WO1994024080A1 - Antagonistes et agonistes de recepteur de progesterone - Google Patents

Antagonistes et agonistes de recepteur de progesterone Download PDF

Info

Publication number
WO1994024080A1
WO1994024080A1 PCT/US1993/010086 US9310086W WO9424080A1 WO 1994024080 A1 WO1994024080 A1 WO 1994024080A1 US 9310086 W US9310086 W US 9310086W WO 9424080 A1 WO9424080 A1 WO 9424080A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
methyl
methylidene
bromo
methoxyphenyl
Prior art date
Application number
PCT/US1993/010086
Other languages
English (en)
Inventor
I. Charles Pathirana
Tina S. Berger
Robert S. Stein
William Fenical
Todd K. Jones
Lawrence G. Hamann
Luc Farmer
Michael G. Johnson
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 AU56641/94A priority Critical patent/AU5664194A/en
Publication of WO1994024080A1 publication Critical patent/WO1994024080A1/fr

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
    • C07C205/13Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by hydroxy groups
    • C07C205/18Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by hydroxy groups having nitro groups or hydroxy groups bound to carbon atoms of rings other than 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
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/01Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and halogen atoms, or nitro or nitroso groups bound to the same carbon skeleton
    • C07C323/09Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and halogen atoms, or nitro or nitroso groups bound to the same carbon skeleton having sulfur atoms of thio groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton
    • 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 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).
  • HREs hormone response elements
  • ligands 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.”
  • 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:
  • McConnell et al. Through silica gel chromatography of an ether-soluble extract of C. barbata, McConnell et al. were able to obtain a 1 : 1 mixture of ⁇ : ⁇ epimers of cyclocymopol. McConnell et al. also obtained a 3:1 mixture of ⁇ : ⁇ epimers of cyclocymopol monomethyl ether, which was enriched to a 4:1 mixture of the ⁇ : ⁇ 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.
  • the forms of cyclocympol and cyclocymopol monomethyl ether obtained by Högberg et al., supra, were pure forms of formulae 1b 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.
  • 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
  • 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
  • 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.
  • 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.
  • 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 (3 R)-cyclocymopol monomethyl ether (compound SO-53). For this compotmd 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 (3S)-cyclocymopol monomethyl ether (compotmd SO-54) and its acetate (compound SO-51).
  • 3S 3S-cyclocymopol monomethyl ether
  • compound SO-51 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 (3 R)-cyclocymopol monomethyl ether (compound SO-9).
  • compound SO-9 progesterone receptor activation by (3 R)-cyclocymopol monomethyl ether
  • Figure 6 presents activation profiles for analysis of glucocorticoid receptor activation by (3R)-cyclocymopol monomethyl ether (compound SO-09).
  • agonist dose response is shown in panel a and 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 RU486 and cyclocymopol monomethyl ether diastereomers.
  • Inhibition by (3 R)-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; and
  • Figure 11 presents profiles showing the inhibition by RU486 of induction of alkaline phosphatase in T47D cells by (3S)-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
  • Y is oxygen, nitrogen, sulfur or a saturated or unsaturated C 1 -C 4 alkyl, optionally substituted with oxygen, nitrogen or sulfur;
  • R 1 is R 17 , -OR 17 ,-N(R 17 )(R 17' ), -SR I7 , fluorine, chlorine, bromine, or -NO 2 ;
  • R 17 and (R 17' ), each independently, are hydrogen, a 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 18 and (R 18' ), each independently are hydrogen, a saturated or unsaturated
  • alkyl groups being branched or straight-chain which optionally may contain hydroxyl, aldehyde, ketbne, nitrile, or ester groups;
  • R 3 is R 17 or -OR I7 , or R 2 and R 3 taken together can form a saturated or unsaturated heterocyclic 3-8 member ring substituted with one or more heteroatoms selected from the group consisting of oxygen, nitrogen, and sulfur, provided, however, that when R 2 and R 3 form such a saturated or unsaturated heterocyclic 3-8 member ring, then the bond between the carbon atoms carrying the R 2 and R 3 substituents can be either a single bond or a double bond;
  • 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 , or OR 17 ;
  • R 7 and R 8 are hydrogen, R 18 , or R 7 and R 8 together are a carbocyclic 3-8 member ring;
  • R 11 and R 13 together are joined in a carbocyclic 3-8 member ring or are -O- to form an epoxide
  • 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 ;
