in one portion. After 10 min at -78°C, N-methyl-N-(2-pyridyl) formamide (28.5 mg, 25 μL, 0.21 mmol, 2.1 equiv) was added as a solution in 1 mL THF, and the reaction mixture was allowed to stir for 30 min before quenching with 1 mL 1:4 acetic acid/methanol. The reaction mixture was then partitioned between hexane and 1.0 M NaHSO4, washed with pH 7 buffer, and the resultant organic phase was dried over Na2SO4 and concentrated under diminished pressure. Purification by radial chromatography (silica gel, 1 mm chromatotron plate, 10-15% ethyl acetate in hexane) gave 28.1 mg (58%) of the protected phenolic aldehyde as a white solid. A portion of this silylated phenol (3.5 mg) was deprotected using 25 μL of 1.0 M tetrabutylammonium fluoride in 1 mL THF at 0°C. The reaction was quenched with pH 7 buffer, and partitioned between hexanes and water. The resultant organic phase was dried over Na2SO4 and concentrated under diminished pressure. Purification by radial chromatography (silica gel, 1 mm chromatotron plate, 15% ethyl acetate in hexanes) gave 2.1 mg (79%) of the desired aldehyde as a white solid. 1H NMR (400 MHz, CDCl3) δ 1.10 and 1.26 (2s, 2 × 3H, geminal-CH3's), 2.70 and 3.01 (d of ABq, 2H, benzylic-H's), 3.84 (s, 3H, OCH3), 4.39 and 4.62 (2s, 2 × 1H, methylidene-CH2), 4.44 (dd, 1H, BrCH), 6.60 and 7.27 (2s, 2 × 1H, Ar-H), 10.33 ppm (s, 1H, CHO). Example 21
(3R)-4 '-Iodocyclocymopol monomethyl ether (30):
This compound was prepared from (3R)-2'-(tert-butyl)dimethylsilyloxycyclocymopol monomethyl ether (15.2 mg, 0.029 mmol) and iodine (300 μL of a 0.25 M solution in benzene, 0.075 mmol, 2.6 equiv) in the manner previously described for cyclocymopol derivative 29, affording 9.2 mg (70%) of the iodocyclocymopol derivative as a colorless oily solid. 1H NMR (400 MHz, CDCl3) δ 1.08 and 1.22 (2s, 2 × 3H, geminal-CH3's), 2.52 and 2.91 (d of ABq, 2H, benzylic-H's), 3.78 (s, 3H, OCH3) , 4.30 and 4.62 (2s, 2 × 1H, methylidene-CH2), 4.42 (dd, 1H, ICH), 4.49 (s, 1H, OH), 6.45 and 7.10 ppm (2s, 2 × 1H, Ar-H).
(This compound is also designated Compound "V" below.)
Example 22
(3R,5R)-5-Hydroxycyclocymopol monomethyl ether (31):
To a flame-dried 25 mL round-bottomed flask containing (3R)-2'-(tert-butyl)dimethylsilyloxycyclocymopol monomethyl ether (13.8 mg, 0.026 mmol) in 3.5 mL anhydrous dichloromethane at room temperature was added selenium dioxide (1.5 mg, 0.013 mmol, 0.50 equiv) and anhydrous t-butyl hydroperoxide (17.3 μL of a 3.0 M solution in 2,2,4-trimethylpentane, 0.052 mmol, 2.00 equiv), and the mixture was allowed to stir at room temperature for 40 h, at which time the solvent was removed by rotary evaporation. Purification by flash column chromatography (silica gel, hexanes/ethyl acetate, gradient elution) afforded 13 mg of material which was dissolved in 1 mL THF, cooled to 0°C, and treated with tetrabutylammonium fluoride (0.03 mL of a 1.0M solution in THF, 0.030 mmol, 1.15 equiv). After 10 min, the reaction was quenched by the addition of pH 7 buffer (5 mL), and extracted with hexanes (2 × 25 mL). The combined organic extracts were dried over. Na2SO4, and concentrated under diminished pressure. Purification by flash column chromatography (silica gel, hexanes/ethyl acetate, gradient elution) afforded 8.8 mg (78%) of the hydroxycyclocymopol as a white sold. 1H NMR (400 MHz, CDCl3) δ 1.04 and 1.23 (2s, 2 × 3H, geminal-CH3's), 3.83 (s, 3H, OCH3), 4.42 (dd, 1H, CHBr), 4.43 and 4.96 (2s, 2 × 2H, methylidene-CH2), 4.73 (dd, 1H, CHOH), 6.54 and 7.00 ppm (2s, 2 × 1H, Ar-H).
(This compound is also designated Compound "E" below.)