  • Representative compounds and derivatives according to the present invention include the following:
  • 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 marine alga Cymopolia barbata (L.) Lamouroux (Dasycladaceae) was collected and frozen. Frozen sample was lyophilized and extracted with 1 : 1 MeOH/CH 2 Cl 2 three times, and the extract was concentrated in vacuo to obtain an aqueous suspension of organic components. The concentrate was re-extracted with CH 2 Cl 2 until no color came into the organic phase, and the CH 2 Cl 2 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/CH 2 Cl 2 , or vacuum flash chromatography on silica using a gradient of ethyl acetate in hexane, and the fractions were examined by thin layer chromatography (TLC).
  • TLC thin layer chromatography
  • the fractions that contained cymopols were pooled together and separated by reversed phase high performance liquid chromatography (HPLC) using 80% MeOH/H 2 O to yield a mixture of
  • 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 transfected 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
  • 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.
  • luciferase When that gene is one that expresses, for example, luciferase, luciferase is produced which catalyzes a light-emitting reaction with its substrate luciferin.
  • luciferase 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.
  • 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.
  • control hormone progesterone
  • agonist analogues cyclocymopol compounds
  • 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).
  • antagonist dose response is shown in panel b.
  • 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% 0 2 /5% CO 2 for 40 hours prior to harvest.
  • radiolabeled progesterone-agonist 3 H-R5020 compound SO-9 (which is identical to compound SO-53), and the progesterone antagonist, RU486.
  • 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 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.
  • (3s)-cyclocymopol monomethyl ether (compound SO-54), and its corresponding acetate (compound SO-51), are functional agonists in this system
  • (3R)-cyclocymopol monomethyl ether (compotmd SO-53), and its corresponding acetate (compound SO-52)
  • the 3S form compounds exhibit increased efficacy at higher concentrations, an effect which has been found to be reproducible.
  • (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 3S-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.
  • reaction mixture was then poured into a separatory funnel containing 50 mL dichloromethane and 50 mL saturated aqueous N ⁇ 4 Cl, the layers were separated, and the organic phase was washed with 50 mL brine, dried over Na 2 SO 4 , 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.
  • 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 trans-ferred 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
  • 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
  • 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 4,4-dimethylcyclopent-2-en-1-one (94) and benzylic bromide (6) in 5 steps in the manner previously described for the synthesis of olefin (13), with the following procedural changes in the last two steps, from intermediate (95).
  • 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 acetoxy-diene as a colorless, oily solid.
  • 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.
  • 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
  • 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 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.
  • 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.
  • This compound was prepared from 5,5-dimethylcyclohex-2-en-1-one (8) and p-bromobenzyl bromide in three steps in the manner described for the synthesis of olefin (13).
  • 1 H NMR 400 MHz, CDCl 3 ) ⁇ 0.94 and 0.97 (2s, 2 x 3H, geminal-CH 3 's), 2.62 and 2.82 (d of ABq, 2H, benzylic-CH 2 ), 4.25 and 4.61 (2s, 2 x 1 H, methylidene-CH 2 ), 6.98 (d, 2H, Ar-H), 7.34 ppm (d, 2H, Ar-H).
  • This compound is also referred to as Compound "O" or 120130]
  • 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 nitro-diene as a colorless oil.
  • TTis 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 (3S)-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 compotmd were identical to those reported for the racemic analog 13. [This compound is also referred to as Compound "G" or 120058]
  • This compound was prepared from (35)-1-debromocyclocymopol monomethyl ether (28) in the manner previously described for the synthesis of acetate (84).
  • the TLC elution properties and 1 H NMR spectral data for this compound are identical to those reported for the racemic analog (84).
  • This compound is also referred to as Compound "B" or 120093]
  • 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. Purification by radial
  • 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.
  • reaction mixture was then poured into a 250 mL erlynmeyer flask containing 50 mL ice-cold 1.0 M aqueous NaHSO 4 , and the resultant biphasic mixture was stirred 5 min before extraction with ethyl acetate (3 ⁇ 50 mL).