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Utilizing the "co-transfection" assay described above, representative synthetic and semisynthetic cyclocymopol derivatives have been tested and found to be antagonists specifically for the intracellular receptor for progesterone. Cultured monkey kidney cells (CV-1's) were transfected with the human receptor cDNA for the progesterone receptor altered at the Tau-1 location and utilized in the co-transfection assay. The assay was also run using T47D (human breast cancer) cells. The antagonist activity assay results are shown below in Table 1. Table 2 presents comparable results for the antagonists RU-486 and (3R)-cyclocymopol monomethyl ether, and for the agonist (35)-cyclocymopol monomethyl ether. Efficacy is reported as the % maximal response observed for each compound relative to RU-486, a compound known to exhibit progesterone receptor antagonist activity. Also reported in Tables 1 and 2 for each compound is its potency or IC50 (which is the concentration (nM), required to reduce the maximal response by 50%), and its binding activity for the progesterone receptor.
Table 1
CV-1 Cells T47D Cells
Compound Efficacy Potency Efficacy Potency Binding
% % % nM Kd,nM
A 55 225 70 200 100
B 45 400 65 85 35
C 55 400 70 200 85
D 45 450 55 150 50
E 85 450 85 475 925
F 100 550 80 200 925
G 45 600 50 300 20
H 40 700 60 775 60
I 95 700 75 125 55
J 95 750 85 175 255
K 70 775 70 275 155
L 55 825 75 200 90 CV-1 Cells T47D Cells
Compound Efficacy Potency Efficacy Potency Binding
% % % nM Kd,nM
M 90 875 80 350 75
N 90 1000 70 325 195
O 90 1000 85 850 445
P 100 1200 60 275 440
Q 95 1200 65 225 245
R 95 1225 85 250 230
S 90 1500 85 400 345
T 90 1575 90 300 185
U 95 1600 80 300 190
V 95 1800 85 325 95
Table 2
CV-1 Cells T47D Cells
Efficacy Potency Efficacy Potency Binding
Compound % nM % nM Kd,nM
RU-486 100 0.1 - - 0.6
(3R)-cyclo85 965 85 385 450 cymopol monomethyl ether
(3S)-cyclo<20 >10,000 - - 325 cymopol monomethyl ether
The synthetic cyclocymopol compounds were also individually tested for cross-reactivity with the other known intracellular receptor classes. This testing showed the compounds not to have activity with the glucocorticoid receptor, in contrast to RU-486 which shows significant activity for that receptor. Some derivative compounds were found to exhibit slight activity for the androgen receptor. Pharmacological and Other Applications
It has been recognized that the co-transfection assay provides a functional assessment of the ligand being tested as either an agonist or antagonist of the specific genetic process sought to be affected, and mimics an in vivo system in the laboratory. Ligands which do not react with other intracellular receptors, as determined by the co-transfection assay, can be expected to result in fewer pharmacological side effects. Because the co-transfection assay is conducted in living cells, the evaluation of a ligand provides an early indicator of the potential toxicity of the candidate ligand at concentrations where a therapeutic benefit would be expected.
As will be discernible to those skilled in the art, the non-steroid progesterone receptor antagonist and agonist compounds disclosed can be readily utilized in pharmacological applications where progesterone receptor antagonist or agonist activity is desired, and where it is desired to minimize cross reactivities with other related intracellular receptors. In vivo applications of the invention include administration of the disclosed compounds to mammalian subjects, and in particular to humans.
The compounds of the present invention are small molecules which are relatively fat soluble or lipophilic and enter the cell by passive diffusion across the plasma membrane. Consequently, these ligands are well suited for administration orally as well as by injection. Upon administration, these ligands can selectively activate progesterone receptors and thereby modulate processes mediated by these receptors.
The pharmaceutical compositions of this invention are prepared in conventional dosage unit forms by incorporating an active compound of the invention, or a mixture of such compounds, with a nontoxic pharmaceutical carrier according to accepted procedures in a nontoxic amount sufficient to produce the desired pharmacodynamic activity in a mammalian and in particular a human subject. Preferably, the composition contains the active ingredient in an active, but nontoxic, amount selected from about 5 mg to about 500 mg of active ingredient per dosage unit. This quantity depends on the specific biological activity desired and the condition of the patient.