  • the combined organic solutions were washed with 50 mL brine, dried over Na 2 SO 4 , and concentrated under diminished pressure. Purification by flash column chromatography (silica gel, hexanes / ethyl acetate, 5:1) afforded 1.57 g (85%) of the desired allylic alcohol as a white solid.
  • Hexanes 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 hexanes 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.
  • This compound was prepared from isophorone (2.065 g, 14.94 mmol) and/Miitrobenzyl bromide (4.06 g, 18.80 mmol, 1.25 equiv) in the manner previously described for the synthesis of enone (11), affording 1.891 g (46%), of the nitro-enone as a pale yellow oil.
  • reaction mixture was then allowed to warm to room temperature, and stirred 45 min before cooling to 0 °C, and the addition of 0.5 mL of 1 N NaOH.
  • the reaction mixture was then diluted with hexanes (50 mL), and filtered through a pad of celite and silica gel. The organic solution was dried over Na 2 SO 4 , and concentrated under diminished pressure. Purification by flash column
  • isophorone (329 mL, 2.19 mmol) was added dropwise as a solution in 2 mL THF, and the reaction mixture was allowed to gradually warm to 0 °C over 90 min before the addition of 4-nitrophenyldisulfide (1.19 g, 3.29 mmol, 1.50 equiv) as a solution in 5 mL THF.
  • 4-nitrophenyldisulfide (1.19 g, 3.29 mmol, 1.50 equiv) as a solution in 5 mL THF.
  • the resultant dark-orange reaction solution was allowed to warm to room temperature, stirring 15 h before cooling to 0 °C, and quenching with 10 mL saturated aqueous ⁇ H 4 Cl.
  • reaction mixture was quenched at 0 °C with saturated aqueous NH 4 Cl, and the reaction mixture was extracted with ethyl acetate (50 mL). The organic phase was washed with brine (20 mL), dried over Na 2 SO 4 , and concentrated under diminished pressure. Purification by flash column chromatography (silica gel, hexanes / ethyl acetate, gradient elution) afforded 20 mg (96%) of the spirocyclopropane as a colorless, viscous oil.
  • This compound was prepared from 4-[2'-(tert-butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]methyl-3,5,5-trimethylcyclohex-2-en-1-one (55) (100 mg, 0.214 mmol) in the manner previously described for the synthesis of olefin (13), affording 43 mg (57%) of the desired diene as a colorless oil.
  • This compound was prepared from isophorone (0.40 g, 2.88 mmol) and 3-[2'-(tert-butyl)dimethylsilyloxy-4'-bromo-5'-methoxyphenyl]prop-2-enyl bromide (43) (0.50 g, 1.15 mmol) in the manner previously described for the synthesis of enone (11), affording 452 mg (80%) of the desired enone as a yellow oil.
  • This compound was prepared from 6-[trans -(2'-hydroxy-4'-bromo-5'-methoxypheny1)-1-propenyl]-3,5,5-trimethylcyclohex-2-en-1-one (59) (50 mg, 0.10 mmol) in the manner previously described for the synthesis of olefin (13), affording 23.0 mg (60%) of the desired triene as a colorless oil.
  • Lithium tris[(3-ethyl-3-pentyl)-oxy]aluminohydride (LiTEPA) (0.90 mL of a 0.5 M solution in THF, 0.45 mmol, 1.1 equiv) was then added at a steady rate over 40 min using a syringe pump.
  • the reaction mixture was allowed to stir at -78 °C for an additional 2 h before the addition of 10 mL 1 :1 methanol / water.
  • the mixture was extracted with ethyl acetate (3 ⁇ 30 mL), and the combined organic layers were dried over Na 2 SO 4 , and concentrated under diminished pressure. Purification by flash column
  • This compound was prepared from trans -4-(2-hydroxy)ethyl-6-(2'-hydroxy-4'-bromo-5'-methoxyphenyl)methyl-3,5,5-trimethylcyclohex-2-en-1-one (62) (50 mg, 0.098 mmol) in the manner previously described for the synthesis of olefin (13), using 5.0 equiv of the (trimethyl)silylmethyllithium reagent, affording 12 mg (26%) of the desired diene as a colorless, oily solid.
  • ketosulfoxide (68) prepared from (R)-(+)-pulegone according to the method of Oppolzer and Petrzika; Helv. Chim. Acta 1978, 61, 2755) in 5 mL of dry THF.
  • reaction mixture was stirred at - 35 oC for 3 h, after which 2-(tert-butyl)dimethylsilyloxy-4-bromo-5-methoxybenzyl bromide (6) was added dropwise as a solution in 10 mL of anhydrous THF.
  • the reaction mixture was allowed to stir for an additional 2 h at -35 oC, quenched with 1 M aqueous NaHSO 4 (15 mL) and extracted with ether
  • the resultant oil was dissolved in 2 mL of dry dichloromethane and 162 mL of pyridine, and to this a solution of hydrogen peroxide 35% (181 mL) in 162 mL of water was added dropwise, keeping the temperature between 30-35 oC and warming if necessary to initiate the reaction.
  • the mixture was stirred at room temperature for 30 minutes, and then poured into a separatory funnel containing dichloromethane-saturated aqueous NaHCO 3 . After extraction of the mixture with dichloromethane, the organic solution was washed successively with 10% aqueous HC1 and brine, and dried over Na 2 SO 4 . The solvent was removed under diminished pressure, and the residue was purified by flash column
  • This intermediate alcohol (14 mg, 0.028 mmol) was dissolved in 1 mL of anhydrous THF containing 60 mL (0.56 mmol, 20.0 equiv) of acetic anhydride, and cooled to 0 oC.