The pharmaceutical carrier or vehicle employed may be, for example, a solid or liquid. A variety of pharmaceutical forms can be employed. Thus, when using a solid carrier, the preparation can be plain milled micronized in oil, tableted, placed in a hard gelatin or enteric-coated capsule in micronized powder or pellet form, or in the form of a troche, lozenge, or suppository. When using a liquid carrier, the preparation can be in the form of a liquid, such as an ampule, or as an aqueous or nonaqueous liquid suspension. The following examples provide illustrative pharmacological composition formulations: Example 23
Hard gelatin capsules are prepared using the following ingredients:
Quantity (mg/capsule)
(3R)-cyclocymopol monomethyl ether 140
Starch, dried 100
Magnesium stearate 10
Total 250 mg
The above ingredients are mixed and filled into hard gelatin capsules in 250 mg quantities.
Example 24
A tablet is prepared using the ingredients below:
Quantity (mg/tablet) (3R)-cyclocymopol monomethyl ether 140
Cellulose, microcrystalline 200
Silicon dioxide, fumed 10 Stearic acid 10
Total 360 mg The components are blended and compressed to form tablets each weighing 665 mg.
Example 25
Tablets, each containing 60 mg of active ingredient, are made as follows:
Quantity
(mg/tablet)
(3R)-cyclocymopol monomethyl ether 60
Starch 45 Cellulose, microcrystalline 35
Polyvinylpyrrolidone (PVP)
(as 10% solution in water) 4
Sodium carboxymethyl starch (SCMS) 4.5
Magnesium stearate 0.5 Talc 1.0
Total 150 mg
The active ingredient, starch, and cellulose are passed through a No. 45 mesh U.S. sieve and mixed thoroughly. The solution of PVP is mixed with the resultant powders, which are then passed through a No. 14 mesh U.S. sieve. The granules so produced are dried at
50°C and passed through a No. 18 mesh U.S. sieve. The
SCMS, magnesium stearate, and talc, previously passed through a No. 60 mesh U.S. sieve, and then added to the granules which, after mixing, are compressed on a tablet machine to yield tablets each weighing 150 mg.
Example 26
Suppositories, each containing 225 mg of active ingredient, may be made as follows:
(3R)-cyclocymopol monomethyl ether 225 mg Saturated fatty acid glycerides 2,000 mg
Total 2,225 mg
The active ingredient is passed through a No. 60 mesh U.S. sieve and suspended in the saturated fatty acid glycerides previously melted using the minimum heat necessary. The mixture is then poured into a suppository mold of normal 2g capacity and allowed to cool.
Example 27
An intravenous formulation may be prepared as follows:
(3R)-cyclocymopol monomethyl ether 100 mg
Isotonic saline 1,000 ml
Glycerol 100 ml
The compound is dissolved in the glycerol and then the solution is slowly diluted with isotonic saline. The solution of the above ingredients is then administered intravenously at a rate of 1 ml per minute to a patient.
The compounds of this invention also have utility when labeled as ligands for use in assays to determine the presence of progesterone receptors. They are particularly useful due to their ability to selectively activate progesterone receptors, and can therefore be used to determine the presence of such receptors in the presence of other related receptors.
Due to the selective specificity of the compounds of this invention for progesterone receptors, these compounds can be used to purify samples of progesterone receptors in vitro . Such purification can be carried out by mixing samples containing progesterone receptors with one or more of the cyclocymopol and derivative compounds disclosed so that the compound (ligand) binds to the receptor, and then separating out the bound ligand/receptor combination by separation techniques which are known to those of skill in the art. These techniques include column separation, filtration, centrifugation, tagging and physical separation, and antibody complexing, among others.
While the preferred embodiments have been described and illustrated, various substitutions and modifications may be made thereto without departing from the scope of the invention. Accordingly, it is to be understood that the present invention has been described by way of illustration and not limitation.