  • Tetra-n-butylammonium fluoride (33 mL of a 1.0 M solution in THF, 0.033 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 5 mL of ethyl acetate and 2 mL of aqueous 1 M NaHSO 4 .
  • the layers were separated, the organic phase was dried over Na 2 SO 4 , and the solvent was removed under diminished pressure.
  • the crude intermediate (12.4 mg) was placed in a 10 mL Nalgene vial containing 2 mL of dry THF, 0.175 mL of premade HF / pyridine complex was added, and the mixture was allowed to stir at room temperature for 18 h. The contents of the vial were then transferred into a separatory funnel containing 5 mL of ethyl acetate and 3 mL of aqueous 1 M NaHSO 4 . The layers were separated, and the organic phase was washed with 2 mL of brine, dried over Na 2 SO 4 , and concentrated under diminished pressure.
  • reaction mixture was stirred at -50 oC for an additional 30 minutes, and then siphoned into cold, vigorously stirred aqueous 2 N HCl. In this procedure it was important to keep the reaction temperature below -40 oC.
  • the resulting mixture was extracted with ether, and the organic extract was washed with saturated aqueous NaHCO 3 , dried over Na 2 SO 4 , and concentrated under diminished pressure.
  • the mixture was allowed to stir at room temperature for 18 h, at which time TLC analysis indicated complete consumption of starting material, and the formation of a less polar product.
  • the reaction mixture was then poured into a separatory funnel containing 5 mL of dichloromethane and 5 mL saturated aqueous NH 4 CI. The layers were separated, and the organic phase was washed with 5 mL of brine, dried over Na 2 SO 4 . and concentrated under diminished pressure.
  • This exo-enone was prepared in two steps from (3R)-1-debromocyclocymopol monomethyl ether, (tert-butyl)dimethylsilyl ether (78) (0.750 g, 1.66 mmol).
  • the first step oxidation with selenium (IV) oxide was carried out in the manner previously described for the synthesis of
  • reaction mixture was then transferred to a 125 mL erlynmeyer flask containing 50 mL 1:1 saturated aqueous NaHCO 3 / 10% Na 2 S 2 O 3 , and the mixture was stirred for an additional 90 min.
  • the mixture was then extracted with dichloromethane (2 ⁇ 40 mL), and the organic phase was washed with brine, dried over Na 2 SO 4 , and concentrated under diminished pressure. Purification by flash column chromatography (silica gel, hexanes / ethyl acetate, 9:1) afforded 289 mg (85%) of the desired enone as a white 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.
  • a successful pregnancy requires not only the effective collision and fusion of egg and spermatozoon, but also the provision of a receptive and supportive uterus.
  • the preparation of the endometrium of the uterus for implantation i.e. decidualization
  • decidualization starts well before the blastocyst arrives in the uterus. Since decidualization of the rodent uterus is a classic response to progesterone action, a progesterone antagonist should inhibit or interrupt this process when given in vivo.
  • Psuedopregnant mice i.e. mated with vasectomized males
  • the key element of this assay model targets the manipulation of the endocrine status of the mice during the early days of pseudopregnancy by perturbing the action of endogenous progesterone, and thereby blocking the decidual cell response. This was acheived by intraluminal administration of various known progesterone antagonists and compounds of the present invention between Day 2 and Day 4 of
  • 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.
  • 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.
  • Hard gelatin capsules are prepared using the following
  • 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 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.
  • 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 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.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Toxicology (AREA)
  • Cell Biology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Immunology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Endocrinology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Composés non stéroïdiens constituant des ligands de haute affinité et de haute spécificité par rapport à des récepteurs de progestérone. Ces composés comprennent des dérivés synthétiques de cyclocymopol et de ses diastéréomères, du monométhyle éther de (3R)-cyclocymopol de pureté spectroscopique et chromatographique, qui fonctionne comme antagoniste de récepteur de progestérone, et du monométhyle éther de (3S)-cyclocymopol de pureté spectroscopique et chromatographique qui fonctionne comme agoniste de récepteur de progestérone. Des procédés d'utilisation de ces composés sont également décrits, lesquels permettent de moduler des processus induits par des récepteurs de progestérone et de traiter des patients requérant une thérapie par antagoniste ou agoniste de récepteur de progestérone.
PCT/US1993/010086 1993-04-16 1993-10-21 Antagonistes et agonistes de recepteur de progesterone WO1994024080A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU56641/94A AU5664194A (en) 1993-04-16 1993-10-21 Progesterone receptor agonist and antagonists