Claims

Claims
1. A compound having the formulae:
Figure imgf000064_0001
or
Figure imgf000064_0002
wherein:
the dotted lines in the structure depict optional double bonds;
X is carbon, oxygen, or nitrogen;
R1 is R17, -OR17, -N(R17)(R17,), -SR17, fluorine, chlorine, bromine, or -NO2;
R17 and (R17.), each independently, are hydrogen, saturated or unsaturated C1-C6 alkyl, C3-C7 cycloalkyl, C5-C7 aryl, or C7 aralkyl, said alkyl groups being branched or straight-chain;
R2 is -NO2, -N(OH)R17, fluorine, chlorine, bromine, iodine, R17, -N(R17)(R17,), -SR17, -S(O)-R17, -S(O)2-R17, -CH2OH, -C(O)-H, -C(O)CH3, -C(O)-OCH3, -C=CH2,
-C=CH-C(O)-OCH3, or R18;
R18 and (R18,), each independently, are hydrogen, saturated or unsaturated C1-C6 alkyl, C3-C6 alkyl, C3-C7 cycloalkyl, C5-C7 aryl, or C7 aralkyl, said alkyl groups being branched or straight-chain which optionally may contain hydroxyl, aldehyde, ketone, nitrile, or ester groups;
R3 is R17 or -OR17;
R4 is hydrogen, -OR17, -OC(O)R17, -OC(O)OR17,
-OC(O)N(R17)(R17,), -OS(O)2R17, or -OS(O)-R17;
R5 is hydrogen or OR17;
R6 is R17;
R7 and R8, each independently, are R18, or R7 and R8 together are a carbocyclic 3-8 member ring;
R9 and R10, each independently, are chlorine, bromine, or R17, or R9 and R10 combined are =0, except when X=0, R9 and R10 are not present, and when X is N, then R10 is not present, or R9 and R10 together are joined in a carbocyclic 3-8 member ring;
R11 and R12, each independently, are -OR17, R18, are =0, or are =CH2, except when R11 is attached to an sp2 carbon atom in the ring, then R12 is not present and R11 is R18, or R11 and R13 together are joined in a carbocyclic 3-8 member ring or are -O- to form an epoxide;
R13 and R14, each independently, are -OR17 or R18, except when R13 is attached to an sp2 carbon atom in the ring, then R14 is not present and R13 is -OR17 or R18;
R15 and R16, each independently, are R18 or OR17, or R15 and R16 together are -CH2-O- forming an epoxide, or R15 and
R16 combined are =0 or =C(R18)(R18,), except when R15 is hydroxyl, then R16 is not hydroxyl, and when R15 is attached to an sp2 carbon atom in the ring, then R16 is not present; but excluding cyclocymopol and cyclocymopol monomethyl ether.
2. A compound as set forth in claim 1 wherein said compound exhibits activity as a progesterone receptor antagonist.
3. A compound as set forth in claim 1 wherein said compound is a diastereomerically pure 3R diastereomer.
4. A compound as set forth in claim 1 wherein said compound is a diastereomerically pure 3S diastereomer.
5. A compound selected from the group consisting of 1-methylidene-2- (2' -acetoxy-4' -bromo-5' -methoxyphenyl)methyl-3,3-dimethylcyclohexane; (35)-1-debromocyclocymopol monomethyl ether,2'-acetate; 1-methylidene-2-(2'-hydroxy-4'-bromo-5'-methoxy¬phenyl)methyl-3,3-dimethylcyclohexane; (35)-1-debromocyclocymopol monomethyl ether, 2'-methylcarbonate; (3R,5R)-5-hydroxycyclocymopol monomethyl ether; 2-(4'-nitrophenyl)methylcyclohexanone; (35)-1-debromocyclocymopolmonomethylether; 1-methylidene-6-(2'-acetoxy-4'-bromo-5'-methoxyphenyl)methyl-3,5,5-trimethylcyclohex-2-ene; 1-methylidene-6-(4'-nitrophenyl)methyl-3,5,5-trimethylcyclohex-2-ene; (3R)-1-debromocyclocymopolmonomethylether; 1-methylidene-6-(2'-hydroxy-4'-bromo-5'-methoxyphenyl)methyl-5,5-dimethylcyclohex-2-ene; 1-methylidene-6- (2 ' -acetoxy-4' -bromo-5 ' -methoxyphenyl)methyl-5,5-dimethylcyclohex-2-ene; 1-methylidene-6-(3'-methyl-4'-nitrophenyl)methyl-5,5-dimethylcyclohex-2-ene;
trans-1-methylidene-6- (2'-acetoxy-4'-bromo-5'-methoxyphenyl)methyl-4,5,5-trimethylcyclohex-2-ene; 1-methylidene-2-(4'-bromophenyl)methyl-3,3-dimethylcyclohexane; 1-methylidene-2-(2'-hydroxy-4'-bromo-5'-methoxyphenyl)methyl-3,3-dimethylcyclopentane; 1-methylidene-2-(4'-nitrophenyl)methylcyclohexane; (3R)-1-debromocyclocymopol monomethyl ether,2'-methylcarbonate; 1-methylidene-2-(2'-hydroxy-4'-bromo-5'-methoxyphenyl)methylcyclohexane; (3R)-cyclocymopol monomethyl ether,2'-methylcarbonate; (3R)-1-debromocyclocymopol monomethyl ether,2'-benzoate; and (3R)-4'-iodocyclocymopol monomethyl ether.
6. (35)-1-Debromocyclocymopol monomethyl ether,2'-acetate.
7. 1-Methylidene-6-(4'-nitrophenyl)methyl-3,5,5-trimethylcyclohex-2-ene.