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US4864693A 1993-04-16 1993-04-16
US08/048,646 1993-04-16

Publications (1)

Publication Number Publication Date
WO1994024080A1 true WO1994024080A1 (fr) 1994-10-27

Family

ID=21955675

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1993/010086 WO1994024080A1 (fr) 1993-04-16 1993-10-21 Antagonistes et agonistes de recepteur de progesterone

Country Status (2)

Country Link
AU (1) AU5664194A (fr)
WO (1) WO1994024080A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6440954B1 (en) 1997-04-18 2002-08-27 President And Fellows Of Harvard College Inhibition of vascular smooth muscle cell proliferation

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4780420A (en) * 1985-09-30 1988-10-25 Yale University Progesterone receptor ligands
EP0474301A2 (fr) * 1990-09-03 1992-03-11 Shell Internationale Researchmaatschappij B.V. Dérivés de la cyclohexanone

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4780420A (en) * 1985-09-30 1988-10-25 Yale University Progesterone receptor ligands
EP0474301A2 (fr) * 1990-09-03 1992-03-11 Shell Internationale Researchmaatschappij B.V. Dérivés de la cyclohexanone

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
A TANAKA ET AL: "SYNTHESIS OF(+-)-CYCLOCYMOPOL", AGRICULTURAL AND BIOLOGICAL CHEMISTRY, vol. 54, no. 1, 1990, TOKYO JP, pages 121 - 123 *
B DAVIS ET AL: "DITERPENE SYNTHESIS", JOURNAL OF THE CHEMICAL SOCIETY, PERKIN TRANSACTIONS 1, 1978, LETCHWORTH GB, pages 2840 - 2844 *
D KAMINSKY ET AL: "FISCHER INDOLE LIKE SYNTHESIS", TETRAHEDRON LETTERS, no. 10, 1967, OXFORD GB, pages 859 - 861 *
L HARWOOD ET AL: "DIRECT 4-ALKYLATION OF 1,3-CYCLOHEXANEDIONES VIA DIANIONIC SPECIES", SYNTHESIS, no. 6, 1986, STUTTGART DE, pages 476 - 480 *
M WALL ET AL: "PLANT ANTIMUTAGENIC AGENTS 7.", JOURNAL OF NATURAL PRODUCTS, vol. 52, no. 5, 1989, COLUMBUS ,OHIO,US, pages 1092 - 1099 *
O MCCONNELL ET AL: "DIASTEREOISOMERS OFCYCLOCYMOPOL AND CYCLOCYMOPOL MONOMETHYL ETHER FROM CYMOPOLIA BARBATA", PHYTOCHEMISTRY, vol. 21, no. 8, 1982, OXFORD,GB, pages 2139 - 2141 *
R THOMSON ET AL: "THE CYMOPOLS,A GROUP OF PRENYLARED BROMOHYDROQUINONES FROM THE GREEN CALCAREOUS ALGA CYMOPOLIA BARBATA", JOURNAL OF THE CHEMICAL SOCIETY, PERKIN TRANSACTIONS 1, no. 16, 1976, LETCHWORTH GB, pages 1696 - 1701 *
S AOKI ET AL: "PALLADIUM-CATALYZED ARYLATION OF SILYLOXYCYCLOPROPANES WITH ARYL TRIFLATES", JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, vol. 110, no. 10, 1988, WASHINGTON, DC US, pages 3296 - 3298 *