8. (3R)-1-Debromocyclocymopol monomethyl ether.
9. (3R)-1-Debromocyclocymopol monomethyl ether,2'-benzoate.
10. A ligand-receptor complex formed by binding of a compound of claim 1 to a progesterone receptor.
11. A ligand-receptor complex formed by binding a compound of claim 5 to a progesterone receptor.
12. A diastereomerically pure 3R diastereomer of cyclocymopol monomethyl ether.
13. A diastereomerically purified 3R diastereomer of cyclocymopol monomethyl ether having a purity such that said purified (3R)-cyclocymopol monomethyl ether exhibits activity as a progesterone receptor antagonist despite the presence of any remaining 3S diastereomer of cyclocymopol monomethyl ether.
14. A pharmacological composition comprising a pharmaceutically acceptable vehicle and one or more compounds from the group consisting of the compounds of claim 1, cyclocymopol, and cyclocymopol monomethyl ether.
15. A pharmacological composition comprising a pharmaceutically acceptable vehicle and one or more compounds of claim 5.
16. A method of purifying a diastereomer of a compound of claim 1, cyclocymopol, or cyclocymopol monomethyl ether, comprising converting said diastereomer to an acetate and separating the diastereomeric acetate.
17- The method of claim 16 wherein the separation is carried out using HPLC.
18. A method of affecting progesterone activity comprising the in vivo administration of a compound from the group consisting of the compounds of claim 1, cyclocymopol, and cyclocymopol monomethyl ether.
19. A method for treating a mammalian subject requiring progesterone receptor antagonist therapy comprising administering to such subject a pharmaceutically effective amount of a compound of claim 1.
20. A method for treating a mammalian subject requiring progesterone receptor antagonist therapy comprising administering to such subject a pharmaceutically effective amount of the 3R diastereomer of cyclocymopol monomethyl ether or of cyclocymopol.
21. A method for treating a mammalian subject requiring progesterone receptor agonist therapy comprising administering to such subject a pharmaceutically effective amount of the 3S diastereomer of cyclocymopol monomethyl ether or of cyclocymopol.
22. A method for modulating a process mediated by progesterone receptors comprising causing said process to be conducted in the presence of at least one compound as set forth in claim 1.
23. A method for modulating a process mediated by progesterone receptors comprising causing said process to be conducted in the presence of at least one compound from the group consisting of cyclocymopol and cyclocymopol monomethyl ether.
24. A method for modulating a process mediated by progesterone receptors comprising administering to a mammalian subject an amount, effective to moderate said process mediated by said progesterone receptors, of a compound of claim 1.
25. A method for modulating a process mediated by progesterone receptors comprising administering to a mammalian subject an amount, effective to moderate said process mediated by said progesterone receptors, of at least one compound from the group consisting of cyclocymopol and cyclocymopol monomethyl ether.
26. A method for determining the presence of one or more progesterone receptors comprising combining at least one compound from the group consisting of the compounds of claim 1, cyclocymopol, and cyclocymopol monomethyl ether with a sample containing one or more unknown receptors and determining whether said compound binds to any receptor in said sample.
27. A method of purifying progesterone receptors comprising combining at least one compound from the group consisting of the compounds of claim 1, cyclocymopol, and cyclocymopol monomethyl ether with a sample containing progesterone receptors, allowing said compound to bind said progesterone receptors, and separating out the bound combination of said compound and said progesterone receptors.
PCT/US1993/003909 1992-04-21 1993-04-21 Non-steroid progesterone receptor agonist and antagonist compounds and methods WO1993021145A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP93910811A EP0637296A4 (en) 1992-04-21 1993-04-21 Non-steroid progesterone receptor agonist and antagonist compounds and methods.

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US87271092A 1992-04-21 1992-04-21
US07/872,710 1992-04-21
US4864693A 1993-04-16 1993-04-16
US08/048,646 1993-04-16

Publications (1)

Publication Number Publication Date
WO1993021145A1 true WO1993021145A1 (en) 1993-10-28

Family

ID=26726365

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1993/003909 WO1993021145A1 (en) 1992-04-21 1993-04-21 Non-steroid progesterone receptor agonist and antagonist compounds and methods

Country Status (5)

Country Link
EP (1) EP0637296A4 (en)
AU (1) AU4117093A (en)
CA (1) CA2133325A1 (en)
MX (1) MX9302312A (en)
WO (1) WO1993021145A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995011215A1 (en) * 1993-10-21 1995-04-27 Ligand Pharmaceuticals Incorporated Non-steroid androgen receptor antagonists
LT4291B (en) 1994-12-23 1998-02-25 Schering Aktiengesellschaft Compounds with progesterone-antagonistic and anti-oestrogen properties intended for combined use in female contraception
US11198699B2 (en) 2019-04-02 2021-12-14 Aligos Therapeutics, Inc. Compounds targeting PRMT5

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR1562130A (en) * 1967-02-10 1969-04-04
US4778621A (en) * 1985-10-14 1988-10-18 Chisso Corporation Ester compound
US4981784A (en) * 1987-12-02 1991-01-01 The Salk Institute For Biological Studies Retinoic acid receptor method
US5017610A (en) * 1988-06-13 1991-05-21 Ono Pharmaceutical Co., Ltd. Derivatives of p-substituted phenyl ester of pivalic acid
US5071773A (en) * 1986-10-24 1991-12-10 The Salk Institute For Biological Studies Hormone receptor-related bioassays
US5128479A (en) * 1989-02-10 1992-07-07 Basf Aktiengesellschaft Oxidized diphenylheteroalkanes
US5135940A (en) * 1982-09-23 1992-08-04 Merck Frosst Canada, Inc. Leukotriene antagonists

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5134155A (en) * 1991-08-08 1992-07-28 Ortho Pharmaceutical Corporation Tetrahydroindazole, tetrahydrocyclopentapyrazole, and hexahydrocycloheptapyrazole compounds and their use as HMG-coA reductase inhibitors

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR1562130A (en) * 1967-02-10 1969-04-04
US5135940A (en) * 1982-09-23 1992-08-04 Merck Frosst Canada, Inc. Leukotriene antagonists
US4778621A (en) * 1985-10-14 1988-10-18 Chisso Corporation Ester compound
US5071773A (en) * 1986-10-24 1991-12-10 The Salk Institute For Biological Studies Hormone receptor-related bioassays
US4981784A (en) * 1987-12-02 1991-01-01 The Salk Institute For Biological Studies Retinoic acid receptor method
US5017610A (en) * 1988-06-13 1991-05-21 Ono Pharmaceutical Co., Ltd. Derivatives of p-substituted phenyl ester of pivalic acid
US5128479A (en) * 1989-02-10 1992-07-07 Basf Aktiengesellschaft Oxidized diphenylheteroalkanes

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
Agric. Biol. Chem., Vol. 54, No. 1, pp. 121-123, (1990), A. TANAKA et al., "Synthesis of ( ) Cyclocymopol". *
CHEMICAL ABSTRACTS, Vol. 106, No. 7, (1986), No. 49640e, (Oxford, U.K., "Direct 4-Alkylation of 1,3-Cyclohexamediones......", Synthesis, (6), 476-80. *
CHEMICAL ABSTRACTS, Vol. 117, No. 19, (1989), No. 19210v, (Calcutta, India, U.R., GHATAK et al., "An Efficient Synthesis of 2-Substituted 3,3-Dimethylcyclohexan-1 Ones: A Simple..........Diterpenoids", Proc.-INT. CONGR. ESSENT. OILS, FRAGRANCES Flavors, 11th Volumne, New Delhi, India). *
CHEMICAL ABSTRACTS, Vol. 118, No. 11, (1992), No. 101626u, (Calcutta, India, S. PAL et al., "A Facile Synthetic Route to 1, 1-Disubstituted......", Synthesis, (11), 1073-5. *
CHEMICAL ABSTRACTS, Vol. 118, No. 13, (1986), No. 115810Jj, (Calcutta, India, B.K. BANIK et al., "Efficient Synthesis of 2-Substituted 3,3-Dimethylcyclohexamones.....", J. Chem. RES., SYNOP (11), 406-7. *
CHEMICAL ABSTRACTS, Vol. 73, No. 8, 4 Apr 1969 (04.04.69) (France, F. BENGUEREL et al., "Anthraquinone Dyes", Sandoz Ltd.; & FR,A,1562130 (1969). *
Jrnl. Chem. Soc. Perkin, Trans. I, pp. 1696-1701, (1976), H. HOGBERG et al., "The Cymopols ..... Cymopolia Barbata". *
Jrnl. of Natural Products, Vol. 52, No. 5, pp. 1092-1099, Sept-Oct. 1989, M.E. WALL et al., "Plant Antimutagenic Agents......... Alga (Cymopolia Barbata)". *
Phyto Chemistry, Vol. 21, No. 8, pp. 2139-2141, (1982), O.J. McCONNELL et al., "Diastereoisomers of Cyclocymopol and ......". *
Science, Vol. 240, pp 889-895, (13 May 1988), R. EVANS, "The Steroid and Thyroid Hormone Receptor Super Family". *
See also references of EP0637296A4 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995011215A1 (en) * 1993-10-21 1995-04-27 Ligand Pharmaceuticals Incorporated Non-steroid androgen receptor antagonists
US5677336A (en) * 1993-10-21 1997-10-14 Ligand Pharmaceuticals Incorporated Non-steroid androgen receptor antagonist compounds and methods
LT4291B (en) 1994-12-23 1998-02-25 Schering Aktiengesellschaft Compounds with progesterone-antagonistic and anti-oestrogen properties intended for combined use in female contraception
US11198699B2 (en) 2019-04-02 2021-12-14 Aligos Therapeutics, Inc. Compounds targeting PRMT5

Also Published As

Publication number Publication date
MX9302312A (en) 1994-06-30
AU4117093A (en) 1993-11-18
EP0637296A1 (en) 1995-02-08
EP0637296A4 (en) 1996-02-07
CA2133325A1 (en) 1993-10-28

Similar Documents

Publication Publication Date Title
US5808139A (en) Non-steroid progesterone receptor agonist and antagonist and compounds and methods
JP3278159B2 (en) (3R, 4R) -Δ ▲ 6-tetrahydrocannabinol-7-acid
US5677336A (en) Non-steroid androgen receptor antagonist compounds and methods
JP2758902B2 (en) Alkoxy-substituted dihydrobenzopyran-2-carboxylate derivatives
US5856535A (en) Aminosterol ester compounds
Fernández-Herrera et al. Probing the selective antitumor activity of 22-oxo-26-selenocyanocholestane derivatives
JPH10502373A (en) Stereochemical wortmannin derivatives
IL185867A (en) Substituted benzopyrans as selective estrogen receptor-beta agonists
CN102796161A (en) Estratriene derivatives and their uses as 17beta-hydr0xyster0id dehydrogenase inhibitors
US5321044A (en) 15,16-seco-19-nor progestins
Revankar et al. A selective ligand for estrogen receptor proteins discriminates rapid and genomic signaling
CN100374444C (en) Substituted benzopyrans as selective estrogen receptor-beta agonists
Zhao et al. Design, synthesis, and estrogenic activity of a novel estrogen receptor modulator a hybrid structure of 17β-estradiol and vitamin E in hippocampal neurons
Dell’Agli et al. Inhibition of NF-kB and metalloproteinase-9 expression and secretion by parthenolide derivatives
JP2002510667A (en) New retinoids and their use
EP2682386B1 (en) Novel vitamin d receptor modulator with partial agonist activity
JPH10505348A (en) Methods for identifying and treating resistant tumors
WO1993021145A1 (en) Non-steroid progesterone receptor agonist and antagonist compounds and methods
JP2001502708A (en) Novel cis-3,4-chroman derivative useful for prevention or treatment of estrogen-related diseases or syndromes
CN112915096B (en) Pharmaceutical application of echinocystic acid-28-O-beta-D-glucoside
Vila et al. Synthesis and biological studies of “Polycerasoidol” and “trans-δ-Tocotrienolic acid” derivatives as PPARα and/or PPARγ agonists
US5994390A (en) Trans-3,4-chroman derivatives useful in the prevention or treatment of estrogen related diseases or syndromes
CN117281816A (en) Asymmetric synthesis and use of compounds in the treatment of disease
WO1994024080A1 (en) Progesterone receptor agonist and antagonists
CA2501833A1 (en) 5-(1&#39;,1&#39;-cycloalkyl/alkenyl)methylidene 1,2-dihydro-5h-chromeno[3,4-f]quinolines as selective progesterone receptor modulator compounds

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU BB BG BR CA CZ FI HU JP KP KR LK MG MN MW NO NZ PL PT RO RU SD SK UA

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES 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)
WWE Wipo information: entry into national phase

Ref document number: 2133325

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1993910811

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1993910811

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1993910811

Country of ref document: EP