U GHATAK ET AL: "A FACILE SYNTHETIC ROUTE TO 1,1-DISUBSTITUTED 2,3-DIHYDRO-1H-BENZ(f) INDENE-4,9-DIONES", SYNTHESIS, no. 11, 1992, STUTTGART DE, pages 1073 - 1075 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6440954B1 (en) 1997-04-18 2002-08-27 President And Fellows Of Harvard College Inhibition of vascular smooth muscle cell proliferation

Also Published As

Publication number Publication date
AU5664194A (en) 1994-11-08

Similar Documents

Publication Publication Date Title
US5808139A (en) Non-steroid progesterone receptor agonist and antagonist and compounds and methods
US5677336A (en) Non-steroid androgen receptor antagonist compounds and methods
Manolov et al. Synthesis, structure, toxicological and pharmacological investigations of 4-hydroxycoumarin derivatives
JP5410314B2 (ja) レソルビン:治療処置用のバイオテンプレート
JP3280975B2 (ja) 20−メチル−置換ビタミンd−誘導体
JP3278159B2 (ja) (3r,4r)−δ▲上6▼−テトラヒドロカンナビノール−7−酸
TW200831109A (en) Progesterone receptor antagonists
FR2485543A1 (fr) Alcynyl-10 steroides utiles comme medicaments, procede et intermediaires pour leur preparation et compositions therapeutiques et formes pharmaceutiques contenant ces medicaments
JPS60502100A (ja) 黄体活性を示すステロイド
JP2000256390A (ja) 性ステロイド活性を抑制するために用いるエストロゲン核誘導体の医薬組成物
US5321044A (en) 15,16-seco-19-nor progestins
FR2485544A1 (fr) (propadiene-1,2 yl)-10 steroides inhibiteurs irreversibles des aromatases, utiles notamment comme medicaments inhibiteurs de la production d'oestrogenes, compositions therapeutiques et formes pharmaceutiques les contenant et intermediaires pour la preparation de ces steroides
JP2643943B2 (ja) 新規な6−もしくは7−メチレンアンドロスタ−1,4−ジエン−3,17−ジオン誘導体およびその製造方法
JP2006524240A (ja) 選択的エストロゲン受容体β作用薬である置換ベンゾピラン
JP2512439B2 (ja) 新規ステロイドおよび医薬
US3478106A (en) Preparation of 13-alkylgona-1,3,5(10),8,14-pentaenes
WO1994024080A1 (fr) Antagonistes et agonistes de recepteur de progesterone
WO1993021145A1 (fr) Composes non steroidiens a activite agoniste et antagoniste par rapport au recepteur de progesterone, et procedes d'utilisation
JPH07507807A (ja) 抗高コレステロール血症化合物及び関連医薬組成物並びに使用方法
US6900191B1 (en) 1α-Hydroxyvitamin D5, its synthesis and use in cancer prevention
Wall Recent advances in the chemistry and metabolism of the cannabinoids
JPH05132499A (ja) ビタミンd関連化合物
GB2212803A (en) Intermediates for seco-sterol compounds
AU716389B2 (en) Cytostatic sterols
JPH0625155A (ja) 22−メチルビタミンd誘導体

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 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)
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA