WO2014203129A1 - Combinations of benzopyran compounds, compositions and uses thereof - Google Patents

Combinations of benzopyran compounds, compositions and uses thereof Download PDF

Info

Publication number
WO2014203129A1
WO2014203129A1 PCT/IB2014/062176 IB2014062176W WO2014203129A1 WO 2014203129 A1 WO2014203129 A1 WO 2014203129A1 IB 2014062176 W IB2014062176 W IB 2014062176W WO 2014203129 A1 WO2014203129 A1 WO 2014203129A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
unsubstituted
inhibitor
combination
pharmaceutically acceptable
Prior art date
Application number
PCT/IB2014/062176
Other languages
French (fr)
Inventor
Peter J. Kushner
Leslie Hodges GALLAGHER
Cyrus L. HARMON
David C. Myles
Original Assignee
Olema Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Olema Pharmaceuticals, Inc. filed Critical Olema Pharmaceuticals, Inc.
Publication of WO2014203129A1 publication Critical patent/WO2014203129A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4025Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil not condensed and containing further heterocyclic rings, e.g. cromakalim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/513Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim having oxo groups directly attached to the heterocyclic ring, e.g. cytosine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones
    • A61P5/30Oestrogens

Definitions

  • This invention is in the field of pharmaceuticals, and in particular, this invention is related to the treatment of cancers mediated by the estrogen receptor by administering a specific benzopyran (in the form of a mixture of S-C2 and R-C2 diastereomers or its pure S-diastereomer) as described in US 2013/0178445 and WO 2013/090921 in combination with particular agents, active agents, anti-cancer agents or chemotherapeutic agents.
  • a specific benzopyran in the form of a mixture of S-C2 and R-C2 diastereomers or its pure S-diastereomer
  • Estrogen receptor modulators are a class of compounds that act on the estrogen receptor. These compounds can be pure agonists (mimicking estrogen), pure antagonists, or mixed agonist-antagonists (sometimes referred to as Selective Estrogen Receptor Modulators (SERMs)). For example, estradiol (A) is a pure agonist, fulvestrant (B) is a complete antagonist, and tamoxifen (C) and raloxifene (D) are SERMs.
  • ER estrogen receptors
  • Partial anti-estrogens like raloxifene and tamoxifen retain some estrogen-like effects, including an estrogen-like stimulation of uterine growth, and also, in some cases, an estrogen-like action during breast cancer progression which actually stimulates tumor growth.
  • fulvestrant a complete anti-estrogen, is free of estrogen-like action on the uterus and is effective in tamoxifen-resistant tumors.
  • fulvestrant is substantially superior to the aromatase inhibitor anastrozole in treating metastatic breast cancer (Robertson et al. J Clin Oncol (2009) 27(27):4530-5).
  • Estradiol is a naturally-occuring female estrogenic hormone.
  • Raloxifene was disclosed by Eli Lilly in 1981 (U.S. Patent No. 4,418,068; 5,478,847; 5,393,763; and 5,457,117) for prevention of breast cancer and treatment of osteoporosis. Fulvestrant was disclosed by Imperial Chemical Industries (ICI) in 1983 (U.S. Patent No. 4,659,516, expired in 2007 with a patent term extension; U.S. Patent Nos. 6,774, 122 and 7,456, 160). Tamoxifen was also disclosed by ICI in the '516 patent. Tamoxifen was developed for the treatment of breast cancer on the basis of strong antagonism of estrogen action in mammary tissue (Jordan, J. Cell. Biochem. 51 (1995)).
  • ovariectomized rodents to test doses of the compound both in the absence (agonist mode) and presence (antagonist mode) of estrogen.
  • Tamoxifen and other partial anti-estrogens stimulate uterine weight gain in the agonist mode and only partly block estrogen-driven uterine weight gain in the antagonist mode.
  • Fulvestrant and other complete anti-estrogens do not stimulate uterine weight gain in the agonist mode and completely block estrogen-driven weight gain in the antagonist mode.
  • the induction of estrogen-regulated alkaline phosphatase expression in human uterine cancer cell growth in culture can be used to distinguish partial and complete anti-estrogenicity and correlates well with the rodent weight gain assay.
  • Tamoxifen and fulvestrant both inhibit cultured human breast cancer cell proliferation provoked by estrogen.
  • fulvestrant more fully inhibits the proliferation when provoked with growth factors, especially of the insulin/insulin-like growth factor family.
  • growth-factor driven breast cancer cell proliferation and the effect on uterine weight provide two assays which can distinguish between complete and partial anti- estrogens.
  • Tamoxifen binding stabilizes the estrogen receptor whereas fulvestrant and chemically related antiestrogens, such as ICI-164384 and RU-58668, cause degradation of the estrogen receptor.
  • fulvestrant and chemically related antiestrogens such as ICI-164384 and RU-58668, cause degradation of the estrogen receptor.
  • some compounds like GW-5638 (Wu et al, Mol Cell.,18,413 (2005), and OP1075, described below, degrade the receptor but are partial estrogens- that is, not complete anti-estrogens.
  • the ability to degrade the estrogen receptor does not ensure complete antiestrogenicity.
  • the ability to induce degradation of the receptor is nonetheless a factor that differentiates the behavior of tamoxifen and fulvestrant and may be desirable in a drug to treat breast cancer.
  • Fulvestrant which degrades the estrogen receptor, incorporates a core of 17-beta estradiol. It has a long flexible aliphatic side chain that blocks oral absorption. The estradiol core blocks oral absorption and the long flexible aliphatic side chain makes the drug very insoluble which worsens the problem. Fulvestrant must be injected because of the poor oral bioavailability. Two 5 ml intramuscular depot injections, one into each buttock, must be administered monthly by a health professional. Furthermore, it is unclear whether these two injections provide sufficient drug exposure for optimal action. The drug does not seem to work in pre-menopausal women.
  • Patent No. 5,395,842 (see claim 29) which taught that EM-343 (H), showed superior binding to the estrogen receptor with no loss of anti-estrogen action.
  • EM-343 differed from the Saeed compounds by including the hydroxyl at the 4'-position of a 4-methyl, 7-hydroxyl benzopyran.
  • Labrie et al. filed a continuation-in-part patent application, which issued in 2000 as U.S. Patent No. 6,060,503, disclosing prodrugs and optically active species of EM- 343.
  • Labrie et al. disclosed a pure isomer of EM-343, EM-652, referred to as acolbifene (I), which is (S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-(2-(piperidin-l- yl)ethoxy)phenyl)-2H-chromen-7-ol.
  • D is -OCH 2 CH 2 N(R 3 )R4 (R 3 and R 4 either being independently selected from the group consisting of C 1 -C 4 alkyl, or R 3 , R 4 and the nitrogen atom to which they are bound together being a ring structure selected from the group consisting of pyrrolidino, dimethyl- 1-pyrrolidino, methyl-l-purrolidinyl, piperidino, hexamethyleneimino and morpholino); and
  • Ri and R 2 are independently selected from the group consisting of hydrogen, hydroxyl and a moiety converted in vivo in to hydroxyl, and
  • Ri and R 2 are independently selected from the group consisting of hydroxyl and a moiety converted in vivo in to hydroxyl;
  • R 3 is a species selected from the group consisting of saturated, unsaturated or substituted pyrrolidinyl, saturated, unsaturated or substituted piperidino, saturated, unsaturated or substituted piperidinyl, saturated, unsaturated or substituted morpholino, nitrogen-containing cyclic moiety, nitrogen-containing polycyclic moiety, and RaRb (Ra and Rb being independently hydrogen, straight or branched Ci-C 6 alkyl, straight or branched C 2 -C 6 alkenyl, and straight of branched C 2 -C 6 alkynyl.
  • Acolbifene binds to the estrogen receptor alpha with three times the affinity of 17- beta estradiol, the native ligand (Katzenellenbogen (2011) J Med Chem 54(15):5271-82). Since anti-estrogens must compete with estradiol for binding to the estrogen receptor, high affinity binding is an important drug virtue.
  • EM-800, a pivalate prodrug of EM-652, and HC1 salts of EM-652 were also described in the '503 patent.
  • Acolbifene was initially thought to be a complete anti-estrogen.
  • careful studies with the rodent uterine assay and human uterine cell alkaline phosphatase assays revealed that it retained some estrogen-like action, about 12% that of estradiol (Labrie et al. "The combination of a novel selective estrogen receptor modulator with an estrogen protects the mammary gland and uterus in a rodent model: the future of postmenopausal women's health?" Endocrinology. 2003 144(11):4700-6). This contrasts with fulvestrant where the residual estrogen-like action is almost unmeasurable.
  • Acolbifene is orally bioavailable and is currently being positioned for Phase III clinical trials for the treatment of breast cancer by the Canadian company Endoceutics (Founded by Dr. Labrie).
  • a daily oral dose of 40 mg of acolbifene or EM800 in women produces mean drug exposures of 8.3 and 15 ng/ml of circulating acolbifene, respectively.
  • the Merck chromane core differs from the acolbifene core by the absence of a double bond in the oxane ring. These structures also had a hydroxyl at position 6 (not 7) of the fused benzene ring.
  • a chromane core with a 2-m ethyl pyrrolidine (but not a 3 -methyl) with a methyl on the linker created a nearly complete anti-estrogen, (see compound 12 of the Blizzard et al. paper).
  • Blizzard et al. commented on the differences among anti-estrogenic activities of variously substituted cores, and noted that the size and stereogenic placement of substituents is crucial for receptor potency and selectivity.
  • Blizzard et al. again studied the dihydrobenzoxathiin core and reported that their studies have resulted in the discovery that addition of a methyl group to the side chain at the appropriate position and with the right stereochemistry, either on the pyrrolidine ring or on the linker substantially increased estrogen antagonist activity in uterine tissue. Blizzard et al. also reported that the best estrogen antagonist activity in this dihydrobenzoxathiin series was determined to have a methyl group on the pyrrolidine and a methyl group on the linker, with the hydroxyl in the 6-position of the fused benzene ring. Blizzard et al.
  • SRMs Selective Estrogen Receptor Modulators
  • Kushner et al. (US 2013/0178445 and WO 2013/090921, both filed Dec. 17, 2012 and both assigned to Olema Pharmaceuticals) describe OP-1038 (3-(4-hydroxyphenyl)-4-methyl- 2-(4- ⁇ 2-[(3R)-3-methylpyrrolidin-l-yl]ethoxy ⁇ phenyl)-2H-chromen-7-ol) and OP-1074 ((2S)-3- (4-hydroxyphenyl)-4-methyl-2-(4- ⁇ 2-[(3R)-3-methylpyrrolidin-l-yl]ethoxy ⁇ phenyl)-2H- chromen-7-ol), as well as pharmaceutical compositions and methods of use.
  • Bazedoxifene is a SERM, under development for prevention and treatment of postmenopausal osteoporosis (Biskobing, D. M. (2007) Clinical interventions in aging 2 (3): 299-303).
  • Lasofoxifene is another SERM under development for the treatment of
  • U.S. Patent 5,254,568 discloses benzopyrans as anti-estrogenic agents.
  • WO2010/145010 discloses a combination of SERM and sex steroid precursor for treating hot flashes and other symptoms.
  • WO2004/091488 discloses benzopyrans as estrogen receptor modulators.
  • U.S. Patent 5,840,735 discloses benzopyrans as sex steroid activity inhibitors.
  • U.S. Patent 6,262,270 discloses a method for the enantiomeric synthesis of acolbifene derivatives.
  • Embodiments described herein relate to a method of treating a disorder modulated, mediated or affected by the estrogen receptor in a subject comprising administering to the subject a compound, which is
  • Ri and R 2 are independently either:
  • R can be independently selected from hydrogen, polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or two R groups are joined to form an substituted or unsubstituted heterocyclic ring;
  • an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic.
  • the compound is in the form of a pharmaceutically acceptable salt.
  • either or both of Rl or R2 is an ester, amide, carbonate or phosphate.
  • compound is in the form of a prodrug, further wherein the prodrug is
  • the compound is (2S)-3-(4- hydroxyphenyl)-4-methyl-2-(4- ⁇ 2-[(3R)-3 -methyl pyrrolidin-1 -yl]ethoxy ⁇ phenyl)-2H-chromen- 7-ol, which has the chemical structure:
  • Embodiments described herein relate to a method of treating a disorder modulated, mediated or affected by the estrogen receptor in a subject comprising administering to the subject a compound, which is (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4- ⁇ 2-[(3R)-3- methylpyrrolidin-l-yl]ethoxy ⁇ phenyl)-2H-chromen-7-ol, which has the chemical structure:
  • the compound is administered in alternation with an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic.
  • Embodiments described herein relate to a combination of a compound, which is
  • Ri and R 2 are independently either:
  • R 9 is independently selected from H, halogen (CI, Br, I or F), natural or non- naturally occurring amino acid (bound through either the OC(O)- or C(0)0- (an ester) or the amino (through either -C(0)-N- or -N-C(O)- (an amide linkage)), R 10 , -OR 10 , or -SR 1(
  • R can be independently selected from hydrogen, polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or two R groups are joined to form an substituted or unsubstituted heterocyclic ring;
  • the compound is in the form of a pharmaceutically acceptable salt.
  • the compound is (2S)-3-(4- hydroxyphenyl)-4-methyl-2-(4- ⁇ 2-[(3R)-3 -methyl pyrrolidin-1 -yl]ethoxy ⁇ phenyl)-2H-chromen- 7-ol, which has the chemical structure:
  • Embodiments described herein relate to a combination of (2S)-3-(4- hydroxyphenyl)-4-methyl-2-(4- ⁇ 2-[(3R)-3 -methyl pyrrolidin-1 -yl]ethoxy ⁇ phenyl)-2H- 7-ol, which has the chemical structure:
  • the disorder is mediated by the estrogen receptor.
  • the disorder is breast cancer.
  • the breast cancer is local, advanced or metastatic estrogen or progesterone receptor positive advanced breast cancer.
  • the breast cancer is estrogen or progesterone receptor positive advanced breast cancer.
  • the breast cancer is estrogen or progesterone receptor negative breast cancer.
  • the disorder is selected from the group consisting of ovarian cancer, endometrial cancer, vaginal cancer, endometriosis, lung cancer, and bronchial cancer.
  • the disorder is selected from the group consisting of ovarian, endometrial, and vaginal cancer and
  • the disorder is retinoblastoma positive breast cancer.
  • the disorder is selected from the group consisting of retinoblastoma positive endometrial, vaginal and ovarian cancers and lung and bronchial cancers.
  • the disorder is lung cancer or bronchial cancer.
  • the subject is a patient.
  • the agent is a
  • the CDK 4/6 inhibitor is PD-0332991, LY2835219, or LEE011.
  • the CDK 4/6 inhibitor is PD-0332991, or a pharmaceutically acceptable salt thereof.
  • the CDK 4/6 inhibitor is PD-0332991.
  • the agent is an mTOR inhibitor.
  • the mTOR inhibitor is rapamycin, everolimus, temsirolimus, AP23573, AZD8055, WYE-354, WYE-600, WYE-687, or Ppl21.
  • the mTOR inhibitor is everolimus.
  • the agent is a
  • the PI3 Kinase inhibitor is BKM-120, XL-147, RG-7321, CH-5132799 and BAY-80-6946.
  • the PI3 Kinase inhibitor is BKM-120.
  • the agent is a taxane.
  • the taxane is paclitaxel or docetaxel.
  • the agent is an antimetabolite.
  • the antimetabolite is 5- fluorouracil.
  • the agent is an antitumor antiobiotic.
  • the antitumor antibiotic is doxorubicin.
  • Embodiments described herein relate to a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically effective amount of a compound, which is
  • Ri and R 2 are independently either:
  • R C1 can be independently selected from hydrogen, polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or two R groups are joined to form an substituted or unsubstituted heterocyclic ring;
  • an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic, and a pharmaceutically acceptable carrier.
  • the compound is in the form of a pharmaceutically acceptable salt.
  • Ri or ester, amide, carbonate or phosphate is in embodiments of the pharmaceutical composition of the invention.
  • the compound is in the form o
  • the compound is (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4- ⁇ 2-[(3R)-3-methylpyrrolidin-l- yl]ethoxy ⁇ phenyl)-2H-chromen-7-ol, which has the chemical structure:
  • Embodiments described herein relate to a pharmaceutical composition of the invention relates to a compound, which is (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4- ⁇ 2-[(3R)-3- methylpyrrolidin-l-yl]ethoxy ⁇ phenyl)-2H-chromen-7-ol, which has the chemical structure:
  • an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic, and a pharmaceutically acceptable carrier.
  • the agent is a
  • the CDK 4/6 inhibitor is PD-0332991, LY2835219, or LEE011.
  • the CDK 4/6 inhibitor is PD-0332991, or a pharmaceutically acceptable salt thereof.
  • the CDK 4/6 inhibitor is PD-0332991.
  • the agent is an mTOR inhibitor.
  • the mTOR inhibitor is rapamycin, everolimus, temsirolimus, AP23573, AZD8055, WYE-354, WYE-600, WYE-687, or Ppl21.
  • the mTOR inhibitor is everolimus.
  • the agent is a
  • the PI3 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N
  • Kinase inhibitor is BKM-120, XL-147, RG-7321, CH-5132799 and BAY-80-6946.
  • the PI3 is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N
  • Kinase inhibitor is BKM-120.
  • the agent is a taxane.
  • the taxane is paclitaxel or docetaxel.
  • the agent is an antimetabolite.
  • the antimetabolite is 5-fluorouracil.
  • the agent is an antitumor antiobiotic.
  • the antitumor antibiotic is doxorubicin.
  • the present invention relates to the treatment of tumors, including cancers, mediated by the estrogen receptor with the combination of a specific benzopyran (in the form of a mixture of S-C2 and R-C2 diastereomers and also its pure S-diastereomer) as described in US 2013/0178445 and WO 2013/090921 in combination or alternation with specific agents, active agents, anti-cancer agents or chemotherapeutic agents, and in some embodiments, in certain improved dosages or formulations.
  • Benefits that can result include increased efficacy and/or lower doses when the specified combinations are used.
  • Tumors, including cancers which benefit from such combinations or alternations are those that are mediated by the estrogen receptor and will also respond to the second agent.
  • agents, active agents, anti-cancer agents or chemotherapeutic agents that can be combined with compounds described in US 2013/0178445 and WO 2013/090921 include an mTOR inhibitor such as everolimus, or a CDK 4/6 inhibitor such as PD-0332991, or a PI3 Kinase Inhibitor such as BKM-120, or a taxane, or an antimetabolite such as 5-fluorouracil, or an antitumor antibiotic such as doxorubicin.
  • the combination therapy includes the described anti-estrogenic compounds combined with PD-0332991 for treatment of retinoblastoma positive breast cancer as well as retinoblastoma positive endometrial, vaginal and ovarian cancers and lung and bronchial cancers.
  • 2013/0178445 and WO 2013/090921 with an agent, active agent, anti-cancer agent or
  • chemotherapeutic agent wherein the concerted use can be either (i) a pharmaceutically acceptable formulation that includes both active agents; or (ii) separate pharmaceutical formulations that independently provide each active and are either administered at the same time or at different times but in a concerted synchronized fashion in a manner that optimizes the benefit of the combination therapy (i.e., alternation therapy).
  • a pharmaceutically acceptable formulation that includes both active agents
  • separate pharmaceutical formulations that independently provide each active and are either administered at the same time or at different times but in a concerted synchronized fashion in a manner that optimizes the benefit of the combination therapy (i.e., alternation therapy).
  • certain actives will normally be administered orally, such as in a tablet, pill or liquid, and others might be normally administered via intravenous injection or other systemic, topical, parenteral, transdermal or other route.
  • OP-1038 3 -(4-hydroxyphenyl)-4-methyl-2-(4- ⁇ 2- [(3R)-3 -methylpyrrolidin- 1 -yl] ethoxy ⁇ phenyl)-2H- chromen-7-ol.
  • OP-1074 is (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4- ⁇ 2- [(3R)-3-methylpyrrolidin-l-yl]ethoxy ⁇ phenyl)-2H-chromen-7-ol.
  • the active compound to be combined or alternated with an agent, active agent, anti-cancer agent or chemotherapeutic agent can be provided if desired as a pharmaceutically acceptable salt, solvate, hydrate, prodrug, stereoisomer, tautomer, N-oxide or R 1 and/or R 2 - substituted derivative optionally in a pharmaceutically acceptable composition to treat a tumor that is modulated or affected by an estrogen receptor, including those treatable with an antiestrogenic compound optimally with virtually no estrogenic effect.
  • OP-1038 and OP-1074 have two chiral carbons and thus there are four possible diastereomers.
  • the chiral carbon at the C2 position is in the S-configuration in OP-1074 (the same configuration in EM-652, acolbifene) and is a mixture of R and S in OP-1038.
  • OP-1038, OP-1074 and their prodrugs are especially useful to treat locally advanced or metastatic breast cancer that is positive for expression of estrogen receptors, progesterone receptors or both (receptor positive advanced breast cancer) and are also responsive to the specific agent, active agent, anti-cancer agent or chemotherapeutic agent.
  • the combinations are used to treat estrogen or progesterone receptor negative breast cancer. The combinations can be used as the initial treatment of advanced breast cancer in patients who have never received previous hormonal therapy for advanced breast cancer.
  • active agents that can be combined with compounds described in US 2013/0178445 and WO 2013/090921 include an mTOR inhibitor such as everolimus, or a CDK 4/6 inhibitor such as PD-0332991, or a PI3 Kinase Inhibitor such as BKM-120, or a taxane, or an mTOR inhibitor such as everolimus, or a CDK 4/6 inhibitor such as PD-0332991, or a PI3 Kinase Inhibitor such as BKM-120, or a taxane, or an mTOR inhibitor such as everolimus, or a CDK 4/6 inhibitor such as PD-0332991, or a PI3 Kinase Inhibitor such as BKM-120, or a taxane, or an mTOR inhibitor such as everolimus, or a CDK 4/6 inhibitor such as PD-0332991, or a PI3 Kinase Inhibitor such as BKM-120, or a taxane, or
  • the combination therapy includes the compounds described in US 2013/0178445 and WO 2013/090921 combined with PD-0332991 for treatment of retinoblastoma positive breast cancer as well as retinoblastoma positive endometrial, vaginal and ovarian cancers and lung and bronchial cancers.
  • the combinations described in the present invention are also useful as adjuvant therapy after surgery to prevent recurrance. Such adjuvant use is often administered for several years, for instance 5 years, or even up to 10 years after surgery and associated chemotherapy and radiotherapy have been concluded.
  • the adjuvant combination therapy includes the compounds described in US 2013/0178445 and WO 2013/090921 combined with PD-0332991 for treatment of retinoblastoma positive breast cancer as well as retinoblastoma positive endometrial, vaginal and ovarian cancers and lung and bronchial cancers.
  • the combinations described in the present invention are also useful for the prevention of breast cancer in women at high risk for breast cancer and can be taken for any desired time period, including indefinitely.
  • a patient typically a woman, with a family history of breast cancer, or who has been determined to carry a mutation in the BRCAl or BRCA2 gene or other genes that predispose a patient to breast cancer may choose to use such preventative treatment instead of a mastectomy or other intervention.
  • the combinations described herein are also useful as neoadjuvants to shrink large tumors prior to surgical removal, both to enable breast conservative surgery and to reduce the risk of recurrence.
  • these combinations are also useful to treat other cancers and other overgrowth diseases of the female reproductive tract including ovarian, endometrial, and vaginal cancer and endometriosis.
  • the combinations are useful in treating lung cancers that are positive for estrogen or progesterone receptors.
  • the combination of the compound described in US 2013/0178445 and WO 2013/090921 with PD-0332991 is beneficial.
  • Fig. 1 A plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or ⁇ OP-1074 in combination with increasing amounts of PD-0332991.
  • IB plots the effects on MCF-7 cell proliferation of vehicle or lOOnM PD-0332991 in combination with increasing amounts of OP- 1074.
  • Fig. 1C plots the effects on MCF-7 cell proliferation of vehicle or 3nM OP- 1074, tamoxifen, or fulvestrant in combination with increasing amounts of PD-0332991. Proliferation was measured using Cyquant fluorescent DNA-binding dye (Invitrogen, Grand Island, NY). Results were from a single representative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing SEM.
  • FIG. 1 plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or lOOnM OP-1074 in combination with increasing amounts of everolimus.
  • Fig. 1 plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or lOOnM OP-1074 in combination with increasing amounts of everolimus.
  • FIG. 2B plots the effects on MCF-7 cell proliferation of vehicle, InM or ⁇ everolimus in combination with increasing amounts of OP-1074.
  • Fig. 2C plots the effects on MCF-7 cell proliferation of vehicle or 3nM OP-1074, tamoxifen, or fulvestrant in combination with increasing amounts of everolimus. Proliferation was measured as in Figure 1 and results were from a single representative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing SEM.
  • FIG. 3 OP-1074 increases the potency of the pan PI3K inhibitor BKM-120 in inhibiting E2-stimulated MCF-7 breast cell proliferation.
  • the antiestrogen OP-1074 enhances the efficacy and potency of the PI3K inhibitor BKM-120 in inhibiting E2-stimulated breast cell proliferation in vitro.
  • MCF-7 cells were treated with the indicated amount of OP-1074 and BKM-120 in hormone-depleted medium for 7 days in the presence of 100 pM 17P-estradiol (E2).
  • Fig. 3 A plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or lOOnM OP-1074 in combination with increasing amounts of BKM-120.
  • Fig. 3B plots the effects on MCF-7 cell proliferation of vehicle, 32nM or lOOnM BKM-120 in combination with increasing amounts of OP-1074. Proliferation was measured as in Figure 1 and results were from a single
  • FIG. 4 OP-1074 increases the potency of 5-Fluorouracil in inhibiting E2- stimulated MCF-7 breast cell proliferation.
  • the antiestrogen OP-1074 enhances the efficacy and potency of the thymidylate synthase inhibitor 5-Fluorouracil in inhibiting E2-stimulated breast cell proliferation in vitro.
  • MCF-7 cells were treated with the indicated amount of OP-1074 and 5- fluorouracil in hormone-depleted medium for 6 days in the presence of 100 pM 17P-estradiol (E2). The effects on MCF-7 cell proliferation of vehicle, 1, 10 or ⁇ OP-1074 in
  • MCF-7 breast cell proliferation The antiestrogen OP-1074 enhances the efficacy and potency of paclitaxel and docetaxel in inhibiting E2-stimulated breast cell proliferation in vitro.
  • MCF-7 cells were treated with the indicated amount of OP-1074 and taxane in hormone-depleted medium for 6 days in the presence of 100 pM 17P-estradiol (E2).
  • Fig. 5 A plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or ⁇ OP-1074 in combination with increasing amounts of paclitaxel.
  • Fig. 5B plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or ⁇ OP- 1074 in combination with increasing amounts of docetaxel. Proliferation was measured as in Figure 1 and results were from a single representative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing SEM.
  • OP-1074 increases the potency of the anthracycline doxorubicin in inhibiting E2-stimulated MCF-7 breast cell proliferation.
  • the antiestrogen OP-1074 enhances the efficacy and potency of the anthracycline doxorubicin in inhibiting E2-stimulated breast cell proliferation in vitro.
  • MCF-7 cells were treated with the indicated amount of OP-1074 and doxorubicin in hormone-depleted medium for 7 days in the presence of 100 pM 17P-estradiol (E2). The effects on MCF-7 cell proliferation of vehicle, 1, 10 or ⁇ OP-1074 in
  • the present invention relates to the treatment of tumors, including cancers, mediated by the estrogen receptor with the combination of a specific benzopyran (in the form of a mixture of S-C2 and R-C2 diastereomers and also its pure S-diastereomer) as described in US 2013/0178445 and WO 2013/090921 in combination or alternation with specific agents, active agents, anti-cancer agents or chemotherapeutic agents, and in some embodiments, in certain improved dosages or formulations.
  • Benefits that can result include increased efficacy and/or lower doses when the specified combinations are used. Cancers which benefit from such combinations or alternations are those that are mediated by the estrogen receptor and will also respond to the second agent.
  • agents, active agents, anti-cancer agents or chemotherapeutic agents that can be combined with compounds described in US 2013/0178445 and5 and WO 2013/090921 include an mTOR inhibitor such as everolimus, or a CDK 4/6 inhibitor such as PD-0332991, or a PI3 Kinase Inhibitor such as BKM-120, or a taxane, or an antimetabolite such as 5-fluorouracil, or an antitumor antibiotic such as doxorubicin.
  • an mTOR inhibitor such as everolimus
  • a CDK 4/6 inhibitor such as PD-0332991
  • a PI3 Kinase Inhibitor such as BKM-120
  • an antimetabolite such as 5-fluorouracil
  • an antitumor antibiotic such as doxorubicin.
  • the combination therapy includes the described anti-estrogenic compounds combined with PD-0332991 for treatment of retinoblastoma positive breast cancer as well as retinoblastoma positive endometrial, vaginal and ovarian cancers and lung and bronchial cancers.
  • the compound to be provided in combination or alternation with particular agents, active agents, anti-cancer agents or chemotherapeutic agents can be provided if desired as a pharmaceutically acceptable salt, solvate, hydrate, prodrug, stereoisomer, tautomer, N-oxide or Ri and/or R 2 -substituted derivative or a pharmaceutically acceptable composition thereof to treat a disorder that is mediated, modulated or affected by an estrogen receptor, including those treatable with an anti-estrogenic compound with virtually no estrogenic effect.
  • Ri and R 2 are independently either:
  • R can be independently selected from hydrogen, polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or two R groups are joined to form an substituted or unsubstituted heterocyclic ring.
  • Ri or R 2 is an ester, amide, carbonate or phosphate.
  • prodrugs of the described compounds that can be combined or alternated with the particular agents, active agents, anti-cancer agents or chemotherapeutic agents are:
  • Examples of useful metabolically cleavable prodrug groups include acetyl, methoxycarbonyl, benzoyl, methoxymethyl and trimethylsilyl groups.
  • Combinations of OP-1038, OP-1074 and their prodrugs are especially useful to treat locally advanced or metastatic breast cancer that is positive for expression of estrogen receptors, progesterone receptors or both (receptor positive advanced breast cancer).
  • the combinations are used to treat estrogen or progesterone receptor negative breast cancer.
  • active agents include an mTOR inhibitor such as everolimus, or a CDK 4/6 inhibitor such as PD-0332991, or a PI3 Kinase Inhibitor such as BKM-120, or a taxane, or an antimetabolite such as 5-fluorouracil, or an antitumor antibiotic such as doxorubicin.
  • an mTOR inhibitor such as everolimus
  • a CDK 4/6 inhibitor such as PD-0332991
  • a PI3 Kinase Inhibitor such as BKM-120
  • a taxane or an antimetabolite such as 5-fluorouracil
  • an antitumor antibiotic such as doxorubicin.
  • 2013/090921 can be combined with a CDK 4/6 inhibitor, for example PD-0332991.
  • OP-1074 is combined with PD-0332991.
  • 2013/090921 can be combined with an mTOR inhibitor, for example everolimus.
  • OP-1074 is combined with everolimus.
  • 2013/090921 can be combined with a PI3 Kinase Inhibitor, for example BKM-120.
  • OP-1074 is combined with BKM-120.
  • 2013/090921 can be combined with a taxane, for example paclitaxel or docetaxel.
  • OP-1074 is combined with paclitaxel or docetaxel.
  • 2013/090921 can be combined with antimetabolites, for example 5-fluorouracil.
  • OP-1074 is combined with 5-fluorouracil.
  • 2013/090921 can be combined with antitumor antibiotics, for example doxorubicin.
  • OP-1074 is combined with doxorubicin.
  • compositions are described of combinations of the compounds as described in US 2013/0178445 and WO 2013/090921 combined or alternated with particular agents, active agents, anti-cancer agents or chemotherapeutic agents.
  • the compositions can be administered in a pharmaceutical composition suitable for oral delivery to the patient, typically a human.
  • the compounds can be delivered in a carrier suitable for topical, transdermal (including by patch), intravenous, parenteral, intraortal, subcutaneous or other desired delivery route, including any method of controlled delivery, for example, using degradable polymers, or with nano or microparticles, liposomes, layered tablets or other structural frameworks which slow delivery.
  • compositions can be in the form of salts of the actives. They can be administered as a pharmaceutically acceptable salt, for example, a pharmaceutically acceptable acid addition salt, including a hydrochloride, hydroiodide, hydrobromide, nitrate, sulfate, bisulfate, phosphate, acetate, lactate, citrate, tartrate, succinate, maleate, fumarate, benzoate, para-toluenesulfonate and the like.
  • a pharmaceutically acceptable salt for example, a pharmaceutically acceptable acid addition salt, including a hydrochloride, hydroiodide, hydrobromide, nitrate, sulfate, bisulfate, phosphate, acetate, lactate, citrate, tartrate, succinate, maleate, fumarate, benzoate, para-toluenesulfonate and the like.
  • the compositions are used to treat or prevent a disorder modulated by the estrogen receptor in an animal, typically a mammal,
  • the combinations can also be used as adjunctive therapy.
  • a therapeutically effective amount of the compound can be used in combination with another anticancer agent, especially for estrogen receptor positive breast cancer, but in some embodiments, for estrogen receptor negative breast cancer.
  • a method of treating a patient with combinations of a once-a-day, orally administered, treatment regimen including selecting one of the plurality of treatment regimens and titrating the dosages supplied to the patient after weekly, monthly or quarterly periods of time.
  • Some embodiments of the present invention facilitate the selection of a combination treatment regimen and/or the change in dosages or titration of dosages of a treatment regimen over time.
  • the combination of the compounds of the present invention with other actives, in particular other agents, active agents, anti-cancer agents or chemotherapeutic actives, can be done in a single fixed dose combination pill such as a multi-pill, combopill or polypill.
  • a polypill is a medication that is a drug product in pill form (i.e., tablet or capsule) that combines multiple active pharmaceutical ingredients. In one embodiment it is manufactured as a fixed-dose combination (FDC) drug product targeting treatment or prevention of a disease.
  • FDC fixed-dose combination
  • Polypills can reduce the number of tablets or capsules (generally orally administered) that need to be taken, which in turn may facilitate handling and administration of pharmaceuticals as well as alleviate patient pill-burden.
  • the multiple drugs in a given polypill might all be aimed at a single underlying condition (or, group of related conditions), for whom a given combination of drugs/dosages might be appropriate (particularly in the case of mass-produced polypills, i.e. FDCs).
  • polypills can also be custom-made for specific patients through a process called pharmacy compounding.
  • the present invention also relates to protocols enabling the physician or other healthcare provider to deliver the medications in a plurality of treatment regimens in a combination or alternation dose packaging (once-a-day, more than once a day, alternate days, every third day, twice a week, once a week, once every other week, once a month and so on) to increase compliance with medical therapy.
  • a combination or alternation dose packaging once-a-day, more than once a day, alternate days, every third day, twice a week, once a week, once every other week, once a month and so on
  • the present invention provides protocols for a plurality of pills into a combination dose packaging or polypill corresponding to the combination treatments mentioned above used in treating a patient for cancer, and in particular breast cancer, as well as other estrogen mediated diseases that would benefit from the compounds of the present invention, and related health issues over a weekly, monthly, quarterly, or longer period of time.
  • the present invention can further greatly improve patient compliance by providing the combination doses over a period of time (e.g. week, month, quarter, or longer) and having the physician titrate the dosages (adjust the dosages of each effective therapeutic ingredient) over the period of time based upon the condition of the patient.
  • the combination treatment pack may provide alternative dosages for treatment at more than one time per day.
  • an agent, active agent, anti-cancer agent or chemotherapeutic agent wherein the concerted use can be either (i) a pharmaceutically acceptable formulation that includes both active agents; or (ii) separate pharmaceutical formulations that independently provide each active and are either administered at the same time or at different times but in a concerted synchronized fashion in a manner that optimizes the benefit of the combination therapy (i.e., alternation therapy).
  • a pharmaceutically acceptable formulation that includes both active agents
  • separate pharmaceutical formulations that independently provide each active and are either administered at the same time or at different times but in a concerted synchronized fashion in a manner that optimizes the benefit of the combination therapy (i.e., alternation therapy).
  • certain actives will normally be administered orally, such as in a tablet, pill or liquid, and others might be normally administered via intravenous injection or other systemic, topical, parenteral, transdermal or other route.
  • a “pharmaceutical composition” can thus contain either one of the active agents or a combination of agents, based on how the active agents are best administered according to compatibility with the carrier, dosage, dosage scheduling, possibility of drug holidays and other general differences in physical properties, convenience, efficacy, toxicity and dosage regimes.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically effective amount of a compound as described in US 2013/0178445 and WO 2013/090921 in combination with a particular agent, active agent, anti-cancer agent or chemotherapeutic agent, and a pharmaceutically acceptable carrier.
  • the amount of the composition administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
  • compositions provided herein can be administered by a variety of routes including oral, topical, parenteral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal with a pharmaceutical carrier suitable for such administration.
  • the compound is administered in a controlled release formulation.
  • compositions for oral administration can take the form of bulk liquid solutions or suspensions, or bulk powders.
  • the compositions are presented in unit dosage forms to facilitate accurate dosing.
  • unit dosage forms refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.
  • Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions.
  • the compound is usually a minor component (as a nonlimiting example, from about 0.1 to about 50% by weight or preferably from about 1 to about 40% by weight) with the remainder being various vehicles or carriers and processing aids helpful for forming the desired dosing form.
  • a minor component as a nonlimiting example, from about 0.1 to about 50% by weight or preferably from about 1 to about 40% by weight
  • compositions can be used for treating a patient with combinations of a once- a-day, orally administered, treatment regimen, including selecting one of the plurality of treatment regimens and titrating the dosages supplied to the patient after weekly, monthly or quarterly periods of time.
  • Some embodiments of the present invention facilitate the selection of a combination treatment regimen and/or the change in dosages or titration of dosages of a treatment regimen over time.
  • the combination of the compounds of the present invention with other actives, in particular other chemotherapeutic actives can be done in a single fixed dose combination pill such as a multi-pill, combopill or polypill.
  • a polypill is a medication that is a drug product in pill form (i.e., tablet or capsule) that combines multiple active pharmaceutical ingredients. In one embodiment it is manufactured as a fixed-dose combination (FDC) drug product targeting treatment or prevention of a disease.
  • FDC fixed-dose combination
  • Polypills can reduce the number of tablets or capsules (generally orally administered) that need to be taken, which in turn may facilitate handling and administration of pharmaceuticals as well as alleviate patient pill-burden.
  • the multiple drugs in a given polypill might all be aimed at a single underlying condition (or, group of related conditions), for whom a given combination of drugs/dosages might be appropriate (particularly in the case of mass-produced polypills, i.e. FDCs).
  • polypills can also be custom-made for specific patients through a process called pharmacy compounding.
  • the present invention also relates to protocols enabling the physician or other healthcare provider to deliver the medications in a plurality of treatment regimens in a combination protocol (once-a-day, more than once a day, alternate days, every third day, twice a week, once a week, once every other week, once a month and so on) to increase compliance with medical therapy.
  • the present invention provides protocols for a plurality of pills into a combination protocol or polypill corresponding to the combination treatments mentioned above used in treating a patient for cancer, and in particular breast cancer, as well as other estrogen mediated diseases that would benefit from the compounds of the present invention, over a weekly, monthly, quarterly, or longer period of time.
  • the present invention can further greatly improve patient compliance by providing the combination doses over a period of time (e.g. week, month, quarter, or longer) and having the physician titrate the dosages (adjust the dosages of each effective therapeutic ingredient) over the period of time based upon the condition of the patient. If necessary, the combination treatment may provide alternative dosages for treatment at more than one time per day.
  • a period of time e.g. week, month, quarter, or longer
  • the combination treatment may provide alternative dosages for treatment at more than one time per day.
  • Liquid forms suitable for oral administration may include a suitable aqueous or nonaqueous vehicle with buffers, suspending and dispensing agents, colorants, flavors and the like.
  • Solid forms may include, for example, any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
  • a binder such as microcrystalline cellulose, gum tragacanth or gelatin
  • an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch
  • Transdermal compositions are typically formulated as a topical ointment or cream containing the active ingredient(s), for example in an amount ranging from about 0.01 to about 20% by weight, preferably from about 0.1 to about 20% by weight, preferably from about 0.1 to about 10%) by weight, and more preferably from about 0.5 to about 15%> by weight.
  • the active ingredients When formulated as an ointment, the active ingredients will typically be combined with either a suitable delivery polymeric composition, or a paraffinic or a water-miscible ointment base.
  • the active ingredients may be formulated in a cream with, for example an oil-in-water cream base.
  • Such transdermal formulations are well-known in the art and generally include additional ingredients to enhance the dermal penetration of stability of the active ingredients or the formulation. All such known transdermal formulations and ingredients are included within the scope provided herein.
  • compositions provided herein can be administered by a transdermal device.
  • Transdermal administration can be accomplished using a patch either of the reservoir or porous membrane type, or of a solid matrix variety.
  • compositions of this invention can also be administered in sustained release forms or from sustained release drug delivery systems.
  • sustained release materials can be found in Remington 's Pharmaceutical Sciences.
  • the formulation comprises water. In another embodiment, the formulation comprises a cyclodextrin derivative. In certain embodiments, the formulation comprises hexapropyl-P-cyclodextrin. In a more particular embodiment, the formulation comprises hexapropyl-P-cyclodextrin (10-50%) in water).
  • the present invention also includes pharmaceutically acceptable acid addition salts of compounds of the compositions of the invention.
  • the acids which are used to prepare the pharmaceutically acceptable salts are those which form non-toxic acid addition salts, i.e. salts containing pharmacologically acceptable anions such as the hydrochloride, hydroiodide, hydrobromide, nitrate, sulfate, bisulfate, phosphate, acetate, lactate, citrate, tartrate, succinate, maleate, fumarate, benzoate, para-toluenesulfonate, and the like.
  • Combinations of OP-1038, OP-1074 and their prodrugs are useful to treat disorders modulated, mediated or affected by the estrogen receptor.
  • the combinations of the present invention can be used to treat local, advanced or metastatic breast cancer that is positive for expression of estrogen receptors, progesterone receptors or both (receptor positive advanced breast cancer).
  • the combinations are used to treat estrogen or progesterone receptor negative breast cancer.
  • the combinations can be used as the initial treatment of advanced breast cancer in patients who have never received previous hormonal therapy for advanced breast cancer, in combination with one or more other anti-cancer agents described below or otherwise known to those skilled in the art.
  • a targeted therapy such as an mTOR inhibitor such as everolimus or a CDK 4/6 inhibitor such as PD-0332991.
  • the combinations of the invention are also useful as adjunctive therapy after or instead of chemotherapy, radiation or surgery. Such adjuvant use is often used for several years, perhaps 5 years, after chemotherapy or other therapies have been concluded, but may optimally be continued for additional years.
  • the combinations of the invention are also useful for the prevention of breast cancer in women at high risk and can be taken for any desired time period, including indefinitely.
  • a patient typically a woman, with a family history of breast cancer, or who has been determined to carry a mutation in the BRCA1 or BRCA2 gene or other genes that predispose a patient to breast cancer may choose to use such preventative treatment instead of a mastectomy or other intervention.
  • the combinations described herein are also useful as neoadjuvants to shrink large tumors prior to surgical removal, both to enable breast conservative surgery and to reduce the risk of recurrence.
  • these combinations are useful in treating other cancers and other overgrowth diseases of the female reproductive tract including ovarian, endometrial, and vaginal cancer and endometriosis. Besides these reproductive tissues the combinations are useful in treating lung cancers that are positive for estrogen or progesterone receptors.
  • the present combinations are used as therapeutic or prophylactic agents for the treatment of conditions in mammals, particularly humans whose conditions are modulated by estrogen receptors.
  • Combinations described herein are useful for treating locally advanced or metastatic breast cancer, preventing recurrence or early breast cancer after surgery, and preventing breast cancer in women at high risk. Such combinations are useful for treating all estrogen-dependent cancers of the reproductive tract including endometrial and ovarian cancers. Such combinations have potential uses in the treatment of lung and bronchial cancers that express estrogen receptors.
  • active agents that can be combined with compounds as described in US 2013/0178445 and WO 2013/090921 include an mTOR inhibitor such as everolimus, or a CDK 4/6 inhibitor such as palbociclib (PD-0332991), or a PI3 Kinase Inhibitor such as BKM-120, or taxanes, or antimetabolites such as 5-fluorouracil, or antitumor antibiotics such as doxorubicin.
  • mTOR inhibitor such as everolimus
  • CDK 4/6 inhibitor such as palbociclib (PD-0332991)
  • a PI3 Kinase Inhibitor such as BKM-120
  • taxanes or antimetabolites
  • antimetabolites such as 5-fluorouracil, or antitumor antibiotics such as doxorubicin.
  • CDK 4/6 inhibitors for the treatment of cancer are presently in the clinic.
  • CDK 4/6 normal function in cell cycle regulation is through phosphorylation of retinoblastoma protein at Serine 780 and 795. Cancers which are Retinoblastoma positive are targets for CDK 4/6 inhibitors.
  • PD-0332991 (palbociclib, Pfizer), 6-acetyl-8-cyclopentyl-5-methyl-2-(5- piperazin-l-yl-pyridin-2-ylamino)-8H-pyrido[2,3- ⁇ i]pyrimidin-7-one or 6-acetyl-8-cyclopentyl-5- methyl-2- ⁇ [5-(l-piperazinyl)-2-pyridinyl]amino ⁇ pyrido[2,3-d]pyrimidin-7(8H)-one was first described and synthesized in Barvian et al. (WO 2003/062236).
  • PD-0332991 is currently being evaluated in numerous tumor types including late- line metastatic breast cancer, liposarcoma, non-small cell lung cancer, liver cancer, ovarian cancer, glioblastoma, refractory solid tumors, multiple myeloma and mantle cell lymphoma.
  • Such cancers are generally categorized as retinoblastoma positive as CDK 4/6 normal function in cell cycle regulation is through phosphorylation of retinoblastoma protein at Serine 780 and 795.
  • PD-0332991 is dosed at 125 mg, orally once daily on Day 1 to Day 21 of every 28-day cycle followed by 7 days off treatment.
  • PD-0332991 is being evaluated in combination with the aromatase inhibitor letrozole in a comparison trial with letrozole plus placebo for 1st line metastatic breast cancer. Preliminary results reported in December 2012 suggest the combination is substantially superior to letrozole alone in progression-free survival.
  • Cancers that would benefit from the combination of compounds of the present invention, including OP- 1038 and OP- 1074 include include advanced or metastatic breast cancer, preventing recurrence or early breast cancer after surgery, and preventing breast cancer in women at high risk.
  • Other cancers include retinoblastoma positive estrogen-dependent cancers of the reproductive tract including endometrial and ovarian cancers as well as retinoblastoma positive lung and bronchial cancers that express estrogen receptors.
  • the amount of PD-0332991 administered as part of the combination according to the present invention will be an amount selected from about 1.25 mg to about 250 mg; suitably, the amount will be selected from about 2 mg to about 150 mg; suitably, the amount will be selected from about 2.5 mg to about 125 mg. Accordingly, the amount of PD-0332991 administered as part of the combination according to the present invention will be an amount selected from about 1.25 mg to about 250 mg.
  • the amount of PD-0332991 administered as part of the combination according to the present invention can be a lower dose than that currently administered alone, such as 1.25 mg, 2.5 mg, 5 mg, 7.5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 105 mg, 110 mg, 115 mg, 120 mg, or 125 mg.
  • the selected amount of PD-0332991 is administered once a day.
  • the selected amount of PD-0332991 is administered twice a day.
  • PD-0332991 administered as part of the combination according to the present invention orally once daily on Day 1 to Day 21 of every 28-day cycle followed by 7 days off treatment.
  • PD-0332991 may be administered with less days off treatment as part of every 28 day cycle, for instance daily on Day 1 to Day 22 of every 28-day cycle followed by 6 days off treatment, daily on Day 1 to Day 23 of every 28-day cycle followed by 5 days off treatment, daily on Day 1 to Day 24 of every 28-day cycle followed by 4 days off treatment, daily on Day 1 to Day 25 of every 28-day cycle followed by 3 days off treatment, daily on Day 1 to Day 26 of every 28-day cycle followed by 2 days off treatment, daily on Day 1 to Day 27 of every 28-day cycle followed by 1 days off treatment, or daily for the entire 28 -day cycle with no days of off treatment.
  • the term mTOR inhibitor, mTOR and derivatives thereof, unless otherwise defined, include but are not limited to rapamycin and its analogs, RAD001 or everolimus (Afinitor), CCI-779 or temsirolimus, AP23573, AZD8055, WYE-354, WYE-600, WYE-687 and Ppl21.
  • the mTOR inhibitor is selected form rapamycin, everolimus (Afinitor) and temsirolimus.
  • the mTOR inhibitor is selected from rapamycin and everolimus (Afinitor).
  • the mTOR inhibitor is everolimus.
  • the amount of everolimus administered as part of the combination according to the present invention will be an amount selected from about 1.25 mg to about 20 mg; suitably, the amount will be selected from about 2 mg to about 15 mg; suitably, the amount will be selected from about 2.5 mg to about 10 mg. Accordingly, the amount of everolimus administered as part of the combination according to the present invention will be an amount selected from about 1.25 mg to about 20 mg. For example, the amount of everolimus
  • administered as part of the combination according to the present invention can be 1.25 mg, 1.5 mg, 2 mg, 2.5 mg, 3 mg, 3.5 mg, 4 mg, 4.5 mg, 5 mg, 5.5 mg, 6 mg, 6.5 mg, 7 mg, 7.5 mg, 8 mg, 8.5 mg, 9 mg, 9.5 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg.
  • the selected amount of everolimus is administered twice a day.
  • the selected amount of everolimus is administered once a day.
  • the administration of everolimus will begin as a loading dose.
  • the loading dose will be an amount from 2 to 100 times the maintenance dose; suitably from 2 to 10 times; suitably from 2 to 5 times; suitably 2 times; suitably 3 times; suitably 4 times; suitably 5 times.
  • the loading does will be administered from 1 to 7 days; suitably from 1 to 5 days; suitably from 1 to 3 days; suitably for 1 day; suitably for 2 days; suitably for 3 days, followed by a maintenance dosing protocol.
  • the amount of temsirolimus administered as part of the combination according to the present invention will be an amount infused over a 30 to 60 minute period, where the amount is selected from about 5 mg to about 50 mg; suitably, the amount will be selected from about 10 mg to about 40 mg; suitably, the amount will be selected from about 15 mg to about 35 mg. Accordingly, the amount of temsirolimus administered as part of the combination according to the present invention will be an amount selected from about 5 mg to about 50 mg.
  • the amount of temsirolimus administered as part of the combination according to the present invention can be 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg.
  • the selected amount of temsirolimus is administered twice a day.
  • the selected amount of temsirolimus is administered once a day.
  • the administration of temsirolimus will begin as a loading dose.
  • the loading dose will be an amount from 2 to 100 times the maintenance dose; suitably from 2 to 10 times; suitably from 2 to 5 times; suitably 2 times; suitably 3 times; suitably 4 times; suitably 5 times.
  • the loading does will be administered from 1 to 7 days; suitably from 1 to 5 days; suitably from 1 to 3 days; suitably for 1 day; suitably for 2 days; suitably for 3 days, followed by a maintenance dosing protocol.
  • PI3K inhibitors may include, without being limited to, BKM-120, XL- 147, RG-
  • Phosphoinositides which are phosphorylated derivatives of
  • phosphatidylinositol are essential in eukaryotic cells, regulating nuclear processes, cytoskeletal dynamics, signaling and membrane trafficking.
  • PI3 -kinases PI3K
  • PI3-kinases phosphorylate phosphatidylinositols or Pis at the 3-position of the inositol ring.
  • the 3- phosphorylated phospholipids generated by PI3K activity bind to the pleckstrin homology (PH) domain of protein kinase B (PKB or AKT), causing translocation of PKB to the cell membrane and subsequent phosphorylation of PKB.
  • Phosphorylated PKB inhibits apoptosis-inducing proteins such as FKHR, Bad, and caspases, and is thought to play an important role in cancer progression.
  • the PDKs are divided into classes I-III, and class I is further subclassified into classes la and lb. Among these isoforms, class la enzymes are thought to play the most important role in cell proliferation in response to growth factor-tyrosine kinase pathway activation
  • PI3-kinases are attractive targets for the treatment of proliferative diseases.
  • PI3 -kinase inhibitors including wortmannin and
  • wortmannin is a potent PI3K inhibitor with a low nanomolar IC 50 value, it has low in vivo anti-tumor activity.
  • a group of morpholine substituted quinazoline, pyridopyrimidine and thienopyrimidine compounds have been reported to be effective in inhibiting PI3 kinase pi 10. alpha.. (Hayakawa, 6847-6858).
  • Oral dosage of a morpholine substituted thienopyrimidine compound has shown tumor suppression in glioblastoma xenografts in vivo.
  • the following publications disclose a series of thienopyrimidine, pyridopyrimidine and quinazoline based PI3-Kinase inhibitors: WO 2008/073785; WO 2008/070740; WO 2007/127183; U.S. Patent Publication 20080242665.
  • Phosphatidylinositol-3 -kinase (PI3-kinase) inhibitors include, but are not limited to, e.g., celecoxib and analogs thereof, such as OSU-03012 and OSU-03013 (e.g., Zhu et al., Cancer Res., 64(12): 4309-18, 2004); 3-deoxy-D-myo-inositol analogs (e.g., U.S. Application No. 20040192770; Meuillet et al., Oncol.
  • quinazoline-4-one derivatives such as IC486068 (e.g., U.S. Application No. 20020161014; Geng et al., Cancer Res., 64:4893-99, 2004); 3-(hetero)aryloxy substituted benzo(b)thiophene derivatives (e.g., WO 04 108715; also WO 04 108713); viridins, including semi -synthetic viridins such as such as PX-866 (acetic acid ( 1 S,4E, 1 OR, 11R, 13 S, 14R)-[4-diallylaminomethylene-6-hydroxy- 1 -methoxymethy- 1-10,13- dimethyl-3,7, 17-trioxo-l,3,4,7, 10,l l, 12,13, 14,15, 16,17-dodecahydro- -2-oxa- cyclopenta[a]phenanthren-l
  • IC486068 e.g., U.S. Application No
  • the PI3K inhibitor is 5-[2,6-di(4-morpholinyl)-4-pyrimidinyl]-4- (trifluoromethyl)-2-pyridinamine (BKM-120).
  • BKM-120 is currently being tested in Phase III clinical trials at a daily dose of 100 mg via oral capsules.
  • the amount of BKM-120 administered as part of the combination according to the present invention will be an amount selected from about 1.25 mg to about 250 mg; suitably, the amount will be selected from about 2 mg to about 150 mg; suitably, the amount will be selected from about 2.5 mg to about 100 mg.
  • the amount of BKM-120 administered as part of the combination according to the present invention will be an amount selected from about 1.25 mg to about 250 mg.
  • administered as part of the combination according to the present invention can be a lower dose than that currently administered alone, such as 1.25 mg, 2.5 mg, 5 mg, 7.5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, or 95 mg.
  • the selected amount ofBKM-120 is administered once a day.
  • the selected amount of BKM-120 is administered twice a day.
  • Efudex is a drug that is a pyrimidine analog which is used in the treatment of cancer. It is a suicide inhibitor and works through irreversible inhibition of thymidylate synthase.
  • 5-FU is an intravenously (IV) administered fluorinated pyrimidine cytotoxic agent that inhibits the function of thymidylate synthase, an enzyme necessary for the production of the thymidine nucleotides required for DNA synthesis.
  • 5-FU has activity in the therapy of a number of tumor types but is most commonly given in the treatment of colorectal cancer, upper gastrointestinal malignancies, and breast cancer.
  • 5-FU is currently administered in various protocols such as 500 mg/m 2 IV on
  • a method that has been used to overcome the poor oral bioavailability of 5-FU involves the administration of a prodrug that has good bioavailability and is ultimately converted to 5-FU.
  • Capecitabine penentyl [l-(3,4-dihydroxy-5-methyltetrahydrofuran-2-yl)-5-fluoro-2-oxo- lH-pyrimidin-4-yl]carbamate, Xeloda) is such a novel oral fluoropyrimidine carbamate. It is readily absorbed from the gastrointestinal tract and is preferentially converted to 5-FU in tumor tissue.
  • capecitabine After oral administration, capecitabine passes intact from the gastrointestinal tract to the liver, where it is converted by carboxylesterases to 5'-deoxy-5-flourocytidine (5'-DFCR), then by cytidine deaminase in liver and tumor tissue to 5'-deoxy-5-flourouridine (5'-DFUR), and finally by thymidine phosphorylase (dThdPase) in tumor tissue to 5-FU.
  • the usual starting dose is 2,500 mg/m2/day in two divided doses, 12 hours apart.
  • One cycle includes two weeks of treatment followed by one week without treatment. Cycles can be repeated every three weeks.
  • the dosage of 5-FU may be, for example, 10-1000 mg/ m 2 /day, preferably 50-1000 mg/ m 2 /day and more preferably 100-600 mg/ m 2 /day.
  • the dosage Capecitabine may be, for example, 100-10000 mg/ m 2 /day, preferably 500-5000 mg/ m 2 /day and more preferably 1000-5000 mg/ m 2 /day.
  • Doxorubicin also known as Adriamycin, is one of the more common cytotoxic agents used for the treatment of breast cancer.
  • the term doxorubicin includes doxorubicin as well as the pharmaceutically acceptable salts of doxorubicin.
  • Doxorubicin or hydroxyldaunorubicin is an antineoplastic drug widely used in chemotherapy (Hortobagyi, Drugs, 54 (Supplement 4): 1-7 (1997)). It is an anthracycline antibiotic and structurally closely related to daunomycin (Minotti et al., Pharmacologiy. Rev., 56; 185-229 (2004).
  • Doxorubicin is commonly used in the treatment of a wide range of cancers, including cancers of the blood, lymph system, bladder, breast, stomach, lung, ovaries, thyroid, nerves, kidneys, bones, soft tissues, including muscles and tendons, multiple myeloma, and others.
  • the hydrochloride salt of doxorubicin is one of the most common forms.
  • doxorubicin hydrochloride 14- hydroxydaunorubicin hydrochloride; 3-hydroxyacetyldaunorubicin hydrochloride; and 5, 12- naphthacenedione, 10-[(3-amino-2,3,6-trideoxy-.alpha.-L-lyxo-hexopyranosyl)oxy]-7,8,9, 10-te- trahydro-6,8,1 l-trihydroxy-8-(hydroxyacetyl)-l-methoxy-, hydrochloride, (8S-cis)-(9Cl).
  • Doxorubicin hydrochloride has the molecular formula C 2 7H 2 9NC)ii.HCl, a molecular weight (MW) of 580.0, and CAS number 25316-40-9.
  • the dosage amount of doxorubicin is preferably in the range from 30 to 100 mg/m 2 /day, more preferably 40 to 80 mg/m 2 /day, more preferably a dose of about 50 mg/m 2 /day or about 60 mg/m 2 /day.
  • Infusion times for doxorubicin are generally up to 6 hours, more preferable 1-3 hours, with 1 hour most preferred.
  • the dosage may be, for example, 10-100 mg/ m 2 /day, preferably 30-100 mg/ m 2 /day and more preferably 30-60 mg/ m 2 /day.
  • Taxane Combination Therapy examples include paclitaxel (trade name: Taxol) and docetaxel (trade name: Taxotere). Poliglumex paclitaxel (trade name: Opaxio) is also included in taxanes. Such taxanes have been approved and developed for application to breast cancer, non-small-cell lung cancer, gastric cancer, head and neck cancer, ovarian cancer, esophageal cancer, gastric cancer, uterine body cancer or the like. In addition, combination therapy for various cancers has also been approved or developed by combining taxane with various drugs, for example, with bevacizumab for breast cancer and with carboplatin for ovarian cancer and non-small-cell lung cancer.
  • Taxane may be administered according to known clinical practice.
  • the dosage and dosing schedule may be altered according to a specific symptom or all symptoms of the patient's disease.
  • the dosage may appropriately be reduced according to age, symptoms or incidence of side effects.
  • taxane may usually, but without limitation, be administered for 0.01-10000 mg/m 2 /day, preferably 0.1- 1000 mg/m 2 /day and more preferably 1-500 mg/m 2 /day for an adult, which may be administered usually once to three times a day.
  • the dosage needs to be reduced if undue toxicity occurs in the patient.
  • the dosage and dosing schedule may be altered when one or more additional agents, active agents, anti-cancer agents or chemotherapeutic agents are used in addition to the combination therapy of the invention.
  • the dosing schedule may be determined by the physician in charge of the treatment of the specific patient.
  • the dosage paclitaxel may be 0.01-10000 mg/m 2 /day, preferably 0.1-1000 mg/m 2 /day and more preferably 1-500 mg/m 2 /day.
  • the dosage of docetaxel may be 0.01-10000 mg/m 2 /day, preferably 0.1-1000 mg/m 2 /day and more preferably 1-500 mg/m 2 /day.
  • the dosage of poliglumex paclitaxel may be 0.01-10000 mg/m 2 /day, preferably 0.1- 1000 mg/m 2 /day and more preferably 1-500 mg/m 2 /day.
  • the combinations of the present invention are provided for use in medical therapy, including for any of the conditions described herein.
  • the use of the present compositions in the manufacture of a medicament for the treatment or prevention of one of the aforementioned conditions and diseases is also provided.
  • Injection dose levels range are provided in any desired dosage of the active agents, for example, from about 0.1 mg/kg/hour to at least 10 mg/kg/hour, all for from about 1 to about 120 hours and especially 24 to 96 hours.
  • a preloading bolus of from about 0.1 mg/kg to about 10 mg/kg or more may also be administered to achieve adequate steady state levels.
  • the maximum total dose is not expected to exceed about 2 g/day for a 40 to 80 kg human patient.
  • any dose of the active agents, as combined, is appropriate that achieved the desired goals.
  • suitable daily dosages of active agents of the present invention are between about 0.1-4000 mg, more typically between 5 mg and 1 gram, more typically between 10 mg and 500 mg, and administered orally once-daily, twice-daily or three times-daily, continuous (every day) or intermittently (e.g., 3-5 days a week).
  • the dose of the compounds of this invention usually ranges between about 0.1 mg, more usually 10, 50, 100, 200.250, 1000 or up to about 2000 mg per day.
  • the compounds provided herein may be isolated and purified by known standard procedures. Such procedures include (but are not limited to) recrystallization, column chromatography or HPLC. The following schemes are presented with details as to the preparation of representative substituted benzopyrans that have been listed herein.
  • the compounds provided herein may be prepared from known or commercially available starting materials and reagents by one skilled in the art of organic synthesis.
  • the diastereomerically or enantiomerically pure compounds provided herein may be prepared according to any techniques known to those of skill in the art. For instance, they may be prepared by chiral or asymmetric synthesis from a suitable optically pure precursor or obtained from a racemate or mixture of diastereomers by any conventional technique, for example, by chromatographic resolution using a chiral column, TLC or by the preparation of diastereoisomers, separation thereof and regeneration of the desired enantiomer or diastereomer. See, e.g., "Enantiomers, Racemates and Resolutions," by J. Jacques, A. Collet, and S.H. Wilen, (Wiley-Interscience, New York, 1981); S.H.
  • a diastereomerically pure compound may be obtained by reaction of the racemate or mix of diastereomers with a suitable optically active acid or base.
  • suitable acids or bases include those described in Bighley et al.., 1995, Salt Forms of Drugs and Adsorption, in Encyclopedia of Pharmaceutical Technology, vol. 13, Swarbrick & Boylan, eds., Marcel Dekker, New York; ten Hoeve & H. Wynberg, 1985, Journal of Organic Chemistry 50:4508-4514; Dale & Mosher, 1973, J. Am. Chem. Soc. 95:512; and CRC Handbook of Optical Resolution via Diastereomeric Salt Formation, the contents of which are hereby incorporated by reference in their entireties.
  • Enantiomerically or diastereomerically pure compounds can also be recovered either from the crystallized diastereomer or from the mother liquor, depending on the solubility properties of the particular acid resolving agent employed and the particular acid enantiomer or diastereomer used.
  • the identity and optical purity of the particular compound so recovered can be determined by polarimetry or other analytical methods known in the art.
  • the diasteroisomers can then be separated, for example, by chromatography or fractional crystallization, and the desired enantiomer or diastereomer regenerated by treatment with an appropriate base or acid.
  • the other enantiomer or diasteromer may be obtained from the racemate or mix of diastereomers in a similar manner or worked up from the liquors of the first separation.
  • enantiomerically or diastereomerically pure compound can be separated from racemic compound or a mixture of diastereomers by chiral
  • chiral columns and eluents for use in the separation of the enantiomers or diastereomers are available and suitable conditions for the separation can be empirically determined by methods known to one of skill in the art.
  • Exemplary chiral columns available for use in the separation of the enantiomers provided herein include, but are not limited to
  • CHIRALPACK® IC CHIRALCEL® OB, CHIRALCEL® OB-H, CHIRALCEL® OD, CHIRALCEL® OD-H, CHIRALCEL® OF, CHIRALCEL® OG, CHIRALCEL® OJ and CHIRALCEL® OK.
  • Step 1 Reaction to produce l-(2,4-Dihydroxyphenyl)-2-(4-hydroxyphenyl)ethanone
  • the solid was transferred to a 2 L Erlenmeyer flask. Isopropyl alcohol (IPA) was used to rinse the flask. The solid was recrystallized from IPA (1.4 L). The suspension was cooled in an ice bath for 30 minutes and the solid collected by vacuum filtration. The solid was rinsed with ice cold IPA until the filtrate was colorless and dried in a vacuum oven to give a white powder (162.24 g). The mother liquor and washes were combined and concentrated to orange oil (38.09 g).
  • IPA isopropyl alcohol
  • the oil bath was cooled to 90 °C and 380 mL of isopropyl alcohol was added in one portion.
  • the reaction mixture became a cloudy white suspension and redissolved to give a solution in less than a minute at 90 °C.
  • the heating to the bath was set to 50 °C and the flask was allowed to gradually cool to 50 °C.
  • a precipitate started to form at 60 °C and gave a suspension at 50 °C.
  • a thick oily mass falls out of solution ⁇ 55-53 °C.
  • Vigorous agitation with overhead stirrer 300 rpm was required to prevent the oily mass from solidifying into one solid as seen with small scale reactions equipped with stir bar. The reaction was left to stir until the mixture cooled to room temperature.
  • the mother liquor was decanted and fresh isopropanol (100 mL) was added to the flask to rinse the solid.
  • the liquid was decanted and combined with the mother liquor.
  • the mother liquor was concentrated to a dark red oil (27.13 g) and DCM (150 mL) was added to the flask to give a red solution.
  • Silica gel 55 g was added to solution and concentrated to dryness.
  • the silica gel mixture was poured into a 600 mL sintered glass funnel filled with silica gel (50 g). The solids were washed with ethyl acetate (1.2 L) and the filtrate concentrated to an orange oil (137.61 g crude).
  • the oil was dissolved into boiling 80 % IP A/water (1.2 L) and the solution allowed to cool to room temperature and stand overnight to give a cake.
  • the cake was filtered and washed with cold IPA (100 mL).
  • the mother liquor was partially concentrated on a rotovap to give a tan powder. This process was repeated until an oil could not be washed away from the powder.
  • the product was pooled and dried in a vacuum oven to give an impure tan powder (118.25 g, 85.6 %).
  • the red oil was dissolved into ethyl acetate (500 mL) and washed with saturated sodium bicarbonate solution (3 x 1 L). The organic layers was washed with brine (1 L), filtered and concentrated to give a red oil (109.32 g, crude). The oil was loaded onto 100 g of silica gel and chromatographed in 40 g portions on silica gel (100 g cartridge, 5-30 % EA/Hex). Fractions containing spots with Rf 0.55 (33 % EA/Hex) were pooled and concentrated to a light red glass (53.37 g). The glass was mixed with DCM (200 mL) and sonicated to give a pink suspension.
  • the solid was filtered through a sintered glass funnel washed with a 20 % DCM in Hexanes solution (250 mL) and dried in a vacuum oven overnight (32.41 g). The mother liquor was concentrated to a glass and the process repeated a second time to give a pink solid (4.2784 g).
  • the impure mixed fractions were pooled and concentrated to a glass (16.71 g).
  • the glass was dissolved into DCM (75 mL) and pink crystals formed on standing (7.0862 g). This process was repeated to give a second crop of pink crystals (2.3643).
  • the mother liquors from both the pure and impure fractions were combined and chromatographed with the same method (2 x 100 g cartridges). The fractions with Rf 0.55 were pooled and concentrated to give a red oil (17.388 g) which did not solidify. The oil was not combined with previous batches but reprotected in a separate reaction.
  • the reaction was diluted with DCM (200 mL), washed with saturated NaHC03 (200 mL), water (200 mL), brine (200 mL), dried over Na2S04, filtered and concentrated to give a red viscous residue.
  • the residue adsorbed onto silica gel (75 g) was purified on a silica gel column (4 x 100 g, 0 - 20 % EA/Hex) to give a white solid which was triturated with methanol and dried in a vacuum oven at 40° C for 16 h to afford the titled compound as a white powder (51.67 g 90.2 %).
  • the reaction mixture was heated in an oil bath at 120 °C. After 91 h of heating the reaction was cooled to room temperature and the mixture filtered through a pad of Celite (3 cm) which was successively washed with DCM (200 mL), EA (200 mL) and MeOH (200 mL). The filtrate was collected and concentrated. The residue was adsorbed onto silica gel (25 g) purified with silica gel (100 g cartridge, 0 - 30% MeOH/DCM) [TLC: 5 % MeOH/DCM, 4 major spots, Rf (SM:0.95), 0.9, 0.83, (prod. 0.43)]. The fractions containing product were pooled and concentrated to give a brown foam (13.64 g, 81.0 %).
  • OP- 1038 was separated into its diastereomers (2S)-3-(4-hydroxyphenyl)-4- methyl-2-(4- ⁇ 2-[(3R)-3-methylpyrrolidin-l-yl]ethoxy ⁇ phenyl)-2H-chromen-7-ol (OP-1074) and (2R)-3-(4-hydroxyphenyl)-4-methyl-2-(4- ⁇ 2-[(3R)-3-methylpyrrolidin-l-yl]ethoxy ⁇ phenyl)-2H- chromen-7-ol (OP-1075) using a Diacel, Chiralpak IC column at room temperature in isocratic mode with 80 % hexanes, 20 % 2-propanol with 0.1 % dimethylethylamine or 0.1% diethyl amine as a modifier. This method was used at analytical and preparative scale. Step 9:
  • (R)-3-methylpyrrolidine hydrochloride (20.000 g, 164.5 mmol, 1.0 equiv.) was added to a round bottom flask and dissolved into anhydrous DCM (45 mL). Freshly distilled Diisopropylethylamine (60.157 ml, 345.4 mmol, 2.1 equiv.) and freshly activated 4 A molecular sieves (- 21 g) was added to the solution and stirred for 10 minutes.
  • the mixture was filtered through a pad of Celite (4 cm). The solids were washed with DCM (5 x 500 mL). The filtrate was poured into a separatory funnel and the layers separated (-200 mL aqueous layer recovered). The aqueous layer was extracted with DCM (3 x 100 mL). The organic layers were combined and washed with brine (500 mL), dried over anhydrous sodium sulfate, filtered and concentrated to a yellow liquid (33.43 g).
  • This liquid was loaded onto silica gel (25 g) and chromatographed through silica gel (2 x 100 g cartridge) with 50-100 % ethyl acetate in hexanes followed by 10-40 % methanol in dichloromethane to give a yellow oil (29.17 g, quant).
  • the oil was taken up in methanol (50 mL) and 25 % sodium methoxide in methanol (9.9 mL, 45.5 mmol, 1 equiv) was added to the methanolic solution to give a white suspension.
  • the mixture was concentrated to dryness and taken up into anhydrous DCM (35 mL).
  • the suspension was centrifuged at 3K rpm for 5 minutes.
  • the clear solution was collected and the solid resuspended into DCM (35 mL). This process was repeated a total of 4 times.
  • the combined solution was concentrated to a yellow liquid (5.6341 g, 95.6 %).
  • OP-1083 was prepared by air oxidation on OP-1074, followed by
  • OP-1083 and OP-1074 chromatographic separation of OP-1083 and OP-1074.
  • the mixture of OP-1074 and OP-1083 (560 mg) was dissolved in methanol (15 mL) and mixed with silica gel (3 g). The mixture was dried to give a dark red powder. This powder was loaded into a cartridge and chromatographed on silica gel (4 g cartridge) with 0-25 % methanol in dichlorom ethane to give OP-1074 as an orange solid (0.261 g, 46.6 %) and OP-1083 as an orange solid (41.1 mg, 15 %)
  • Method 0 % MeOH for 4 min, gradient to 5 % MeOH/DCM over 5 minutes, hold at 5 % for 6 minutes, gradient to 10 % MeOH/DCM over 2 minutes, hold at 10 % MeOH/DCM for 8 minutes, gradient to 25 % MeOH/DCM over 0 minutes, hold at 25 % for 5 minutes.
  • TLC (5 % MeOH/DCM): 4 spots, 0.84, 0.42, 0.26 (Product), 0.16 (Mono carbonate).
  • Gradient method 0 % MeOH/DCM hold for 2 minutes, gradient to 5 % MeOH/DCM over 5 minutes, hold at 5 % MeOH/DCM for 3 minutes, gradient to 15 % MeOH/DCM over 3 minutes, hold at 15 % MeOH/DCM for 2 minutes.
  • the reaction stirred at this temperature for 1.5 h. The solution became slurry after -45 minutes. The reaction was allowed to reach room temperature over 45 min and stirred at room temperature for 2 h. A sample was quenched with water and analysis showed the mass of the desired product.
  • the reaction mixture was concentrated to dryness.
  • the crude mixture was suspended into 2 N HC1 (5 mL). The suspension was sonicated and centrifuged at 5000 rpm for 6 minutes. The supernatant was decanted and the solid resuspended into 5 mL of 2 N HC1 and the process was repeated. The solid was dried under vacuum to give 129 mg of the crude product.
  • MCF-7 Proliferation in MCF-7 was measured using a fluorescent DNA binding dye 6-8 days after treatment in triplicate with anti-estrogens in the presence of 100 pM E2. Specifically, MCF-7 cells were treated with anti-estrogens alone or in combination with other
  • chemotherapeutics in hormone-depleted medium for 6-8 days in the presence of 100 pM E2. Proliferation was measured using Cyquant fluorescent DNA-binding dye (Invitrogen, Grand Island, NY). Results were from a single representative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing SEM.
  • Fig. 1 A plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or ⁇ OP-1074 in combination with increasing amounts of PD-0332991.
  • IB plots the effects on MCF-7 cell proliferation of vehicle or ⁇ PD-0332991 in combination with increasing amounts of OP-1074.
  • Fig. 1C plots the effects on MCF-7 cell proliferation of vehicle or 3nM OP-1074, tamoxifen, or fulvestrant in combination with increasing amounts of PD- 0332991.
  • the combination of OP-1074 with PD- 0332991 at several concentrations leads to a lower percentage of proliferation of MCF-7 breast cells than either agent by themselves.
  • OP-1074 increases the potency of the mTOR inhibitor everolimus in inhibiting E2-stimulated MCF-7 breast cell proliferation.
  • the antiestrogen OP- 1074 enhances the efficacy and potency of the mTOR inhibitor everolimus in inhibiting E2- stimulated breast cell proliferation in vitro.
  • MCF-7 cells were treated with the indicated amount of OP-1074 and everolimus in hormone-depleted medium for 6 days in the presence of 100 pM 17P-estradiol (E2).
  • Fig. 2 A plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or ⁇ OP-1074 in combination with increasing amounts of everolimus.
  • FIG. 2B plots the effects on MCF-7 cell proliferation of vehicle, InM or lOnM everolimus in combination with increasing amounts of OP-1074.
  • FIG. 2C plots the effects on MCF-7 cell proliferation of vehicle or 3nM OP- 1074, tamoxifen, or fulvestrant in combination with increasing amounts of everolimus.
  • OP-1074 with everolimus at several concentrations leads to a lower percentage of proliferation of MCF-7 breast cells than either agent by themselves.
  • OP-1074 increases the potency of the pan PI3K inhibitor
  • Fig. 3 A plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or ⁇ OP-1074 in combination with increasing amounts of BKM-120.
  • Fig. 3B plots the effects on MCF-7 cell proliferation of vehicle, 32nM or ⁇ BKM-120 in combination with increasing amounts of OP-1074.
  • OP-1074 increases the potency of 5-fluorouracil in inhibiting E2-stimulated MCF-7 breast cell proliferation.
  • the antiestrogen OP-1074 enhances the efficacy and potency of the thymidylate synthase inhibitor 5-fluorouracil in inhibiting E2- stimulated breast cell proliferation in vitro.
  • MCF-7 cells were treated with the indicated amount of OP-1074 and 5-fluorouracil in hormone-depleted medium for 6 days in the presence of 100 pM 17p-estradiol (E2).
  • E2 100 pM 17p-estradiol
  • OP-1074 increases the potency of taxanes in inhibiting El- stimulated MCF-7 breast cell proliferation.
  • the antiestrogen OP-1074 enhances the efficacy and potency of paclitaxel and docetaxel in inhibiting E2-stimulated breast cell proliferation in vitro.
  • MCF-7 cells were treated with the indicated amount of OP-1074 and taxane in hormone-depleted medium for 6 days in the presence of 100 pM 17P-estradiol (E2).
  • Fig. 5 A plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or ⁇ OP-1074 in combination with increasing amounts of paclitaxel.
  • OP-1074 increases the potency of the anthracycline doxorubicin in inhibiting E2-stimulated MCF-7 breast cell proliferation.
  • the antiestrogen OP- 1074 enhances the efficacy and potency of the anthracycline doxorubicin in inhibiting El- stimulated breast cell proliferation in vitro.
  • MCF-7 cells were treated with the indicated amount of OP-1074 and Doxorubicin in hormone-depleted medium for 7 days in the presence of 100 pM 17p-estradiol (E2).
  • E2 100 pM 17p-estradiol
  • Compounds described herein can comprise one or more asymmetric centers, and thus can exist in various isomeric forms, e.g., enantiomers and/or diastereomers.
  • the compounds described herein can be in the form of an individual enantiomer, diastereomer or geometric isomer, or can be in the form of a mixture of stereoisomers, including racemic mixtures and mixtures enriched in one or more stereoisomer.
  • Isomers can be isolated from mixtures by methods known to those skilled in the art, including chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of chiral salts; or preferred isomers can be prepared by asymmetric syntheses.
  • HPLC high pressure liquid chromatography
  • analogue means one analogue or more than one analogue.
  • “Pharmaceutically acceptable salt” refers to a salt of a compound of the invention that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound.
  • such salts are non-toxic may be inorganic or organic acid addition salts and base addition salts.
  • such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethane-disulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid
  • pharmaceutically acceptable cation refers to an acceptable cationic counter-ion of an acidic functional group. Such cations are exemplified by sodium, potassium, calcium, magnesium, ammonium, tetraalkyl ammonium cations, and the like (see, e.g., Berge, et al., J. Pharm. Sci. 66(1): 1-79 (Jan.'77).
  • “Pharmaceutically acceptable vehicle” refers to a diluent, adjuvant, excipient or carrier with which a compound of the invention is administered.
  • “Pharmaceutically acceptable metabolically cleavable group” refers to a group which is cleaved in vivo to yield the parent molecule indicated herein.
  • Prodrugs refers to compounds, including derivatives of the compounds of the invention, which have cleavable groups and become by solvolysis or under physiological conditions a compound of the invention that are pharmaceutically active in vivo.
  • Solvate refers to forms of the compound that are associated with a solvent or water (also referred to as "hydrate”), usually by a solvolysis reaction. This physical association includes hydrogen bonding.
  • solvents include water, ethanol, acetic acid and the like.
  • the compounds of the invention may be prepared e.g. in crystalline or liquid form and may be solvated or hydrated.
  • Suitable solvates include pharmaceutically acceptable solvates, such as hydrates, and further include both stoichiometric solvates and non-stoichiometric solvates. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid.
  • “Solvate” encompasses both solution-phase and isolable solvates. Representative solvates include hydrates, ethanolates and methanolates.
  • a "subject" to which administration is contemplated includes, but is not limited to, humans ⁇ i.e., a male or female of any age group, e.g., a pediatric subject (e.g, infant, child, adolescent) or adult subject (e.g., young adult, middle-aged adult or senior adult)) and/or a non- human animal, e.g., a mammal such as primates (e.g., cynomolgus monkeys, rhesus monkeys), cattle, pigs, horses, sheep, goats, rodents, cats, and/or dogs.
  • the subject is a human.
  • the subject is a non-human animal.
  • the terms "human", “patient” and “subject” are used interchangeably herein.
  • the term “enantiomerically pure” or “pure enantiomer” denotes that the compound comprises more than 95% by weight.
  • the term may refer to more than 96% by weight, more than 97% by weight, more than 98%) by weight, more than 98.5%> by weight, more than 99% by weight, more than 99.2% by weight, more than 99.5% by weight, more than 99.6% by weight, more than 99.7% by weight, more than 99.8% by weight or more than 99.9% by weight, of the enantiomer.
  • the weights are based upon total weight of all enantiomers or stereoisomers of the compound.
  • the term “enantiomerically pure R- compound” refers to at least 95% by weight R-compound and at most about 5% by weight S- compound. In alternative embodiments, when specified, the term can refer to at least about 99% by weight R-compound and at most about 1% by weight S-compound or at least about 99.9% by weight R-compound or at most about 0.1% by weight S-compound. In certain embodiments, the weights are based upon total weight of compound.
  • the term “enantiomerically pure S- compound” or “S-compound” refers to at least about 95% by weight S-compound and at most about 5% by weight R-compound. In alternative embodiments, when specified, the term can refer to at least about 99% by weight S-compound and at most about 1% by weight R-compound or at least about 99.9% by weight S-compound and at most about 0.1% by weight R-compound. In certain embodiments, the weights are based upon total weight of compound.

Abstract

This invention relates to a method of treating a disorder modulated, mediated or affected by the estrogen receptor in a subject comprising administering to the subject a specific benzopyran (in the form of a mixture of S-C2 and R-C2 diastereomers or its pure S-diastereomer) in combination with an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic.

Description

COMBINATIONS OF BENZOPYRAN COMPOUNDS,
COMPOSITIONS AND USES THEREOF
FIELD
[0001] This invention is in the field of pharmaceuticals, and in particular, this invention is related to the treatment of cancers mediated by the estrogen receptor by administering a specific benzopyran (in the form of a mixture of S-C2 and R-C2 diastereomers or its pure S-diastereomer) as described in US 2013/0178445 and WO 2013/090921 in combination with particular agents, active agents, anti-cancer agents or chemotherapeutic agents.
BACKGROUND
[0002] Estrogen receptor modulators are a class of compounds that act on the estrogen receptor. These compounds can be pure agonists (mimicking estrogen), pure antagonists, or mixed agonist-antagonists (sometimes referred to as Selective Estrogen Receptor Modulators (SERMs)). For example, estradiol (A) is a pure agonist, fulvestrant (B) is a complete antagonist, and tamoxifen (C) and raloxifene (D) are SERMs.
[0003] Most breast cancers express estrogen receptors (ER), and their growth is driven by the action of estrogen at its receptors, primarily at ER alpha. This type of cancer is treated with an estrogen antagonist, which competes with estrogen for binding to the receptor, but does not activate it, preventing estrogen driven growth. Partial anti-estrogens like raloxifene and tamoxifen retain some estrogen-like effects, including an estrogen-like stimulation of uterine growth, and also, in some cases, an estrogen-like action during breast cancer progression which actually stimulates tumor growth. In contrast, fulvestrant, a complete anti-estrogen, is free of estrogen-like action on the uterus and is effective in tamoxifen-resistant tumors. A recent study also suggests that fulvestrant is substantially superior to the aromatase inhibitor anastrozole in treating metastatic breast cancer (Robertson et al. J Clin Oncol (2009) 27(27):4530-5).
[0004] Estradiol is a naturally-occuring female estrogenic hormone. Raloxifene was disclosed by Eli Lilly in 1981 (U.S. Patent No. 4,418,068; 5,478,847; 5,393,763; and 5,457,117) for prevention of breast cancer and treatment of osteoporosis. Fulvestrant was disclosed by Imperial Chemical Industries (ICI) in 1983 (U.S. Patent No. 4,659,516, expired in 2007 with a patent term extension; U.S. Patent Nos. 6,774, 122 and 7,456, 160). Tamoxifen was also disclosed by ICI in the '516 patent. Tamoxifen was developed for the treatment of breast cancer on the basis of strong antagonism of estrogen action in mammary tissue (Jordan, J. Cell. Biochem. 51 (1995)).
Figure imgf000004_0001
tamoxifen raloxifene
C D
[0005] The degree of anti-estrogenicity is often assayed by exposing female, immature
(preferably ovariectomized) rodents to test doses of the compound both in the absence (agonist mode) and presence (antagonist mode) of estrogen. Tamoxifen and other partial anti-estrogens stimulate uterine weight gain in the agonist mode and only partly block estrogen-driven uterine weight gain in the antagonist mode. Fulvestrant and other complete anti-estrogens do not stimulate uterine weight gain in the agonist mode and completely block estrogen-driven weight gain in the antagonist mode. The induction of estrogen-regulated alkaline phosphatase expression in human uterine cancer cell growth in culture can be used to distinguish partial and complete anti-estrogenicity and correlates well with the rodent weight gain assay. [0006] Tamoxifen and fulvestrant both inhibit cultured human breast cancer cell proliferation provoked by estrogen. However, fulvestrant more fully inhibits the proliferation when provoked with growth factors, especially of the insulin/insulin-like growth factor family. Thus the inhibition of growth-factor driven breast cancer cell proliferation and the effect on uterine weight provide two assays which can distinguish between complete and partial anti- estrogens.
[0007] Tamoxifen binding stabilizes the estrogen receptor whereas fulvestrant and chemically related antiestrogens, such as ICI-164384 and RU-58668, cause degradation of the estrogen receptor. (Dodge et al, J. Bone Miner. Res., 8 (Suppl 1, S278 (1993); Wakeling, Breast Cancer Res. Treat. 25, 1 (1993); Baer et al, Calcified Tissue Int., 55, 338 (1994). However, some compounds, like GW-5638 (Wu et al, Mol Cell.,18,413 (2005), and OP1075, described below, degrade the receptor but are partial estrogens- that is, not complete anti-estrogens. Thus the ability to degrade the estrogen receptor does not ensure complete antiestrogenicity. The ability to induce degradation of the receptor is nonetheless a factor that differentiates the behavior of tamoxifen and fulvestrant and may be desirable in a drug to treat breast cancer.
[0008] Fulvestrant, which degrades the estrogen receptor, incorporates a core of 17-beta estradiol. It has a long flexible aliphatic side chain that blocks oral absorption. The estradiol core blocks oral absorption and the long flexible aliphatic side chain makes the drug very insoluble which worsens the problem. Fulvestrant must be injected because of the poor oral bioavailability. Two 5 ml intramuscular depot injections, one into each buttock, must be administered monthly by a health professional. Furthermore, it is unclear whether these two injections provide sufficient drug exposure for optimal action. The drug does not seem to work in pre-menopausal women.
[0009] In 1990, an important step in oral anti-estrogen development came with the discovery of a family of high-affinity benzopyran anti-estrogens by Kapil and coworkers.
(Sharma et al. (1990) J Med Chem, 33(12):3222-9; Sharma et al. (1990) J Med Chem,
33(12):3216-22). The numbering scheme of benzopyrans is typically:
Figure imgf000005_0001
F
[0010] Sharma et al. showed that the combination of 7-hydroxyl and 4' -hydroxyl groups conferred high-affinity binding of the benzopyran core to the estrogen receptor (Compound G; see Compound 25 of Sharma et -9 where R1 and R2 are OH).
Figure imgf000006_0001
G
Further, Sharma et al. reported that the presence of a methyl group at the 4 position of the benzopyran core enhanced receptor binding affinity, without a hydroxyl group at the 4'-position.
[0011] In 1991, Labrie and coworkers filed a patent application which issued as U.S.
Patent No. 5,395,842 (see claim 29) which taught that EM-343 (H), showed superior binding to the estrogen receptor with no loss of anti-estrogen action. EM-343 differed from the Saeed compounds by including the hydroxyl at the 4'-position of a 4-methyl, 7-hydroxyl benzopyran.
Figure imgf000006_0002
EM-343
H
[0012] In 1995, Labrie et al. filed a continuation-in-part patent application, which issued in 2000 as U.S. Patent No. 6,060,503, disclosing prodrugs and optically active species of EM- 343. Particularly, Labrie et al. disclosed a pure isomer of EM-343, EM-652, referred to as acolbifene (I), which is (S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-(2-(piperidin-l- yl)ethoxy)phenyl)-2H-chromen-7-ol.
Figure imgf000007_0001
I
[0013] Labrie et al. in WO 01/54699 (see Figure 4a and 4b) also presented several broad generic Markush formulae of benzopyran-containing compounds, including acolbifene analogs, in which the side chain terminates in various substituted ring systems including pyrrolidinyl, piperidinyl, and methyl- 1 -pyrrolidinyl and dimethyl- 1 -pyrrolidinyl.
[0014] U.S. Patent Nos. 7,005,428 and 6,465,445 to Labrie, which claim priority to a
June 1998 application describe the following generic formulas for use as anti-estrogenic compounds:
Figure imgf000007_0002
wherein D is -OCH2CH2N(R3)R4 (R3 and R4 either being independently selected from the group consisting of C1-C4 alkyl, or R3, R4 and the nitrogen atom to which they are bound together being a ring structure selected from the group consisting of pyrrolidino, dimethyl- 1-pyrrolidino, methyl-l-purrolidinyl, piperidino, hexamethyleneimino and morpholino); and
wherein Ri and R2 are independently selected from the group consisting of hydrogen, hydroxyl and a moiety converted in vivo in to hydroxyl, and
Figure imgf000008_0001
wherein Ri and R2 are independently selected from the group consisting of hydroxyl and a moiety converted in vivo in to hydroxyl;
wherein R3 is a species selected from the group consisting of saturated, unsaturated or substituted pyrrolidinyl, saturated, unsaturated or substituted piperidino, saturated, unsaturated or substituted piperidinyl, saturated, unsaturated or substituted morpholino, nitrogen-containing cyclic moiety, nitrogen-containing polycyclic moiety, and RaRb (Ra and Rb being independently hydrogen, straight or branched Ci-C6 alkyl, straight or branched C2-C6 alkenyl, and straight of branched C2-C6 alkynyl.
[0015] Acolbifene binds to the estrogen receptor alpha with three times the affinity of 17- beta estradiol, the native ligand (Katzenellenbogen (2011) J Med Chem 54(15):5271-82). Since anti-estrogens must compete with estradiol for binding to the estrogen receptor, high affinity binding is an important drug virtue. Both the Labrie '842 and the Labrie '503 patents disclosed benzopyran compounds that can contain an unsubstituted pyrrolidine in the "tail" or R3 position as depicted in Compound F. EM-800, a pivalate prodrug of EM-652, and HC1 salts of EM-652 were also described in the '503 patent.
[0016] Acolbifene was initially thought to be a complete anti-estrogen. However, careful studies with the rodent uterine assay and human uterine cell alkaline phosphatase assays revealed that it retained some estrogen-like action, about 12% that of estradiol (Labrie et al. "The combination of a novel selective estrogen receptor modulator with an estrogen protects the mammary gland and uterus in a rodent model: the future of postmenopausal women's health?" Endocrinology. 2003 144(11):4700-6). This contrasts with fulvestrant where the residual estrogen-like action is almost unmeasurable. Furthermore, fulvestrant binding induces dramatic degradation of the estrogen receptor, while acolbifene induces either no or modest receptor degradation. Raloxifene and bazedoxifene don't degrade the receptor, but stabilize the receptor to a much lesser degree than tamoxifen. [0017] Acolbifene is orally bioavailable and is currently being positioned for Phase III clinical trials for the treatment of breast cancer by the Canadian company Endoceutics (Founded by Dr. Labrie). A daily oral dose of 40 mg of acolbifene or EM800 in women produces mean drug exposures of 8.3 and 15 ng/ml of circulating acolbifene, respectively. In preclinical studies both forms of the drug are effective against tamoxifen-resistant human breast cancer xenografts growing on immunocompromised mice. In clinical studies the 40mg dose of EM800 was numerically as effective as anastrozole in preventing progression of metastatic estrogen receptor positive breast cancer.
[0018] Starting in 2005, Blizzard and coworkers at Merck published a series of papers on estrogen receptor ligands. They first focused on using a dihydrobenzoxathiin core (J) with alkyl substituted pyrrolidine side chains and linkers as SERAMs (Selective Estrogen Receptor Alpha Modulators) (Blizzard et al. (2005) Bioorg Med Chem Lett. 15(1): 107-13).
Figure imgf000009_0001
Merck Dihydrobenzoxathiin Core
J
[0019] The group tried to maximize the estrogen receptor α/β selectivity ratio and minimize uterine activity (e.g., maximize antagonism of uterine activity). They reported that the unbranched linker with 3 -methyl pyrrolidinyl and 3,4-methyl pyrrolidinyl as well as the a-m ethyl (i.e., a methyl on the a-position of the ethylene) linker with an unsubstituted pyrrolidinyl side chains were noteworthy. Blizzard et al. concluded that minor modifications in the side chain or linker resulted in significant effects on biological activity, especially in uterine tissue.
[0020] Blizzard et al. also studied a chromane core (Blizzard et al. (2005) Bioorg Med
Chem Lett. 15(6): 1675-81) (Compound K).
Figure imgf000010_0001
Merck Chromane Core
K
[0021] The Merck chromane core differs from the acolbifene core by the absence of a double bond in the oxane ring. These structures also had a hydroxyl at position 6 (not 7) of the fused benzene ring. A chromane core with a 2-m ethyl pyrrolidine (but not a 3 -methyl) with a methyl on the linker created a nearly complete anti-estrogen, (see compound 12 of the Blizzard et al. paper). Blizzard et al. commented on the differences among anti-estrogenic activities of variously substituted cores, and noted that the size and stereogenic placement of substituents is crucial for receptor potency and selectivity.
[0022] In the third publication of this series (Blizzard et al. (2005) Bioorg Med Chem
Lett. 15(17):3912-6); Blizzard et al. again studied the dihydrobenzoxathiin core and reported that their studies have resulted in the discovery that addition of a methyl group to the side chain at the appropriate position and with the right stereochemistry, either on the pyrrolidine ring or on the linker substantially increased estrogen antagonist activity in uterine tissue. Blizzard et al. also reported that the best estrogen antagonist activity in this dihydrobenzoxathiin series was determined to have a methyl group on the pyrrolidine and a methyl group on the linker, with the hydroxyl in the 6-position of the fused benzene ring. Blizzard et al. also noted that to their knowledge, their optimized side chain with two methyl groups represented the first example where a relatively small structural modification of an exisiting SERM resulted in a conversion of a SERM to a SERAM/SERD (Selective Estrogen Receptor alpha Modulator and Down- regulator).
[0023] The Merck team then investigated whether the optimized side chain modification reported for the dihyrobenzoxathiin core was "portable" and could confer strong anti- estrogenicity when appended to different cores (Blizzard et al. (2005) Bioorg Med Chem Lett. 15(23):5214-8). Merck demonstrated that none of the three cores tested (raloxifene,
bazedoxifene, or lasofoxifene) became more anti-estrogenic with either the 3-methyl pyrrolidine or the chiral side chain modifications. Blizzard et al. concluded that "The lack of a dramatic effect on the uterine profile upon incorporation of side chains A and B clearly indicates that the side chain Structure Activity Relationship of the dihydrobenzoxathiin SERAMs is not transferable to other platforms."
[0024] In yet another 2005 research publication, Gauthier, Labrie and colleagues reported the synthesis and structure-activity relationships of analogs of acolbifene (Gauthier et al. (2005) J Enzyme Inhib Med Chem, 20(2): 165-77). They attempted to improve on the anti-estrogenicity of acolbifene by creating analogs in which the terminal piperidine was either replaced by pyrrolidine or substituted in various ways. All of these analogs proved to be more estrogenic than acolbifene as revealed by the rodent uterus assay. This experience suggests that improvement of the anti-estrogenicity of acolbifene will be a challenge and modifications provide unpredictable results.
[0025] Blizzard reviewed the Merck research on anti-estrogens in 2008 (Curr Top Med
Chem. 8(9):792-812). He noted that:
"Selective Estrogen Receptor Modulators (SERMs) have been the subject of extensive medicinal chemistry efforts at several pharmaceutical companies, including
Merck The Merck SERM project involved a large number of talented and dedicated chemists and biologists who worked for several years to discover novel classes of SERMs with a range of selectivities....no drugs have yet reached the market as a result of this effort."
[0026] Indeed, the Merck effort began in the early 1990's and continued well into the
2000' s, reflecting impressive science but no commercial products. Their most promising compounds, which included side chains in which the piperidine ring was replaced with a mono- or di-substituted pyrrolidine ring appended to a benzoxathiin core, especially with a 3-R methyl pyrrolidine terminus, showed anti-estrogenicity, although not as complete as fulvestrant in the rodent uterus assay. A chiral methyl on atom 2 of the flexible linker also conferred improved anti-estrogenicity. The two features together in a doubly substituted side chain conferred anti- estrogenicity that was similar to fulvestrant. Unfortunately the Merck core had problematic reactive metabolites when investigated in primates, which halted clinical development.
[0027] Aragon Pharmaceuticals filed PCT/US2011/039669 (published December 15,
2011 as WO2011/156518), which claimed priority to U.S. Provisional Application 61/353,531 titled "Estrogen Receptor Modulators and Uses Thereof filed in June 2010. Aragon disclosed large genuses of benzopyran derivatives and at least 71 acolbifene analogs intended for treatment of tamoxifen resistant breast cancer. Aragon appears to have taken the prior art teachings of Merck regarding how to optimize the dihydrobenzoxathiin core, and applied the teachings to the acolbifene benzopyran core. Aragon is considering advancing a drug to the clinic for late stage progressive metastatic disease.
[0028] Kushner et al. (US 2013/0178445 and WO 2013/090921, both filed Dec. 17, 2012 and both assigned to Olema Pharmaceuticals) describe OP-1038 (3-(4-hydroxyphenyl)-4-methyl- 2-(4-{2-[(3R)-3-methylpyrrolidin-l-yl]ethoxy}phenyl)-2H-chromen-7-ol) and OP-1074 ((2S)-3- (4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3-methylpyrrolidin-l-yl]ethoxy}phenyl)-2H- chromen-7-ol), as well as pharmaceutical compositions and methods of use.
[0029] Bazedoxifene is a SERM, under development for prevention and treatment of postmenopausal osteoporosis (Biskobing, D. M. (2007) Clinical interventions in aging 2 (3): 299-303). Lasofoxifene is another SERM under development for the treatment of
postmenopausal osteoporosis and vaginal atrophy (Gennari et al. (2006), Expert Opin Investig Drugs 15 (9): 1091-103).
[0030] U.S. Patent 5,254,568 discloses benzopyrans as anti-estrogenic agents.
[0031] WO2010/145010 discloses a combination of SERM and sex steroid precursor for treating hot flashes and other symptoms.
[0032] WO2004/091488 discloses benzopyrans as estrogen receptor modulators.
[0033] U.S. Patent 5,840,735 discloses benzopyrans as sex steroid activity inhibitors.
[0034] U.S. Patent 6,262,270 discloses a method for the enantiomeric synthesis of acolbifene derivatives.
[0035] Thus, there is a need for new combinations and pharmaceutical compositions for the treatment of medical disorders that are mediated or affected by an estrogen receptor.
SUMMARY OF THE INVENTION
[0036] Each of the embodiments described below can be combined with any other embodiment described herein not inconsistent with the embodiment with which it is combined. The phrase "or a pharmaceutically acceptable salt thereof is implicit in the description of all compounds described herein; however, in one aspect of any of the embodiments herein, the compound is in the form of a free base.
[0037] Embodiments described herein relate to a method of treating a disorder modulated, mediated or affected by the estrogen receptor in a subject comprising administering to the subject a compound, which is
Figure imgf000013_0001
OP-1038
Figure imgf000013_0002
Figure imgf000013_0003
OP-1074 or
Figure imgf000013_0004
wherein Ri and R2 are independently either:
(i) OH or OR9,
(ii) wherein R9 is independently selected from H, halogen (CI, Br, I or F), natural or non- naturally occurring amino acid (bound through either the OC(O)- or C(0)0- (an ester) or the amino (through either -C(0)-N- or -N-C(O)- (an amide linkage)), R10, -OR10, or -SR1( where R10 is -C(=0)RC1, -C(=0)0RC1, -C(=0)SRC1, -C(=0)N(RC1)2; or polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;
-S(=0)2RC1, -S(=0)2ORcl, -S(=0)RC1, -S(=0)ORcl, -P(=0)2RC1, -P(=0)2ORcl, -P(=0)(ORcl)2, -P(=0)(RC1)2, or -P(RC1)(0RC1); or oxygen attached to an oxygen protecting group (to produce OH on administration), sulfur attached to a sulfur protecting group (to produce SH or a disulfide on administration), or nitrogen attached to a nitrogen protecting group (to produce - H- on administration);
and R can be independently selected from hydrogen, polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or two R groups are joined to form an substituted or unsubstituted heterocyclic ring;
or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, stereoisomer, tautomer, or N- oxide thereof,
in combination with an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic.
[0038] In embodiments of the method of the invention, the compound is in the form of a pharmaceutically acceptable salt.
[0039] In embodiments of the method of the invention, either or both of Rl or R2 is an ester, amide, carbonate or phosphate.
[0040] In embodiments of the method of the invention, compound is in the form of a prodrug, further wherein the prodrug is
Figure imgf000014_0001
OP-1088
Figure imgf000015_0001
Figure imgf000015_0002
Figure imgf000015_0003
OP-1084
Figure imgf000015_0004
OP-1085
Figure imgf000015_0005
OP-1083
Figure imgf000016_0001
OP-1087
[0041] In embodiments of the method of the invention, the compound is (2S)-3-(4- hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3 -methyl pyrrolidin-1 -yl]ethoxy}phenyl)-2H-chromen- 7-ol, which has the chemical structure:
OP-1074
or a pharmaceutically acceptable salt, solvate, hydrate, or N-oxide thereof.
[0042] Embodiments described herein relate to a method of treating a disorder modulated, mediated or affected by the estrogen receptor in a subject comprising administering to the subject a compound, which is (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3- methylpyrrolidin-l-yl]ethoxy}phenyl)-2H-chromen-7-ol, which has the chemical structure:
Figure imgf000016_0003
OP-1074
or a pharmaceutically acceptable salt thereof,
in combination with an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic. [0043] In embodiments of the method of the invention, the compound is administered in alternation with an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic.
[0044] Embodiments described herein relate to a combination of a compound, which is
Figure imgf000017_0001
wherein Ri and R2 are independently either:
(i) OH or OR9,
(ii) wherein R9 is independently selected from H, halogen (CI, Br, I or F), natural or non- naturally occurring amino acid (bound through either the OC(O)- or C(0)0- (an ester) or the amino (through either -C(0)-N- or -N-C(O)- (an amide linkage)), R10, -OR10, or -SR1(
and R can be independently selected from hydrogen, polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or two R groups are joined to form an substituted or unsubstituted heterocyclic ring;
or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, stereoisomer, tautomer, or N- oxide thereof,
and an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic, for the treatment of a disorder modulated, mediated or affected by the estrogen receptor.
[0045] In embodiments of the combination of the invention, the compound is in the form of a pharmaceutically acceptable salt.
[0046] In embodiments of the combination of the invention, the compound is (2S)-3-(4- hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3 -methyl pyrrolidin-1 -yl]ethoxy}phenyl)-2H-chromen- 7-ol, which has the chemical structure:
Figure imgf000018_0001
OP-1074
harmaceutically acceptable salt, solvate, hydrate, or N-oxide thereof. [0047] Embodiments described herein relate to a combination of (2S)-3-(4- hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3 -methyl pyrrolidin-1 -yl]ethoxy}phenyl)-2H- 7-ol, which has the chemical structure:
Figure imgf000019_0001
OP-1074
or a pharmaceutically acceptable salt thereof,
and an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic for the treatment of a disorder modulated, mediated or affected by the estrogen receptor.
[0048] In embodiments of the method and combination of the invention, the disorder is mediated by the estrogen receptor.
[0049] In embodiments of the method and combination of the invention, the disorder is breast cancer.
[0050] In embodiments of the method and combination of the invention, the breast cancer is local, advanced or metastatic estrogen or progesterone receptor positive advanced breast cancer.
[0051] In embodiments of the method and combination of the invention, the breast cancer is estrogen or progesterone receptor positive advanced breast cancer.
[0052] In embodiments of the method and combination of the invention, the breast cancer is estrogen or progesterone receptor negative breast cancer.
[0053] In embodiments of the method and combination of the invention, the disorder is selected from the group consisting of ovarian cancer, endometrial cancer, vaginal cancer, endometriosis, lung cancer, and bronchial cancer.
[0054] In embodiments of the method and combination of the invention, the disorder is selected from the group consisting of ovarian, endometrial, and vaginal cancer and
endometriosis.
[0055] In embodiments of the method and combination of the invention, the disorder is retinoblastoma positive breast cancer. [0056] In embodiments of the method and combination of the invention, the disorder is selected from the group consisting of retinoblastoma positive endometrial, vaginal and ovarian cancers and lung and bronchial cancers.
[0057] In embodiments of the method and combination of the invention, the disorder is lung cancer or bronchial cancer.
[0058] In embodiments of the method and combination of the invention, the subject is a patient.
[0059] In embodiments of the method and combination of the invention, the agent is a
CDK 4/6 inhibitor.
[0060] In embodiments of the method and combination of the invention, the CDK 4/6 inhibitor is PD-0332991, LY2835219, or LEE011.
[0061] In embodiments of the method and combination of the invention, the CDK 4/6 inhibitor is PD-0332991, or a pharmaceutically acceptable salt thereof.
[0062] In embodiments of the method and combination of the invention, the CDK 4/6 inhibitor is PD-0332991.
[0063] In embodiments of the method and combination of the invention, the agent is an mTOR inhibitor.
[0064] In embodiments of the method and combination of the invention, the mTOR inhibitor is rapamycin, everolimus, temsirolimus, AP23573, AZD8055, WYE-354, WYE-600, WYE-687, or Ppl21.
[0065] In embodiments of the method and combination of the invention, the mTOR inhibitor is everolimus.
[0066] In embodiments of the method and combination of the invention, the agent is a
PI3 Kinase inhibitor.
[0067] In embodiments of the method and combination of the invention, the PI3 Kinase inhibitor is BKM-120, XL-147, RG-7321, CH-5132799 and BAY-80-6946.
[0068] In embodiments of the method and combination of the invention, the PI3 Kinase inhibitor is BKM-120.
[0069] In embodiments of the method and combination of the invention, the agent is a taxane.
[0070] In embodiments of the method and combination of the invention, the taxane is paclitaxel or docetaxel. [0071] In embodiments of the method and combination of the invention, the agent is an antimetabolite.
[0072] In embodiments of the method of the invention, the antimetabolite is 5- fluorouracil.
[0073] In embodiments of the method and combination of the invention, the agent is an antitumor antiobiotic.
[0074] In embodiments of the method and combination of the invention, the antitumor antibiotic is doxorubicin.
[0075] Embodiments described herein relate to a pharmaceutical composition comprising a pharmaceutically effective amount of a compound, which is
Figure imgf000021_0001
OP-1074
Figure imgf000022_0001
wherein Ri and R2 are independently either:
(i) OH or OR9,
(ii) wherein R9 is independently selected from H, halogen (CI, Br, I or F), natural or non- naturally occurring amino acid (bound through either the OC(O)- or C(0)0- (an ester) or the amino (through either -C(0)-N- or -N-C(O)- (an amide linkage)), R10, -OR10, or -SR10 where R10 is -C(=0)RC1, -C(=0)0RC1, -C(=0)SRC1, -C(=0)N(RC1)2; or polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;
-S(=0)2RC1, -S(=0)2ORcl, -S(=0)RC1, -S(=0)ORcl, -P(=0)2RC1, -P(=0)2ORcl, -P(=0)(ORcl)2, -P(=0)(RC1)2, or -P(RC1)(0RC1); or oxygen attached to an oxygen protecting group (to produce OH on administration), sulfur attached to a sulfur protecting group (to produce SH or a disulfide on administration), or nitrogen attached to a nitrogen protecting group (to produce - H- on administration);
and RC1 can be independently selected from hydrogen, polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or two R groups are joined to form an substituted or unsubstituted heterocyclic ring;
or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, stereoisomer, tautomer, or N- oxide thereof,
in combination with an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic, and a pharmaceutically acceptable carrier.
[0076] In embodiments of the pharmaceutical composition of the invention, the compound is in the form of a pharmaceutically acceptable salt. [0077] In embodiments of the pharmaceutical composition of the invention, Ri or ester, amide, carbonate or phosphate.
[0078] In embodiments of the pharmaceutical composition of the invention, the compound is in the form o
Figure imgf000023_0001
OP-1084
Figure imgf000024_0001
Figure imgf000024_0002
Figure imgf000024_0003
OP-1087
[0079] In embodiments of the pharmaceutical composition of the invention, the compound is (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3-methylpyrrolidin-l- yl]ethoxy}phenyl)-2H-chromen-7-ol, which has the chemical structure:
Figure imgf000024_0004
OP-1074
or a pharmaceutically acceptable salt, solvate, hydrate, or N-oxide thereof.
[0080] Embodiments described herein relate to a pharmaceutical composition of the invention relates to a compound, which is (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3- methylpyrrolidin-l-yl]ethoxy}phenyl)-2H-chromen-7-ol, which has the chemical structure:
Figure imgf000025_0001
OP-1074
or a pharmaceutically acceptable salt thereof,
in combination with an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic, and a pharmaceutically acceptable carrier.
[0081] In embodiments of the pharmaceutical composition of the invention, the agent is a
CDK 4/6 inhibitor.
[0082] In embodiments of the pharmaceutical composition of the invention, the CDK 4/6 inhibitor is PD-0332991, LY2835219, or LEE011.
[0083] In embodiments of the pharmaceutical composition of the invention, the CDK 4/6 inhibitor is PD-0332991, or a pharmaceutically acceptable salt thereof.
[0084] In embodiments of the pharmaceutical composition of the invention, the CDK 4/6 inhibitor is PD-0332991.
[0085] In embodiments of the pharmaceutical composition of the invention, the agent is an mTOR inhibitor.
[0086] In embodiments of the pharmaceutical composition of the invention, the mTOR inhibitor is rapamycin, everolimus, temsirolimus, AP23573, AZD8055, WYE-354, WYE-600, WYE-687, or Ppl21.
[0087] In embodiments of the pharmaceutical composition of the invention, the mTOR inhibitor is everolimus.
[0088] In embodiments of the pharmaceutical composition of the invention, the agent is a
PI3 Kinase inhibitor.
[0089] In embodiments of the pharmaceutical composition of the invention, the PI3
Kinase inhibitor is BKM-120, XL-147, RG-7321, CH-5132799 and BAY-80-6946.
[0090] In embodiments of the pharmaceutical composition of the invention, the PI3
Kinase inhibitor is BKM-120.
[0091] In embodiments of the pharmaceutical composition of the invention, the agent is a taxane. [0092] In embodiments of the pharmaceutical composition of the invention, the taxane is paclitaxel or docetaxel.
[0093] In embodiments of the pharmaceutical composition of the invention, the agent is an antimetabolite.
[0094] In embodiments of the pharmaceutical composition of the invention, the antimetabolite is 5-fluorouracil.
[0095] In embodiments of the pharmaceutical composition of the invention, the agent is an antitumor antiobiotic.
[0096] In embodiments of the pharmaceutical composition of the invention, the antitumor antibiotic is doxorubicin.
[0097] The present invention relates to the treatment of tumors, including cancers, mediated by the estrogen receptor with the combination of a specific benzopyran (in the form of a mixture of S-C2 and R-C2 diastereomers and also its pure S-diastereomer) as described in US 2013/0178445 and WO 2013/090921 in combination or alternation with specific agents, active agents, anti-cancer agents or chemotherapeutic agents, and in some embodiments, in certain improved dosages or formulations. Benefits that can result include increased efficacy and/or lower doses when the specified combinations are used. Tumors, including cancers which benefit from such combinations or alternations are those that are mediated by the estrogen receptor and will also respond to the second agent. Specific examples of agents, active agents, anti-cancer agents or chemotherapeutic agents that can be combined with compounds described in US 2013/0178445 and WO 2013/090921 include an mTOR inhibitor such as everolimus, or a CDK 4/6 inhibitor such as PD-0332991, or a PI3 Kinase Inhibitor such as BKM-120, or a taxane, or an antimetabolite such as 5-fluorouracil, or an antitumor antibiotic such as doxorubicin. In one embodiment, the combination therapy includes the described anti-estrogenic compounds combined with PD-0332991 for treatment of retinoblastoma positive breast cancer as well as retinoblastoma positive endometrial, vaginal and ovarian cancers and lung and bronchial cancers.
[0098] The term "combinations of the invention" or the like (such as "combined with") as used herein refers to the concerted use of one or more of the compounds described in US
2013/0178445 and WO 2013/090921 with an agent, active agent, anti-cancer agent or
chemotherapeutic agent, wherein the concerted use can be either (i) a pharmaceutically acceptable formulation that includes both active agents; or (ii) separate pharmaceutical formulations that independently provide each active and are either administered at the same time or at different times but in a concerted synchronized fashion in a manner that optimizes the benefit of the combination therapy (i.e., alternation therapy). For example, certain actives will normally be administered orally, such as in a tablet, pill or liquid, and others might be normally administered via intravenous injection or other systemic, topical, parenteral, transdermal or other route.
[0099] The compounds that can be combined or alternated with agents, active agents, anti-cancer agents or chemotherapeutic agents are described in US 2013/0178445 and WO 2013/090921 and depicted below as OP-1038 and OP-1074. The chemical name for OP-1038 is 3 -(4-hydroxyphenyl)-4-methyl-2-(4- { 2- [(3R)-3 -methylpyrrolidin- 1 -yl] ethoxy } phenyl)-2H- chromen-7-ol. The chemical name for OP-1074 is (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-{2- [(3R)-3-methylpyrrolidin-l-yl]ethoxy}phenyl)-2H-chromen-7-ol.
[00100] The active compound to be combined or alternated with an agent, active agent, anti-cancer agent or chemotherapeutic agent, can be provided if desired as a pharmaceutically acceptable salt, solvate, hydrate, prodrug, stereoisomer, tautomer, N-oxide or R1 and/or R2- substituted derivative optionally in a pharmaceutically acceptable composition to treat a tumor that is modulated or affected by an estrogen receptor, including those treatable with an antiestrogenic compound optimally with virtually no estrogenic effect.
Figure imgf000027_0001
OP-1038
Figure imgf000027_0002
Figure imgf000028_0001
OP-1074
Figure imgf000028_0002
[00101] OP-1038 and OP-1074 have two chiral carbons and thus there are four possible diastereomers. The chiral carbon at the C2 position is in the S-configuration in OP-1074 (the same configuration in EM-652, acolbifene) and is a mixture of R and S in OP-1038.
[00102] OP-1038, OP-1074 and their prodrugs (including esters, carbonates and phosphates), derivatives and their salts when combined with specific agents, active agents, anticancer agents or chemotherapeutic agents, are especially useful to treat locally advanced or metastatic breast cancer that is positive for expression of estrogen receptors, progesterone receptors or both (receptor positive advanced breast cancer) and are also responsive to the specific agent, active agent, anti-cancer agent or chemotherapeutic agent. In an alternative embodiment, the combinations are used to treat estrogen or progesterone receptor negative breast cancer. The combinations can be used as the initial treatment of advanced breast cancer in patients who have never received previous hormonal therapy for advanced breast cancer. Specific examples of active agents that can be combined with compounds described in US 2013/0178445 and WO 2013/090921 include an mTOR inhibitor such as everolimus, or a CDK 4/6 inhibitor such as PD-0332991, or a PI3 Kinase Inhibitor such as BKM-120, or a taxane, or an
antimetabolite such as 5-fluorouracil, or an antitumor antibiotic such as doxorubicin. In one embodiment, the combination therapy includes the compounds described in US 2013/0178445 and WO 2013/090921 combined with PD-0332991 for treatment of retinoblastoma positive breast cancer as well as retinoblastoma positive endometrial, vaginal and ovarian cancers and lung and bronchial cancers. [00103] The combinations described in the present invention are also useful as adjuvant therapy after surgery to prevent recurrance. Such adjuvant use is often administered for several years, for instance 5 years, or even up to 10 years after surgery and associated chemotherapy and radiotherapy have been concluded. In one embodiment, the adjuvant combination therapy includes the compounds described in US 2013/0178445 and WO 2013/090921 combined with PD-0332991 for treatment of retinoblastoma positive breast cancer as well as retinoblastoma positive endometrial, vaginal and ovarian cancers and lung and bronchial cancers.
[00104] The combinations described in the present invention are also useful for the prevention of breast cancer in women at high risk for breast cancer and can be taken for any desired time period, including indefinitely. For example, a patient, typically a woman, with a family history of breast cancer, or who has been determined to carry a mutation in the BRCAl or BRCA2 gene or other genes that predispose a patient to breast cancer may choose to use such preventative treatment instead of a mastectomy or other intervention. The combinations described herein are also useful as neoadjuvants to shrink large tumors prior to surgical removal, both to enable breast conservative surgery and to reduce the risk of recurrence. In addition to breast cancer these combinations are also useful to treat other cancers and other overgrowth diseases of the female reproductive tract including ovarian, endometrial, and vaginal cancer and endometriosis. Besides these reproductive tissues the combinations are useful in treating lung cancers that are positive for estrogen or progesterone receptors. When such cancers are retinoblastoma positive, the combination of the compound described in US 2013/0178445 and WO 2013/090921 with PD-0332991 is beneficial.
[00105] Other objects and advantages will become apparent to those skilled in the art from a consideration of the ensuing detailed description. All variations and modifications of the disclosed invention are considered within the scope of this invention.
BRIEF DESCRIPTION OF THE FIGURES
[00106] Figure 1. OP-1074 increases the potency and efficacy of the CDK 4/6 inhibitor
PD-0332991 in inhibiting E2-stimulated MCF-7 breast cell proliferation. The antiestrogen OP- 1074 enhances the efficacy and potency of PD-0332991 in inhibiting E2-stimulated breast cell proliferation in vitro. MCF-7 cells were treated with the indicated amount of antiestrogen and PD-0332991 in hormone-depleted medium for 6-7 days in the presence of 100 pM 17P-estradiol (E2). Fig. 1 A plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or ΙΟΟηΜ OP-1074 in combination with increasing amounts of PD-0332991. Fig. IB plots the effects on MCF-7 cell proliferation of vehicle or lOOnM PD-0332991 in combination with increasing amounts of OP- 1074. Fig. 1C plots the effects on MCF-7 cell proliferation of vehicle or 3nM OP- 1074, tamoxifen, or fulvestrant in combination with increasing amounts of PD-0332991. Proliferation was measured using Cyquant fluorescent DNA-binding dye (Invitrogen, Grand Island, NY). Results were from a single representative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing SEM.
[00107] Figure 2. OP- 1074 increases the potency of the mTOR inhibitor everolimus in inhibiting E2-stimulated MCF-7 breast cell proliferation. The antiestrogen OP- 1074 enhances the efficacy and potency of the mTOR inhibitor everolimus in inhibiting E2-stimulated breast cell proliferation in vitro. MCF-7 cells were treated with the indicated amount of OP-1074 and everolimus in hormone-depleted medium for 6 days in the presence of 100 pM 17P-estradiol (E2). Fig. 2A plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or lOOnM OP-1074 in combination with increasing amounts of everolimus. Fig. 2B plots the effects on MCF-7 cell proliferation of vehicle, InM or ΙΟηΜ everolimus in combination with increasing amounts of OP-1074. Fig. 2C plots the effects on MCF-7 cell proliferation of vehicle or 3nM OP-1074, tamoxifen, or fulvestrant in combination with increasing amounts of everolimus. Proliferation was measured as in Figure 1 and results were from a single representative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing SEM.
[00108] Figure 3. OP-1074 increases the potency of the pan PI3K inhibitor BKM-120 in inhibiting E2-stimulated MCF-7 breast cell proliferation. The antiestrogen OP-1074 enhances the efficacy and potency of the PI3K inhibitor BKM-120 in inhibiting E2-stimulated breast cell proliferation in vitro. MCF-7 cells were treated with the indicated amount of OP-1074 and BKM-120 in hormone-depleted medium for 7 days in the presence of 100 pM 17P-estradiol (E2). Fig. 3 A plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or lOOnM OP-1074 in combination with increasing amounts of BKM-120. Fig. 3B plots the effects on MCF-7 cell proliferation of vehicle, 32nM or lOOnM BKM-120 in combination with increasing amounts of OP-1074. Proliferation was measured as in Figure 1 and results were from a single
representative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing SEM.
[00109] Figure 4. OP-1074 increases the potency of 5-Fluorouracil in inhibiting E2- stimulated MCF-7 breast cell proliferation. The antiestrogen OP-1074 enhances the efficacy and potency of the thymidylate synthase inhibitor 5-Fluorouracil in inhibiting E2-stimulated breast cell proliferation in vitro. MCF-7 cells were treated with the indicated amount of OP-1074 and 5- fluorouracil in hormone-depleted medium for 6 days in the presence of 100 pM 17P-estradiol (E2). The effects on MCF-7 cell proliferation of vehicle, 1, 10 or ΙΟΟηΜ OP-1074 in
combination with increasing amounts of 5-fluorouracil are plotted. Proliferation was measured as in Figure 1 and results were from a single representative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing SEM.
[00110] Figure 5. OP-1074 increases the potency of taxanes in inhibiting E2-stimulated
MCF-7 breast cell proliferation. The antiestrogen OP-1074 enhances the efficacy and potency of paclitaxel and docetaxel in inhibiting E2-stimulated breast cell proliferation in vitro. MCF-7 cells were treated with the indicated amount of OP-1074 and taxane in hormone-depleted medium for 6 days in the presence of 100 pM 17P-estradiol (E2). Fig. 5 A plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or ΙΟΟηΜ OP-1074 in combination with increasing amounts of paclitaxel. Fig. 5B plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or ΙΟΟηΜ OP- 1074 in combination with increasing amounts of docetaxel. Proliferation was measured as in Figure 1 and results were from a single representative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing SEM.
[00111] Figure 6. OP-1074 increases the potency of the anthracycline doxorubicin in inhibiting E2-stimulated MCF-7 breast cell proliferation. The antiestrogen OP-1074 enhances the efficacy and potency of the anthracycline doxorubicin in inhibiting E2-stimulated breast cell proliferation in vitro. MCF-7 cells were treated with the indicated amount of OP-1074 and doxorubicin in hormone-depleted medium for 7 days in the presence of 100 pM 17P-estradiol (E2). The effects on MCF-7 cell proliferation of vehicle, 1, 10 or ΙΟΟηΜ OP-1074 in
combination with increasing amounts of doxorubicin are plotted. Proliferation was measured as in Figure 1 and results were from a single representative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing SEM.
DETAILED DESCRIPTION OF THE INVENTION
[00112] The present invention relates to the treatment of tumors, including cancers, mediated by the estrogen receptor with the combination of a specific benzopyran (in the form of a mixture of S-C2 and R-C2 diastereomers and also its pure S-diastereomer) as described in US 2013/0178445 and WO 2013/090921 in combination or alternation with specific agents, active agents, anti-cancer agents or chemotherapeutic agents, and in some embodiments, in certain improved dosages or formulations. Benefits that can result include increased efficacy and/or lower doses when the specified combinations are used. Cancers which benefit from such combinations or alternations are those that are mediated by the estrogen receptor and will also respond to the second agent. Specific examples of agents, active agents, anti-cancer agents or chemotherapeutic agents that can be combined with compounds described in US 2013/0178445 and5 and WO 2013/090921 include an mTOR inhibitor such as everolimus, or a CDK 4/6 inhibitor such as PD-0332991, or a PI3 Kinase Inhibitor such as BKM-120, or a taxane, or an antimetabolite such as 5-fluorouracil, or an antitumor antibiotic such as doxorubicin. In one embodiment, the combination therapy includes the described anti-estrogenic compounds combined with PD-0332991 for treatment of retinoblastoma positive breast cancer as well as retinoblastoma positive endometrial, vaginal and ovarian cancers and lung and bronchial cancers.
[00113] The compound to be provided in combination or alternation with particular agents, active agents, anti-cancer agents or chemotherapeutic agents, can be provided if desired as a pharmaceutically acceptable salt, solvate, hydrate, prodrug, stereoisomer, tautomer, N-oxide or Ri and/or R2-substituted derivative or a pharmaceutically acceptable composition thereof to treat a disorder that is mediated, modulated or affected by an estrogen receptor, including those treatable with an anti-estrogenic compound with virtually no estrogenic effect.
Figure imgf000032_0001
OP-1038
Figure imgf000032_0002
Figure imgf000033_0001
Figure imgf000033_0002
wherein Ri and R2 are independently either:
(i) OH or OR9,
(ii) wherein R9 is independently selected from H, halogen (CI, Br, I or F), natural or non- naturally occurring amino acid (bound through either the OC(O)- or C(0)0- (an ester) or the amino (through either -C(0)-N- or -N-C(O)- (an amide linkage)), R10, -OR10, or -SR10 where R10 is -C(=0)RC1, -C(=0)0RC1, -C(=0)SRC1, -C(=0)N(RC1)2; or polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;
-S(=0)2RC1, -S(=0)2ORcl, -S(=0)RC1, -S(=0)ORcl, -P(=0)2RC1, -P(=0)2ORcl, -P(=0)(ORcl)2, -P(=0)(RC1)2, or -P(RC1)(0RC1); or oxygen attached to an oxygen protecting group (to produce OH on administration), sulfur attached to a sulfur protecting group (to produce SH or a disulfide on administration), or nitrogen attached to a nitrogen protecting group (to produce - H- on administration);
and R can be independently selected from hydrogen, polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or two R groups are joined to form an substituted or unsubstituted heterocyclic ring.
[00114] In certain embodiments either or both of Ri or R2 is an ester, amide, carbonate or phosphate.
[00115] Specific examples of prodrugs of the described compounds that can be combined or alternated with the particular agents, active agents, anti-cancer agents or chemotherapeutic agents are:
Figure imgf000034_0001
Figure imgf000034_0002
OP-1086
Figure imgf000034_0003
OP-1084
Figure imgf000035_0001
Figure imgf000035_0002
Figure imgf000035_0003
OP-1087
[00116] Examples of useful metabolically cleavable prodrug groups include acetyl, methoxycarbonyl, benzoyl, methoxymethyl and trimethylsilyl groups.
[00117] Combinations of OP-1038, OP-1074 and their prodrugs (including esters, carbonates and phosphates), derivatives and their salts as described in US 2013/0178445 and WO 2013/090921 when combined with particular agents, active agents, anti-cancer agents or chemotherapeutic agents, are especially useful to treat locally advanced or metastatic breast cancer that is positive for expression of estrogen receptors, progesterone receptors or both (receptor positive advanced breast cancer). In an alternative embodiment, the combinations are used to treat estrogen or progesterone receptor negative breast cancer. Specific examples of active agents that can be combined with compounds of the present invention include an mTOR inhibitor such as everolimus, or a CDK 4/6 inhibitor such as PD-0332991, or a PI3 Kinase Inhibitor such as BKM-120, or a taxane, or an antimetabolite such as 5-fluorouracil, or an antitumor antibiotic such as doxorubicin. [00118] In one embodiment, the compounds as described in US 2013/0178445 and WO
2013/090921 can be combined with a CDK 4/6 inhibitor, for example PD-0332991. In another embodiment, OP-1074 is combined with PD-0332991.
[00119] In one embodiment, the compounds as described in US 2013/0178445 and WO
2013/090921 can be combined with an mTOR inhibitor, for example everolimus. In another embodiment, OP-1074 is combined with everolimus.
[00120] In one embodiment, the compounds as described in US 2013/0178445 and WO
2013/090921 can be combined with a PI3 Kinase Inhibitor, for example BKM-120. In another embodiment, OP-1074 is combined with BKM-120.
[00121] In one embodiment, the compounds as described in US 2013/0178445 and WO
2013/090921 can be combined with a taxane, for example paclitaxel or docetaxel. In another embodiment, OP-1074 is combined with paclitaxel or docetaxel.
[00122] In one embodiment, the compounds as described in US 2013/0178445 and WO
2013/090921 can be combined with antimetabolites, for example 5-fluorouracil. In another embodiment, OP-1074 is combined with 5-fluorouracil.
[00123] In one embodiment, the compounds as described in US 2013/0178445 and WO
2013/090921 can be combined with antitumor antibiotics, for example doxorubicin. In another embodiment, OP-1074 is combined with doxorubicin.
[00124] Compositions are described of combinations of the compounds as described in US 2013/0178445 and WO 2013/090921 combined or alternated with particular agents, active agents, anti-cancer agents or chemotherapeutic agents. The compositions can be administered in a pharmaceutical composition suitable for oral delivery to the patient, typically a human.
Alternatively, the compounds can be delivered in a carrier suitable for topical, transdermal (including by patch), intravenous, parenteral, intraortal, subcutaneous or other desired delivery route, including any method of controlled delivery, for example, using degradable polymers, or with nano or microparticles, liposomes, layered tablets or other structural frameworks which slow delivery.
[00125] The compositions can be in the form of salts of the actives. They can be administered as a pharmaceutically acceptable salt, for example, a pharmaceutically acceptable acid addition salt, including a hydrochloride, hydroiodide, hydrobromide, nitrate, sulfate, bisulfate, phosphate, acetate, lactate, citrate, tartrate, succinate, maleate, fumarate, benzoate, para-toluenesulfonate and the like. [00126] The compositions are used to treat or prevent a disorder modulated by the estrogen receptor in an animal, typically a mammal, and most typically a human.
[00127] The combinations can also be used as adjunctive therapy. For example, a therapeutically effective amount of the compound can be used in combination with another anticancer agent, especially for estrogen receptor positive breast cancer, but in some embodiments, for estrogen receptor negative breast cancer.
[00128] In another aspect of an embodiment of the present invention, a method of treating a patient with combinations of a once-a-day, orally administered, treatment regimen is provided, including selecting one of the plurality of treatment regimens and titrating the dosages supplied to the patient after weekly, monthly or quarterly periods of time. Some embodiments of the present invention facilitate the selection of a combination treatment regimen and/or the change in dosages or titration of dosages of a treatment regimen over time. The combination of the compounds of the present invention with other actives, in particular other agents, active agents, anti-cancer agents or chemotherapeutic actives, can be done in a single fixed dose combination pill such as a multi-pill, combopill or polypill. A polypill is a medication that is a drug product in pill form (i.e., tablet or capsule) that combines multiple active pharmaceutical ingredients. In one embodiment it is manufactured as a fixed-dose combination (FDC) drug product targeting treatment or prevention of a disease. Polypills can reduce the number of tablets or capsules (generally orally administered) that need to be taken, which in turn may facilitate handling and administration of pharmaceuticals as well as alleviate patient pill-burden. Sometimes the multiple drugs in a given polypill might all be aimed at a single underlying condition (or, group of related conditions), for whom a given combination of drugs/dosages might be appropriate (particularly in the case of mass-produced polypills, i.e. FDCs). In addition to the noted fixed-dose types of polypills, polypills can also be custom-made for specific patients through a process called pharmacy compounding.
[00129] In alternative embodiments, rather than combining the customized combination of medications into a single treatment regimen, the present invention also relates to protocols enabling the physician or other healthcare provider to deliver the medications in a plurality of treatment regimens in a combination or alternation dose packaging (once-a-day, more than once a day, alternate days, every third day, twice a week, once a week, once every other week, once a month and so on) to increase compliance with medical therapy.
[00130] The present invention provides protocols for a plurality of pills into a combination dose packaging or polypill corresponding to the combination treatments mentioned above used in treating a patient for cancer, and in particular breast cancer, as well as other estrogen mediated diseases that would benefit from the compounds of the present invention, and related health issues over a weekly, monthly, quarterly, or longer period of time.
[00131] The present invention can further greatly improve patient compliance by providing the combination doses over a period of time (e.g. week, month, quarter, or longer) and having the physician titrate the dosages (adjust the dosages of each effective therapeutic ingredient) over the period of time based upon the condition of the patient. If necessary, the combination treatment pack may provide alternative dosages for treatment at more than one time per day.
[00132] Additional embodiments within the scope provided herein are set forth in non-limiting fashion elsewhere herein and in the examples. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting in any manner.
PHARMACEUTICAL COMPOSITIONS
[00133] The term "combinations of the invention" or the like as used herein refers to the concerted use of one or more of the compounds described in US 2013/0178445 and WO
2013/090921 with an agent, active agent, anti-cancer agent or chemotherapeutic agent, wherein the concerted use can be either (i) a pharmaceutically acceptable formulation that includes both active agents; or (ii) separate pharmaceutical formulations that independently provide each active and are either administered at the same time or at different times but in a concerted synchronized fashion in a manner that optimizes the benefit of the combination therapy (i.e., alternation therapy). For example, certain actives will normally be administered orally, such as in a tablet, pill or liquid, and others might be normally administered via intravenous injection or other systemic, topical, parenteral, transdermal or other route. A "pharmaceutical composition" can thus contain either one of the active agents or a combination of agents, based on how the active agents are best administered according to compatibility with the carrier, dosage, dosage scheduling, possibility of drug holidays and other general differences in physical properties, convenience, efficacy, toxicity and dosage regimes.
[00134] In one aspect, the invention provides a pharmaceutical composition comprising a pharmaceutically effective amount of a compound as described in US 2013/0178445 and WO 2013/090921 in combination with a particular agent, active agent, anti-cancer agent or chemotherapeutic agent, and a pharmaceutically acceptable carrier. The amount of the composition administered will typically be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.
[00135] The pharmaceutical compositions provided herein can be administered by a variety of routes including oral, topical, parenteral, rectal, transdermal, subcutaneous, intravenous, intramuscular, and intranasal with a pharmaceutical carrier suitable for such administration. In one embodiment, the compound is administered in a controlled release formulation.
[00136] The compositions for oral administration can take the form of bulk liquid solutions or suspensions, or bulk powders. Typically, the compositions are presented in unit dosage forms to facilitate accurate dosing. The term "unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subjects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include prefilled, premeasured ampules or syringes of the liquid compositions or pills, tablets, capsules or the like in the case of solid compositions. In such compositions, the compound is usually a minor component (as a nonlimiting example, from about 0.1 to about 50% by weight or preferably from about 1 to about 40% by weight) with the remainder being various vehicles or carriers and processing aids helpful for forming the desired dosing form.
[00137] The compositions can be used for treating a patient with combinations of a once- a-day, orally administered, treatment regimen, including selecting one of the plurality of treatment regimens and titrating the dosages supplied to the patient after weekly, monthly or quarterly periods of time. Some embodiments of the present invention facilitate the selection of a combination treatment regimen and/or the change in dosages or titration of dosages of a treatment regimen over time. The combination of the compounds of the present invention with other actives, in particular other chemotherapeutic actives, can be done in a single fixed dose combination pill such as a multi-pill, combopill or polypill. A polypill is a medication that is a drug product in pill form (i.e., tablet or capsule) that combines multiple active pharmaceutical ingredients. In one embodiment it is manufactured as a fixed-dose combination (FDC) drug product targeting treatment or prevention of a disease. Polypills can reduce the number of tablets or capsules (generally orally administered) that need to be taken, which in turn may facilitate handling and administration of pharmaceuticals as well as alleviate patient pill-burden. Sometimes the multiple drugs in a given polypill might all be aimed at a single underlying condition (or, group of related conditions), for whom a given combination of drugs/dosages might be appropriate (particularly in the case of mass-produced polypills, i.e. FDCs). In addition to the noted fixed-dose types of polypills, polypills can also be custom-made for specific patients through a process called pharmacy compounding.
[00138] In addition, rather than combining the customized combination of medications into a single treatment regimen, the present invention also relates to protocols enabling the physician or other healthcare provider to deliver the medications in a plurality of treatment regimens in a combination protocol (once-a-day, more than once a day, alternate days, every third day, twice a week, once a week, once every other week, once a month and so on) to increase compliance with medical therapy.
[00139] The present invention provides protocols for a plurality of pills into a combination protocol or polypill corresponding to the combination treatments mentioned above used in treating a patient for cancer, and in particular breast cancer, as well as other estrogen mediated diseases that would benefit from the compounds of the present invention, over a weekly, monthly, quarterly, or longer period of time.
[00140] The present invention can further greatly improve patient compliance by providing the combination doses over a period of time (e.g. week, month, quarter, or longer) and having the physician titrate the dosages (adjust the dosages of each effective therapeutic ingredient) over the period of time based upon the condition of the patient. If necessary, the combination treatment may provide alternative dosages for treatment at more than one time per day.
[00141] Liquid forms suitable for oral administration may include a suitable aqueous or nonaqueous vehicle with buffers, suspending and dispensing agents, colorants, flavors and the like. Solid forms may include, for example, any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, Primogel, or corn starch; a lubricant such as magnesium stearate; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, or orange flavoring.
[00142] Injectable compositions are typically based upon injectable sterile saline or phosphate-buffered saline or other injectable carriers known in the art. [00143] Transdermal compositions are typically formulated as a topical ointment or cream containing the active ingredient(s), for example in an amount ranging from about 0.01 to about 20% by weight, preferably from about 0.1 to about 20% by weight, preferably from about 0.1 to about 10%) by weight, and more preferably from about 0.5 to about 15%> by weight. When formulated as an ointment, the active ingredients will typically be combined with either a suitable delivery polymeric composition, or a paraffinic or a water-miscible ointment base. Alternatively, the active ingredients may be formulated in a cream with, for example an oil-in-water cream base. Such transdermal formulations are well-known in the art and generally include additional ingredients to enhance the dermal penetration of stability of the active ingredients or the formulation. All such known transdermal formulations and ingredients are included within the scope provided herein.
[00144] The compositions provided herein can be administered by a transdermal device. Transdermal administration can be accomplished using a patch either of the reservoir or porous membrane type, or of a solid matrix variety.
[00145] The above-described components for orally administrable, injectable or topically administrable compositions are merely representative. Other materials as well as processing techniques and the like are set forth in Part 8 of Remington 's Pharmaceutical Sciences, 17th edition, 1985, Mack Publishing Company, Easton, Pennsylvania, which is incorporated herein by reference.
[00146] The compositions of this invention can also be administered in sustained release forms or from sustained release drug delivery systems. A description of representative sustained release materials can be found in Remington 's Pharmaceutical Sciences.
[00147] In certain embodiments, the formulation comprises water. In another embodiment, the formulation comprises a cyclodextrin derivative. In certain embodiments, the formulation comprises hexapropyl-P-cyclodextrin. In a more particular embodiment, the formulation comprises hexapropyl-P-cyclodextrin (10-50%) in water).
[00148] The present invention also includes pharmaceutically acceptable acid addition salts of compounds of the compositions of the invention. The acids which are used to prepare the pharmaceutically acceptable salts are those which form non-toxic acid addition salts, i.e. salts containing pharmacologically acceptable anions such as the hydrochloride, hydroiodide, hydrobromide, nitrate, sulfate, bisulfate, phosphate, acetate, lactate, citrate, tartrate, succinate, maleate, fumarate, benzoate, para-toluenesulfonate, and the like. USE OF COMPOUNDS IN MEDICAL THERAPY
[00149] Combinations of OP-1038, OP-1074 and their prodrugs (including esters, carbonates and phosphates), derivatives and their salts as described in US 2013/0178445 and WO 2013/090921 with particular agent, active agent, anti-cancer agent or chemotherapeutic agents are useful to treat disorders modulated, mediated or affected by the estrogen receptor.
[00150] In one embodiment, the combinations of the present invention can be used to treat local, advanced or metastatic breast cancer that is positive for expression of estrogen receptors, progesterone receptors or both (receptor positive advanced breast cancer). In an alternative embodiment, the combinations are used to treat estrogen or progesterone receptor negative breast cancer. The combinations can be used as the initial treatment of advanced breast cancer in patients who have never received previous hormonal therapy for advanced breast cancer, in combination with one or more other anti-cancer agents described below or otherwise known to those skilled in the art. They are also useful for second line therapy for treatment after a previous hormonal therapy has failed, either by itself or in combination with another anti-cancer agent, for example, a targeted therapy such as an mTOR inhibitor such as everolimus or a CDK 4/6 inhibitor such as PD-0332991.
[00151] The combinations of the invention are also useful as adjunctive therapy after or instead of chemotherapy, radiation or surgery. Such adjuvant use is often used for several years, perhaps 5 years, after chemotherapy or other therapies have been concluded, but may optimally be continued for additional years.
[00152] The combinations of the invention are also useful for the prevention of breast cancer in women at high risk and can be taken for any desired time period, including indefinitely. For example, a patient, typically a woman, with a family history of breast cancer, or who has been determined to carry a mutation in the BRCA1 or BRCA2 gene or other genes that predispose a patient to breast cancer may choose to use such preventative treatment instead of a mastectomy or other intervention. The combinations described herein are also useful as neoadjuvants to shrink large tumors prior to surgical removal, both to enable breast conservative surgery and to reduce the risk of recurrence. In addition to breast cancer these combinations also are useful in treating other cancers and other overgrowth diseases of the female reproductive tract including ovarian, endometrial, and vaginal cancer and endometriosis. Besides these reproductive tissues the combinations are useful in treating lung cancers that are positive for estrogen or progesterone receptors. [00153] The present combinations are used as therapeutic or prophylactic agents for the treatment of conditions in mammals, particularly humans whose conditions are modulated by estrogen receptors.
[00154] Combinations described herein are useful for treating locally advanced or metastatic breast cancer, preventing recurrence or early breast cancer after surgery, and preventing breast cancer in women at high risk. Such combinations are useful for treating all estrogen-dependent cancers of the reproductive tract including endometrial and ovarian cancers. Such combinations have potential uses in the treatment of lung and bronchial cancers that express estrogen receptors.
[00155] Specific examples of active agents that can be combined with compounds as described in US 2013/0178445 and WO 2013/090921 include an mTOR inhibitor such as everolimus, or a CDK 4/6 inhibitor such as palbociclib (PD-0332991), or a PI3 Kinase Inhibitor such as BKM-120, or taxanes, or antimetabolites such as 5-fluorouracil, or antitumor antibiotics such as doxorubicin. Such combinations are described in more detail below.
CDK 4/6 Combination Therapy
[00156] CDK 4/6 inhibitors for the treatment of cancer are presently in the clinic.
Examples include PD-0332991 (palbociclib, Pfizer), LY2835219 (Lilly), and LEEOl l (Novartis). CDK 4/6 normal function in cell cycle regulation is through phosphorylation of retinoblastoma protein at Serine 780 and 795. Cancers which are Retinoblastoma positive are targets for CDK 4/6 inhibitors.
[00157] PD-0332991 (palbociclib, Pfizer), 6-acetyl-8-cyclopentyl-5-methyl-2-(5- piperazin-l-yl-pyridin-2-ylamino)-8H-pyrido[2,3-<i]pyrimidin-7-one or 6-acetyl-8-cyclopentyl-5- methyl-2-{[5-(l-piperazinyl)-2-pyridinyl]amino}pyrido[2,3-d]pyrimidin-7(8H)-one was first described and synthesized in Barvian et al. (WO 2003/062236). Further disclosures of its use, specific salts and methods of synthesis can be found, for instance, in WO 2005/005426; WO 2008/032157; US 6,936,612; US 7,208,489; US 7,456, 168; US 7,863,278; US 7,345, 171; and US 7,781,583.
[00158] PD-0332991 is currently being evaluated in numerous tumor types including late- line metastatic breast cancer, liposarcoma, non-small cell lung cancer, liver cancer, ovarian cancer, glioblastoma, refractory solid tumors, multiple myeloma and mantle cell lymphoma. Such cancers are generally categorized as retinoblastoma positive as CDK 4/6 normal function in cell cycle regulation is through phosphorylation of retinoblastoma protein at Serine 780 and 795. Currently PD-0332991 is dosed at 125 mg, orally once daily on Day 1 to Day 21 of every 28-day cycle followed by 7 days off treatment.
[00159] PD-0332991 is being evaluated in combination with the aromatase inhibitor letrozole in a comparison trial with letrozole plus placebo for 1st line metastatic breast cancer. Preliminary results reported in December 2012 suggest the combination is substantially superior to letrozole alone in progression-free survival.
[00160] Cancers that would benefit from the combination of compounds of the present invention, including OP- 1038 and OP- 1074 include include advanced or metastatic breast cancer, preventing recurrence or early breast cancer after surgery, and preventing breast cancer in women at high risk. Other cancers include retinoblastoma positive estrogen-dependent cancers of the reproductive tract including endometrial and ovarian cancers as well as retinoblastoma positive lung and bronchial cancers that express estrogen receptors.
[00161] Suitably, the amount of PD-0332991 administered as part of the combination according to the present invention will be an amount selected from about 1.25 mg to about 250 mg; suitably, the amount will be selected from about 2 mg to about 150 mg; suitably, the amount will be selected from about 2.5 mg to about 125 mg. Accordingly, the amount of PD-0332991 administered as part of the combination according to the present invention will be an amount selected from about 1.25 mg to about 250 mg. For example, the amount of PD-0332991 administered as part of the combination according to the present invention can be a lower dose than that currently administered alone, such as 1.25 mg, 2.5 mg, 5 mg, 7.5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, 95 mg, 100 mg, 105 mg, 110 mg, 115 mg, 120 mg, or 125 mg. Suitably, the selected amount of PD-0332991 is administered once a day. Suitably, the selected amount of PD-0332991 is administered twice a day.
[00162] PD-0332991 administered as part of the combination according to the present invention orally once daily on Day 1 to Day 21 of every 28-day cycle followed by 7 days off treatment. Alternatively, PD-0332991 may be administered with less days off treatment as part of every 28 day cycle, for instance daily on Day 1 to Day 22 of every 28-day cycle followed by 6 days off treatment, daily on Day 1 to Day 23 of every 28-day cycle followed by 5 days off treatment, daily on Day 1 to Day 24 of every 28-day cycle followed by 4 days off treatment, daily on Day 1 to Day 25 of every 28-day cycle followed by 3 days off treatment, daily on Day 1 to Day 26 of every 28-day cycle followed by 2 days off treatment, daily on Day 1 to Day 27 of every 28-day cycle followed by 1 days off treatment, or daily for the entire 28 -day cycle with no days of off treatment.
M-Tor Inhibitor Combination Therapy
[00163] For use herein, the term mTOR inhibitor, mTOR and derivatives thereof, unless otherwise defined, include but are not limited to rapamycin and its analogs, RAD001 or everolimus (Afinitor), CCI-779 or temsirolimus, AP23573, AZD8055, WYE-354, WYE-600, WYE-687 and Ppl21. Suitably, the mTOR inhibitor is selected form rapamycin, everolimus (Afinitor) and temsirolimus. Suitably, the mTOR inhibitor is selected from rapamycin and everolimus (Afinitor). Suitably the mTOR inhibitor is everolimus.
[00164] Suitably, the amount of everolimus administered as part of the combination according to the present invention will be an amount selected from about 1.25 mg to about 20 mg; suitably, the amount will be selected from about 2 mg to about 15 mg; suitably, the amount will be selected from about 2.5 mg to about 10 mg. Accordingly, the amount of everolimus administered as part of the combination according to the present invention will be an amount selected from about 1.25 mg to about 20 mg. For example, the amount of everolimus
administered as part of the combination according to the present invention can be 1.25 mg, 1.5 mg, 2 mg, 2.5 mg, 3 mg, 3.5 mg, 4 mg, 4.5 mg, 5 mg, 5.5 mg, 6 mg, 6.5 mg, 7 mg, 7.5 mg, 8 mg, 8.5 mg, 9 mg, 9.5 mg, 10 mg, 11 mg, 12 mg, 13 mg, 14 mg, 15 mg, 16 mg, 17 mg, 18 mg, 19 mg, 20 mg. Suitably, the selected amount of everolimus is administered twice a day. Suitably, the selected amount of everolimus is administered once a day. Suitably, the administration of everolimus will begin as a loading dose. Suitably, the loading dose will be an amount from 2 to 100 times the maintenance dose; suitably from 2 to 10 times; suitably from 2 to 5 times; suitably 2 times; suitably 3 times; suitably 4 times; suitably 5 times. Suitably, the loading does will be administered from 1 to 7 days; suitably from 1 to 5 days; suitably from 1 to 3 days; suitably for 1 day; suitably for 2 days; suitably for 3 days, followed by a maintenance dosing protocol.
[00165] Suitably, the amount of temsirolimus administered as part of the combination according to the present invention will be an amount infused over a 30 to 60 minute period, where the amount is selected from about 5 mg to about 50 mg; suitably, the amount will be selected from about 10 mg to about 40 mg; suitably, the amount will be selected from about 15 mg to about 35 mg. Accordingly, the amount of temsirolimus administered as part of the combination according to the present invention will be an amount selected from about 5 mg to about 50 mg. For example, the amount of temsirolimus administered as part of the combination according to the present invention can be 5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg. Suitably, the selected amount of temsirolimus is administered twice a day. Suitably, the selected amount of temsirolimus is administered once a day. Suitably, the administration of temsirolimus will begin as a loading dose. Suitably, the loading dose will be an amount from 2 to 100 times the maintenance dose; suitably from 2 to 10 times; suitably from 2 to 5 times; suitably 2 times; suitably 3 times; suitably 4 times; suitably 5 times. Suitably, the loading does will be administered from 1 to 7 days; suitably from 1 to 5 days; suitably from 1 to 3 days; suitably for 1 day; suitably for 2 days; suitably for 3 days, followed by a maintenance dosing protocol.
PI3 Kinase Inhibitors Combination Therapy
[00166] PI3K inhibitors may include, without being limited to, BKM-120, XL- 147, RG-
7321 (GDC-0941), CH-5132799 and BAY-80-6946.
[00167] Phosphoinositides (Pis), which are phosphorylated derivatives of
phosphatidylinositol, are essential in eukaryotic cells, regulating nuclear processes, cytoskeletal dynamics, signaling and membrane trafficking. Among the enzymes involved in PI metabolism, PI3 -kinases (PI3K) have attracted special attention because of their oncogenic properties and potential as drug targets. PI3-kinases phosphorylate phosphatidylinositols or Pis at the 3-position of the inositol ring. (Lindmo et. al. Journal of Cell Science 119, 605-614, 2006). The 3- phosphorylated phospholipids generated by PI3K activity bind to the pleckstrin homology (PH) domain of protein kinase B (PKB or AKT), causing translocation of PKB to the cell membrane and subsequent phosphorylation of PKB. Phosphorylated PKB inhibits apoptosis-inducing proteins such as FKHR, Bad, and caspases, and is thought to play an important role in cancer progression. The PDKs are divided into classes I-III, and class I is further subclassified into classes la and lb. Among these isoforms, class la enzymes are thought to play the most important role in cell proliferation in response to growth factor-tyrosine kinase pathway activation
(Hayakawa et al., Bioorganic & Medicinal Chemistry 14 6847-6858, 2006). Three frequent mutations in cancer constitutively activate PI3K. alpha, and, when expressed in cells, they drive the oncogenic transformation and chronic activation of downstream signaling by molecules such as PKB, S6K and 4E bpl that is commonly seen in cancer cells. (Stephens et. al., Current Opinion in Pharmacology, 5(4) 357-365, 2005). As such, PI3-kinases are attractive targets for the treatment of proliferative diseases. [00168] There are several known PI3 -kinase inhibitors including wortmannin and
LY294002. Although wortmannin is a potent PI3K inhibitor with a low nanomolar IC 50 value, it has low in vivo anti-tumor activity. (Hayakawa et al, Bioorg Med Chem 14(20), 6847-6858 (2006)). Recently, a group of morpholine substituted quinazoline, pyridopyrimidine and thienopyrimidine compounds have been reported to be effective in inhibiting PI3 kinase pi 10. alpha.. (Hayakawa, 6847-6858). Oral dosage of a morpholine substituted thienopyrimidine compound (GDC-0941) has shown tumor suppression in glioblastoma xenografts in vivo. (Folkes et al., Journal of Medicinal Chemistry, 51, 5522-5532, 2008). The following publications disclose a series of thienopyrimidine, pyridopyrimidine and quinazoline based PI3-Kinase inhibitors: WO 2008/073785; WO 2008/070740; WO 2007/127183; U.S. Patent Publication 20080242665.
[00169] Examples of Phosphatidylinositol-3 -kinase (PI3-kinase) inhibitors include, but are not limited to, e.g., celecoxib and analogs thereof, such as OSU-03012 and OSU-03013 (e.g., Zhu et al., Cancer Res., 64(12): 4309-18, 2004); 3-deoxy-D-myo-inositol analogs (e.g., U.S. Application No. 20040192770; Meuillet et al., Oncol. Res., 14:513-27, 2004), such as PX-316; 2'-substituted 3'-deoxy-phosphatidyl-myo-inositol analogs (e.g., Tabellini et al., Br. J. Haematol., 126(4): 574-82, 2004); fused heteroaryl derivatives (U.S. Pat. No. 6,608,056); 3-(imidazo[l,2- a]pyridin-3-yl) derivatives (e.g., U.S. Pat. Nos. 6,403,588 and 6,653,320); Ly294002 (e.g., Vlahos, et al., J. Biol., Chem., 269(7) 5241-5248, 1994); quinazoline-4-one derivatives, such as IC486068 (e.g., U.S. Application No. 20020161014; Geng et al., Cancer Res., 64:4893-99, 2004); 3-(hetero)aryloxy substituted benzo(b)thiophene derivatives (e.g., WO 04 108715; also WO 04 108713); viridins, including semi -synthetic viridins such as such as PX-866 (acetic acid ( 1 S,4E, 1 OR, 11R, 13 S, 14R)-[4-diallylaminomethylene-6-hydroxy- 1 -methoxymethy- 1-10,13- dimethyl-3,7, 17-trioxo-l,3,4,7, 10,l l, 12,13, 14,15, 16,17-dodecahydro- -2-oxa- cyclopenta[a]phenanthren-l l-yl ester) (e.g., Ihle et al., Mol Cancer Ther., 3(7):763-72, 2004; U.S. Application No. 20020037276; U.S. Pat. No. 5,726,167); and wortmannin and derivatives thereof (e.g., U.S. Pat. Nos. 5,504, 103; 5,480,906, 5,468,773; 5,441,947; 5,378,725; 3,668,222). In a certain embodiment the PI3K inhibitor is 5-[2,6-di(4-morpholinyl)-4-pyrimidinyl]-4- (trifluoromethyl)-2-pyridinamine (BKM-120).
[00170] BKM-120 is currently being tested in Phase III clinical trials at a daily dose of 100 mg via oral capsules. Suitably, the amount of BKM-120 administered as part of the combination according to the present invention will be an amount selected from about 1.25 mg to about 250 mg; suitably, the amount will be selected from about 2 mg to about 150 mg; suitably, the amount will be selected from about 2.5 mg to about 100 mg. Accordingly, the amount of BKM-120 administered as part of the combination according to the present invention will be an amount selected from about 1.25 mg to about 250 mg. For example, the amount of BKM-120
administered as part of the combination according to the present invention can be a lower dose than that currently administered alone, such as 1.25 mg, 2.5 mg, 5 mg, 7.5 mg, 10 mg, 15 mg, 20 mg, 25 mg, 30 mg, 35 mg, 40 mg, 45 mg, 50 mg, 55 mg, 60 mg, 65 mg, 70 mg, 75 mg, 80 mg, 85 mg, 90 mg, or 95 mg. Suitably, the selected amount ofBKM-120 is administered once a day. Suitably, the selected amount of BKM-120 is administered twice a day.
5-Flurouracil Combination Therapy
[00171] Fluorouracil or 5-FU (5-fluoro-lH,3H-pyrimidine-2,4-dione, trademarked as
Efudex) is a drug that is a pyrimidine analog which is used in the treatment of cancer. It is a suicide inhibitor and works through irreversible inhibition of thymidylate synthase. 5-FU is an intravenously (IV) administered fluorinated pyrimidine cytotoxic agent that inhibits the function of thymidylate synthase, an enzyme necessary for the production of the thymidine nucleotides required for DNA synthesis. 5-FU has activity in the therapy of a number of tumor types but is most commonly given in the treatment of colorectal cancer, upper gastrointestinal malignancies, and breast cancer.
[00172] 5-FU is currently administered in various protocols such as 500 mg/m2 IV on
Days 1-5, or 450-600 mg/m2 IV weekly, or 200-400 mg/m2 rV continuous infusion every day; generally not to exceed 800 mg/day.
[00173] A method that has been used to overcome the poor oral bioavailability of 5-FU involves the administration of a prodrug that has good bioavailability and is ultimately converted to 5-FU. Capecitabine (pentyl [l-(3,4-dihydroxy-5-methyltetrahydrofuran-2-yl)-5-fluoro-2-oxo- lH-pyrimidin-4-yl]carbamate, Xeloda) is such a novel oral fluoropyrimidine carbamate. It is readily absorbed from the gastrointestinal tract and is preferentially converted to 5-FU in tumor tissue. After oral administration, capecitabine passes intact from the gastrointestinal tract to the liver, where it is converted by carboxylesterases to 5'-deoxy-5-flourocytidine (5'-DFCR), then by cytidine deaminase in liver and tumor tissue to 5'-deoxy-5-flourouridine (5'-DFUR), and finally by thymidine phosphorylase (dThdPase) in tumor tissue to 5-FU. The usual starting dose is 2,500 mg/m2/day in two divided doses, 12 hours apart. One cycle includes two weeks of treatment followed by one week without treatment. Cycles can be repeated every three weeks. [00174] More specifically, when the compound of the invention is combined with 5-FU, the dosage of 5-FU may be, for example, 10-1000 mg/ m2/day, preferably 50-1000 mg/ m2/day and more preferably 100-600 mg/ m2/day.
[00175] More specifically, when the compound of the invention is combined with
Capecitabine, the dosage Capecitabine may be, for example, 100-10000 mg/ m2/day, preferably 500-5000 mg/ m2/day and more preferably 1000-5000 mg/ m2/day.
Doxorubicin Combination Therapy
[00176] Doxorubicin, also known as Adriamycin, is one of the more common cytotoxic agents used for the treatment of breast cancer. The term doxorubicin includes doxorubicin as well as the pharmaceutically acceptable salts of doxorubicin. Doxorubicin or hydroxyldaunorubicin is an antineoplastic drug widely used in chemotherapy (Hortobagyi, Drugs, 54 (Supplement 4): 1-7 (1997)). It is an anthracycline antibiotic and structurally closely related to daunomycin (Minotti et al., Pharmacologiy. Rev., 56; 185-229 (2004). Doxorubicin (DOX) is commonly used in the treatment of a wide range of cancers, including cancers of the blood, lymph system, bladder, breast, stomach, lung, ovaries, thyroid, nerves, kidneys, bones, soft tissues, including muscles and tendons, multiple myeloma, and others. The hydrochloride salt of doxorubicin is one of the most common forms. It is referred to by various names, such as doxorubicin hydrochloride; 14- hydroxydaunorubicin hydrochloride; 3-hydroxyacetyldaunorubicin hydrochloride; and 5, 12- naphthacenedione, 10-[(3-amino-2,3,6-trideoxy-.alpha.-L-lyxo-hexopyranosyl)oxy]-7,8,9, 10-te- trahydro-6,8,1 l-trihydroxy-8-(hydroxyacetyl)-l-methoxy-, hydrochloride, (8S-cis)-(9Cl).
Doxorubicin hydrochloride has the molecular formula C27H29NC)ii.HCl, a molecular weight (MW) of 580.0, and CAS number 25316-40-9.
[00177] The dosage amount of doxorubicin is preferably in the range from 30 to 100 mg/m2/day, more preferably 40 to 80 mg/m2/day, more preferably a dose of about 50 mg/m2/day or about 60 mg/m2/day. Infusion times for doxorubicin are generally up to 6 hours, more preferable 1-3 hours, with 1 hour most preferred.
[00178] More specifically, when the compound of the present invention is combined with doxorubicin, the dosage may be, for example, 10-100 mg/ m2/day, preferably 30-100 mg/ m2/day and more preferably 30-60 mg/ m2/day.
Taxane Combination Therapy [00179] Examples of taxane include paclitaxel (trade name: Taxol) and docetaxel (trade name: Taxotere). Poliglumex paclitaxel (trade name: Opaxio) is also included in taxanes. Such taxanes have been approved and developed for application to breast cancer, non-small-cell lung cancer, gastric cancer, head and neck cancer, ovarian cancer, esophageal cancer, gastric cancer, uterine body cancer or the like. In addition, combination therapy for various cancers has also been approved or developed by combining taxane with various drugs, for example, with bevacizumab for breast cancer and with carboplatin for ovarian cancer and non-small-cell lung cancer.
[00180] Taxane may be administered according to known clinical practice. The dosage and dosing schedule may be altered according to a specific symptom or all symptoms of the patient's disease. The dosage may appropriately be reduced according to age, symptoms or incidence of side effects. Upon use of the pharmaceutical composition and/or the kit of the invention, taxane may usually, but without limitation, be administered for 0.01-10000 mg/m2/day, preferably 0.1- 1000 mg/m2/day and more preferably 1-500 mg/m2/day for an adult, which may be administered usually once to three times a day. The dosage needs to be reduced if undue toxicity occurs in the patient. The dosage and dosing schedule may be altered when one or more additional agents, active agents, anti-cancer agents or chemotherapeutic agents are used in addition to the combination therapy of the invention. The dosing schedule may be determined by the physician in charge of the treatment of the specific patient.
[00181] More specifically, when the compound of the invention is combined with paclitaxel, the dosage paclitaxel may be 0.01-10000 mg/m2/day, preferably 0.1-1000 mg/m2/day and more preferably 1-500 mg/m2/day.
[00182] Furthermore, when the compound of the invention is combined with docetaxel, the dosage of docetaxel may be 0.01-10000 mg/m2/day, preferably 0.1-1000 mg/m2/day and more preferably 1-500 mg/m2/day.
[00183] In addition, when the compound of the invention is combined with poliglumex paclitaxel, the dosage of poliglumex paclitaxel may be 0.01-10000 mg/m2/day, preferably 0.1- 1000 mg/m2/day and more preferably 1-500 mg/m2/day.
[00184] In another embodiment, the combinations of the present invention are provided for use in medical therapy, including for any of the conditions described herein. The use of the present compositions in the manufacture of a medicament for the treatment or prevention of one of the aforementioned conditions and diseases is also provided. [00185] Injection dose levels range are provided in any desired dosage of the active agents, for example, from about 0.1 mg/kg/hour to at least 10 mg/kg/hour, all for from about 1 to about 120 hours and especially 24 to 96 hours. In one embodiment, a preloading bolus of from about 0.1 mg/kg to about 10 mg/kg or more may also be administered to achieve adequate steady state levels. The maximum total dose is not expected to exceed about 2 g/day for a 40 to 80 kg human patient.
[00186] For oral dosing, any dose of the active agents, as combined, is appropriate that achieved the desired goals. In one example, suitable daily dosages of active agents of the present invention are between about 0.1-4000 mg, more typically between 5 mg and 1 gram, more typically between 10 mg and 500 mg, and administered orally once-daily, twice-daily or three times-daily, continuous (every day) or intermittently (e.g., 3-5 days a week). For example, when used to treat any disorder described herein, the dose of the compounds of this invention usually ranges between about 0.1 mg, more usually 10, 50, 100, 200.250, 1000 or up to about 2000 mg per day.
GENERAL SYNTHETIC PROCEDURES
[00187] The compounds provided herein can be prepared from readily available starting materials using the following general methods and procedures. See, e.g., Synthetic Schemes below. It will be appreciated that where typical or preferred process conditions {i.e., reaction temperatures, times, mole ratios of reactants, solvents, pressures, etc.) are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvent used, but such conditions can be determined by one skilled in the art by routine optimization procedures.
[00188] Additionally, as will be apparent to those skilled in the art, conventional protecting groups may be necessary to prevent certain functional groups from undergoing undesired reactions. The choice of a suitable protecting group for a particular functional group as well as suitable conditions for protection and deprotection are well known in the art. For example, numerous protecting groups, and their introduction and removal, are described in T. W. Greene and P. G. M. Wuts, Protecting Groups in Organic Synthesis, Second Edition, Wiley, New York, 1991, and references cited therein.
[00189] The compounds provided herein may be isolated and purified by known standard procedures. Such procedures include (but are not limited to) recrystallization, column chromatography or HPLC. The following schemes are presented with details as to the preparation of representative substituted benzopyrans that have been listed herein. The compounds provided herein may be prepared from known or commercially available starting materials and reagents by one skilled in the art of organic synthesis.
[00190] The diastereomerically or enantiomerically pure compounds provided herein may be prepared according to any techniques known to those of skill in the art. For instance, they may be prepared by chiral or asymmetric synthesis from a suitable optically pure precursor or obtained from a racemate or mixture of diastereomers by any conventional technique, for example, by chromatographic resolution using a chiral column, TLC or by the preparation of diastereoisomers, separation thereof and regeneration of the desired enantiomer or diastereomer. See, e.g., "Enantiomers, Racemates and Resolutions," by J. Jacques, A. Collet, and S.H. Wilen, (Wiley-Interscience, New York, 1981); S.H. Wilen, A. Collet, and J. Jacques, Tetrahedron, 2725 (1977); E.L. Eliel Stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); and S.H. Wilen Tables of Resolving Agents and Optical Resolutions 268 (E.L. Eliel ed., Univ. of Notre Dame Press, Notre Dame, IN, 1972, Stereochemistry of Organic Compounds, Ernest L. Eliel, Samuel H. Wilen and Lewis N. Manda (1994 John Wiley & Sons, Inc.), and Stereoselective Synthesis A Practical Approach, Mihaly Nogradi (1995 VCH Publishers, Inc., NY, NY).
[00191] In certain embodiments, a diastereomerically pure compound may be obtained by reaction of the racemate or mix of diastereomers with a suitable optically active acid or base. Suitable acids or bases include those described in Bighley et al.., 1995, Salt Forms of Drugs and Adsorption, in Encyclopedia of Pharmaceutical Technology, vol. 13, Swarbrick & Boylan, eds., Marcel Dekker, New York; ten Hoeve & H. Wynberg, 1985, Journal of Organic Chemistry 50:4508-4514; Dale & Mosher, 1973, J. Am. Chem. Soc. 95:512; and CRC Handbook of Optical Resolution via Diastereomeric Salt Formation, the contents of which are hereby incorporated by reference in their entireties.
[00192] Enantiomerically or diastereomerically pure compounds can also be recovered either from the crystallized diastereomer or from the mother liquor, depending on the solubility properties of the particular acid resolving agent employed and the particular acid enantiomer or diastereomer used. The identity and optical purity of the particular compound so recovered can be determined by polarimetry or other analytical methods known in the art. The diasteroisomers can then be separated, for example, by chromatography or fractional crystallization, and the desired enantiomer or diastereomer regenerated by treatment with an appropriate base or acid. The other enantiomer or diasteromer may be obtained from the racemate or mix of diastereomers in a similar manner or worked up from the liquors of the first separation.
[00193] In certain embodiments, enantiomerically or diastereomerically pure compound can be separated from racemic compound or a mixture of diastereomers by chiral
chromatography. Various chiral columns and eluents for use in the separation of the enantiomers or diastereomers are available and suitable conditions for the separation can be empirically determined by methods known to one of skill in the art. Exemplary chiral columns available for use in the separation of the enantiomers provided herein include, but are not limited to
CHIRALPACK® IC, CHIRALCEL® OB, CHIRALCEL® OB-H, CHIRALCEL® OD, CHIRALCEL® OD-H, CHIRALCEL® OF, CHIRALCEL® OG, CHIRALCEL® OJ and CHIRALCEL® OK.
[00194] General processes for preparing compounds of the instant invention are provided as further embodiments of the invention and are illustrated in the following Schemes.
Synthesis of 3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3-methylpyrrolidin-l- yl]ethoxy}phenyl)-2H-chromen-7-ol (OP-1038) and Separation and Purification of
-1074 and OP-1075)
Figure imgf000053_0001
Step 1: Reaction to produce l-(2,4-Dihydroxyphenyl)-2-(4-hydroxyphenyl)ethanone
[00195] Resorcinol (1,3-dihydroxybenzene) (62.000 g, 563.1 mmol, 1.0 equiv.) and 4-
Hydroxyphenyl acetic acid (94.237 g, 619.4 mmol, 1.1 equiv.) were added to a 3 neck 2 L round bottomed flask fitted with a paddle, a pressure equalizing addition funnel and a thermometer and a heating mantle. Toluene (350 mL) was added to the flask to give a suspension. The reaction purged with nitrogen and the addition funnel filled with Boron trifluoride etherate (198.201 ml, 1578.0 mmol, 2.8 equiv.) via canula. The reaction was stirred at 150 rpm and boron trifluoride etherate was added in portions of 3-4 mL and the reaction heated. During addition the internal temperature rose to 100 °C. The reaction went through various changes in color from yellow to dark red. After complete addition of boron trifluoride etherate the addition funnel was removed and replaced with a condenser. The reaction was stirred for 1.5 h at an internal temperature of 108 °C. A sample was taken and HPLC analysis indicated the reaction was complete. The reaction was cooled and stirring stopped to give a biphasic solution. A 12 % aqueous solution of sodium acetate (41 g, 336 mL) was slowly added to the reaction with stirring. The reaction was stirred for 16 hours. A precipitate formed overnight and was collected in a sintered glass funnel. The solid was dried on a vacuum oven for 16 h to give the product as a white powder (119.67 g, 87.0 %).
Step 2:
Reaction to produce l-(2-hydroxy-4-((tetrahydro-2H-pyran-2-yl)oxy)phenyl)-2-(4- ((tetrahydro-2H-pyran-2-yl)oxy)phenyl)ethanone
[00196] l-(2,4-Dihydroxyphenyl)-2-(4-hydroxyphenyl)ethanone (119.000 g, 487.2 mmol,
1.0 equiv.) and ethyl acetate (400 mL) was added to a 2 L 3 neck round bottomed flask equipped with a stir bar a thermometer, a condenser and a nitrogen inlet. The flask was flushed with nitrogen for 2 minutes and 3,4-dihydro-2H-pyran (222.252 ml, 2436.1 mmol, 5.0 equiv.) was added from a graduated cylinder. The suspension was flushed with nitrogen for 2 minutes and p- toluenesulfonic acid (0.378 g, 2.2 mmol, 0.0 equiv.) was added to the reaction. An exothermic reaction took place and the temperature rose from 20 to 33 °C over 5 minutes. The yellow suspension became a red solution within 1 minute of PTSA addition. The reaction was stirred for 66 h at room temperature. The reaction was monitored by HPLC at 4, 5 and 6 hours. The chromatograms indicated the reaction was 74 %, 90 % and 100 % complete at the time indicated respectively. TEA (5 mL) was added to the cream colored slurry to stop the reaction. The slurry was transferred to a round bottomed flask (2 L) and the three neck flask rinsed with ethyl acetate. The slurry was concentrated on a rotovap to give a cream colored powdery solid. The solid was transferred to a 2 L Erlenmeyer flask. Isopropyl alcohol (IPA) was used to rinse the flask. The solid was recrystallized from IPA (1.4 L). The suspension was cooled in an ice bath for 30 minutes and the solid collected by vacuum filtration. The solid was rinsed with ice cold IPA until the filtrate was colorless and dried in a vacuum oven to give a white powder (162.24 g). The mother liquor and washes were combined and concentrated to orange oil (38.09 g).
Step 3:
Reaction to produce 2-(4-iodophenyl)-7-((tetrahydro-2H-pyran-2-yl)oxy)-3-(4-((tetrahydro-
2H-pyran-2-yl)oxy)phenyl)chroman-4-one
[00197] l-(2-hydroxy-4-((tetrahydro-2H-pyran-2-yl)oxy)phenyl)-2-(4-((tetrahydro-2H- pyran-2-yl)oxy)phenyl)ethanone (16.228 g, 39.34 mmol) was added to a 3 neck 1 L RB flask. 2- Butanol (380 mL, 0.197 M) and 4-iodobenzaldehyde (51.700 g, 222.8 mmol, 1.0 equiv.) was added to the flask to give a suspension. Piperidine (7.300 ml, 73.9 mmol, 0.3 equiv.) and 1,8- Diazabicyclo[5.4.0]undec-7-ene (11.300 ml, 75.6 mmol, 0.3 equiv.) was added to the suspension. The flask was fitted with a Dean-Stark apparatus and condenser, a thermometer, a stirrer shaft and heated in an oil bath at 130 °C to give an orange solution (became a solution when the internal temperature was 80 °C). Half the solvent (190 mL) was collected over 1.5 hours. The Dean-Stark trap was removed and the condenser was placed on the flask the reaction heated for a further 1 hour. The solution gradually darkens to an orange color. The oil bath was cooled to 90 °C and 380 mL of isopropyl alcohol was added in one portion. The reaction mixture became a cloudy white suspension and redissolved to give a solution in less than a minute at 90 °C. The heating to the bath was set to 50 °C and the flask was allowed to gradually cool to 50 °C. A precipitate started to form at 60 °C and gave a suspension at 50 °C. A thick oily mass falls out of solution ~ 55-53 °C. Vigorous agitation with overhead stirrer (300 rpm) was required to prevent the oily mass from solidifying into one solid as seen with small scale reactions equipped with stir bar. The reaction was left to stir until the mixture cooled to room temperature. The oily mass solidified into a cake even with vigorous agitation. The mother liquor was decanted and fresh isopropanol (100 mL) was added to the flask to rinse the solid. The liquid was decanted and combined with the mother liquor. The mother liquor was concentrated to a dark red oil (27.13 g) and DCM (150 mL) was added to the flask to give a red solution. Silica gel (55 g) was added to solution and concentrated to dryness. The silica gel mixture was poured into a 600 mL sintered glass funnel filled with silica gel (50 g). The solids were washed with ethyl acetate (1.2 L) and the filtrate concentrated to an orange oil (137.61 g crude). The oil was dissolved into boiling 80 % IP A/water (1.2 L) and the solution allowed to cool to room temperature and stand overnight to give a cake. The cake was filtered and washed with cold IPA (100 mL). The mother liquor was partially concentrated on a rotovap to give a tan powder. This process was repeated until an oil could not be washed away from the powder. The product was pooled and dried in a vacuum oven to give an impure tan powder (118.25 g, 85.6 %).
Step 4a:
Reaction to produce 2-(4-iodophenyl)-4-methyl-7-((tetrahydro-2H-pyran-2-yl)oxy)-3-(4- ((tetrahydro-2H-pyran-2-yl)oxy)phenyl)chroman-4-ol
[00198] To a solution of 90.0% 2-(4-iodophenyl)-7-((tetrahydro-2H-pyran-2-yl)oxy)-3-(4-
((tetrahydro-2H-pyran-2-yl)oxy)phenyl)chroman-4-one (104.891 g, 150.7 mmol, 1.0 equiv.) in THF (1.2 L) at 5°C, was added Methylmagnesium chloride 3.0 M solution in THF (160.000 ml, 480.0 mmol, 3.2 equiv.) by addition funnel over 30 minutes. The temperature did not rise about 8 °C during the addition. The reaction was removed from the ice bath and stirred at room temperature and stirred for another hour. TLC (20% ethyl acetate in hexanes) showed the reaction had no starting material. The solution was cooled in an ice bath, and carefully quenched with saturated ammonium chloride (35 mL). Ethyl acetate (1.2 L) and water (1.2 L) were added to the reaction mixture, and the layers were separated. The aqueous layer was extracted with EA (1 L). The combined organic layer was washed with brine (1 L), dried over anhydrous Na2S04, filtered and concentrated in vacuo to yield a pale yellow foam (111.26 g crude). This material was used without further purification.
Step 4b:
Reaction to produce 3-(4-hydroxyphenyl)-2-(4-iodophenyl)-4-methyl-2H-chromen-7-ol
[00199] 2-(4-iodophenyl)-4-methyl-7-((tetrahydro-2H-pyran-2-yl)oxy)-3-(4-((tetrahydro- 2H-pyran-2-yl)oxy)phenyl)chroman-4-ol (96.820 g, 150.7 mmol, 1.0 equiv.) and 80% acetic acid in H20 (686 mL) was added to a 2 L RB flask. The suspension was degassed, flushed with nitrogen and heated at 90 °C for 1.5 hours. TLC analysis (1 :2 EA/Hex) of the reaction showed no starting material was present. The solvent was removed to give a red oil. The red oil was dissolved into ethyl acetate (500 mL) and washed with saturated sodium bicarbonate solution (3 x 1 L). The organic layers was washed with brine (1 L), filtered and concentrated to give a red oil (109.32 g, crude). The oil was loaded onto 100 g of silica gel and chromatographed in 40 g portions on silica gel (100 g cartridge, 5-30 % EA/Hex). Fractions containing spots with Rf 0.55 (33 % EA/Hex) were pooled and concentrated to a light red glass (53.37 g). The glass was mixed with DCM (200 mL) and sonicated to give a pink suspension. The solid was filtered through a sintered glass funnel washed with a 20 % DCM in Hexanes solution (250 mL) and dried in a vacuum oven overnight (32.41 g). The mother liquor was concentrated to a glass and the process repeated a second time to give a pink solid (4.2784 g). The impure mixed fractions were pooled and concentrated to a glass (16.71 g). The glass was dissolved into DCM (75 mL) and pink crystals formed on standing (7.0862 g). This process was repeated to give a second crop of pink crystals (2.3643). The mother liquors from both the pure and impure fractions were combined and chromatographed with the same method (2 x 100 g cartridges). The fractions with Rf 0.55 were pooled and concentrated to give a red oil (17.388 g) which did not solidify. The oil was not combined with previous batches but reprotected in a separate reaction.
[00200] Gradient method: (5-30 % EA/Hex) 5 % EA hold for 2 minutes, gradient to 15 % over 3 minutes and hold at 15 % EA/ Hex for 7 minutes, gradient to 30 % over 7 minutes and hold at 30 % EA/ Hex for 17 minutes. Fractions with Rf 0.55 (33 % EA/Hex) were pooled and concentrated to a light pink oil which was triturated with DCM.
Step 5:
Reaction to produce 2-(4-iodophenyl)-4-methyl-7-((tetrahydro-2H-pyran-2-yl)oxy)-3-(4- ((tetrahydro-2H-pyran-2-yl)oxy)phenyl)-2H-chromene
[00201] To a solution of 3-(4-hydroxyphenyl)-2-(4-iodophenyl)-4-methyl-2H-chromen-7- ol (41.860 g, 91.7 mmol, 1.0 equiv.) and pyridinium para-toluene sulfonate (4.822 g, 19.3 mmol, 0.2 equiv.) in DCM (200 mL) was added 3,4-dihydro-2H-pyran (49.226 ml, 539.6 mmol, 5.9 equiv.). The reaction was stirred at room temperature overnight (17 h). TLC showed major desired product. The reaction was diluted with DCM (200 mL), washed with saturated NaHC03 (200 mL), water (200 mL), brine (200 mL), dried over Na2S04, filtered and concentrated to give a red viscous residue. The residue adsorbed onto silica gel (75 g) was purified on a silica gel column (4 x 100 g, 0 - 20 % EA/Hex) to give a white solid which was triturated with methanol and dried in a vacuum oven at 40° C for 16 h to afford the titled compound as a white powder (51.67 g 90.2 %).
[00202] 1H MR (300 MHz, CDC13 ): δ 7.53 (d, J = 5.4 Hz, 2H)), 7.18 (d, J = 8.7 Hz,
1H), 7.06 (parent t, J = 7.8 Hz, 4H), 6.71 (s, 1H), 6.59 (d, J = 2.4 Hz, 1H), 6.45 (d, J = 2.4 Hz, 1H), 5.15 (s, 2H), 4.59 (s, 2H), 4.63 (d, J = 5.7 Hz, 2H), 3.98 (s, 3H), 3.84 (s, 3H).
Step 6:
Reaction to produce -(3R)-3-methyl-l-(2-(4-(4-methyl-7-((tetrahydro-2H-pyran-2-yl)oxy)-3- (4-((tetrahydro-2H-pyran-2-yl)oxy)phenyl)-2H-chromen-2-yl)phenoxy)ethyl)pyrrolidine
[00203] A mixture of 2-(4-iodophenyl)-4-methyl-7-((tetrahydro-2H-pyran-2-yl)oxy)-3-(4-
((tetrahydro-2H-pyran-2-yl)oxy)phenyl)-2H-chromene (16.800 g, 26.9 mmol, 1.0 equiv.), (R)~2- (3-methylpyrrolidin-l-yl)ethanol (10.416 g, 80.6 mmol, 3.0 equiv.), 1,10-Phenanthroline (0.970 g, 5.4 mmol, 0.2 equiv.), and Cesium carbonate (17.530 g, 53.8 mmol, 2.0 equiv.) in butyronitrile (84 mL) was charged into a 250 mL round bottom flask which was evacuated and backfilled with argon (3 x), Copper(I) iodide (5.123 g, 26.9 mmol, 1.0 equiv.) was added to the suspension and evacuated and backfilled with argon (3 x). The reaction mixture was heated in an oil bath at 120 °C. After 91 h of heating the reaction was cooled to room temperature and the mixture filtered through a pad of Celite (3 cm) which was successively washed with DCM (200 mL), EA (200 mL) and MeOH (200 mL). The filtrate was collected and concentrated. The residue was adsorbed onto silica gel (25 g) purified with silica gel (100 g cartridge, 0 - 30% MeOH/DCM) [TLC: 5 % MeOH/DCM, 4 major spots, Rf (SM:0.95), 0.9, 0.83, (prod. 0.43)]. The fractions containing product were pooled and concentrated to give a brown foam (13.64 g, 81.0 %).
[00204] Gradient method 0 % MeOH 4 minutes, gradient to 1 % MeOH/DCM over 3 minutes, hold at 1 % MeOH/DCM for 10 minutes, gradient to 5 % MeOH/DCM over 3 minutes, hold at 5 % MeOH/DCM for 12 minutes, gradient to 25 % MeOH/DCM over zero minutes, hold at 25 % MeOH/DCM for 15 minutes. Many fractions contained a mixture of the starting material and product all fractions were pooled, concentrated, and rechromatographed on silica gel (loaded onto 15 g and 100 g cartridge) and gradient eluted with this gradient method (0 % MeOH 4 minutes, gradient to 1 % MeOH/DCM over 3 minutes, hold at 1 % MeOH/DCM for 20 minutes, gradient to 5 % MeOH/DCM over 5 minutes, hold at 5 % MeOH/DCM for 20 minutes).
Fractions 74 to 126 were pooled and concentrated to a brown oil which solidified to foam (13.64 g, 81 %) (Late eluting fractions from the first column contained a spot which corresponded to the amino alcohol. These fractions were pooled and concentrated to give a red black liquid (6.38 g).
Step 7:
Reaction to produce 3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3-methylpyrrolidin-l- yl] ethoxy}phenyl)-2H-chromen-7-ol (OP- 1038)
[00205] (3R)-3 -methyl- l-(2-(4-(4-methyl-7-((tetrahydro-2H-pyran-2-yl)oxy)-3 -(4-
((tetrahydro-2H-pyran-2-yl)oxy)phenyl)-2H-chromen-2-yl)phenoxy)ethyl)pyrrolidine (15.130 g, 24.2 mmol, 1.0 equiv.) was dissolved into 80% acetic acid/water (150 mL). The solution was heated in an oil bath at 90 °C for 1 hour. HPLC analysis of the reaction mixture indicated the reaction was complete. The dark red solution was concentrated to a dark red oil. The oil was suspended into ethyl acetate (600 mL) and washed with saturated NaHC03 (3 x 300 mL). The combined aqueous layer was extracted with ethyl acetate (2 x 100 mL). The combined organic layer was washed with brine (2 x 200 mL), dried over anhydrous magnesium sulfate, filtered and concentrated to give a red oil (14.03 g, crude). The oil was adsorbed onto silica gel (30 g) and chromatographed on silica gel (2 x 100 g cartridge) with 0-10 % MeOH in DCM. Fractions containing the product were pooled and concentrated to give a red colored foam (6.68 g). Impure fractions were concentrated and repurified with the same conditions to give an additional 0.9496 g of red foam which was combined with the previous foam. Total yield 7.6296 g, 69.0 %.
[00206] Gradient method 0 % MeOH/DCM for 5 minutes, gradient to 10 % MeOH/DCM over 20 minutes, hold at 10 % MeOH/DCM for 10 minutes. TLC conditions (UV and 12): 10 % MeOH/DCM 5 spots 0.64, 0.48, (product) 0.31, 0.21, 0.07. HPLC: 100 % purity (0 - 90% acetonitrile/water). LC_MS: [M+l]+ = 458.3.
Step 8:
[00207] OP- 1038 was separated into its diastereomers (2S)-3-(4-hydroxyphenyl)-4- methyl-2-(4-{2-[(3R)-3-methylpyrrolidin-l-yl]ethoxy}phenyl)-2H-chromen-7-ol (OP-1074) and (2R)-3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3-methylpyrrolidin-l-yl]ethoxy}phenyl)-2H- chromen-7-ol (OP-1075) using a Diacel, Chiralpak IC column at room temperature in isocratic mode with 80 % hexanes, 20 % 2-propanol with 0.1 % dimethylethylamine or 0.1% diethyl amine as a modifier. This method was used at analytical and preparative scale. Step 9:
Reaction to produce (R)-2-(benzyloxy)-l-(3-methylpyrrolidin-l-yl)ethanone
[00208] (R)-3-methylpyrrolidine hydrochloride (20.000 g, 164.5 mmol, 1.0 equiv.) was added to a round bottom flask and dissolved into anhydrous DCM (45 mL). Freshly distilled Diisopropylethylamine (60.157 ml, 345.4 mmol, 2.1 equiv.) and freshly activated 4 A molecular sieves (- 21 g) was added to the solution and stirred for 10 minutes. 2-(Benzyloxy)acetyl chloride (31.881 g, 172.7 mmol, 1.1 equiv.) dissolved into DCM (50 mL) was added to the reaction at room temperature dropwise via syringe over 20 minutes with a room temperature water bath for cooling. After complete addition the reaction was stirred for 17 hours. TLC analysis (1 : 1, EA/Hex, Rf: 0.83, 0.33, 0.05) showed no presence of acid chloride. The reaction poured into a separatory funnel and the organic layer washed successively with 1 M HCl (2 x 200 mL), saturated sodium bicarbonate (200 mL) and brine (200 mL). The organic layer was dried over anhydrous MgS04, filtered and concentrated to an orange oil (42.40 g). The oil was loaded onto silica gel (30 g) and the mixture split into -18 g portions and chromatographed on silica gel (4 x 100 g cartridges) with a gradient method 10-80 % EA/Hex. Fraction with Rf 0.33 spot were pooled and concentrated to give a yellow oil (34.02 g, 88.7 %).
[00209] Gradient method: 10 % EA/Hex hold 5 minutes, gradient to 80 % EA/Hex over 15 minutes, hold at 80 % EA/Hex for 10 minutes. Fractions with Rf 0.33 were pooled and concentrated.
Step 10:
Reaction to produce (R)-l-(2-(benzyloxy)ethyl)-3-methylpyrrolidine
[00210] Aluminum trichloride (54.513 g, 408.8 mmol, 3.0 equiv.) was dissolved into anhydrous THF (750 mL) and cooled in an ice bath. Lithium aluminum hydride (35.688 g, 940.3 mmol, 6.9 equiv.) was added in small portions via a powder addition funnel to the above suspension over 35 minutes and stirred for an additional 10 minutes. The suspension was cooled to -78 °C for 15 minutes and a solution of (R)-2-(Benzyloxy)-l-(3-methylpyrrolidin-l- yl)ethanone (33.980 g, 136.3 mmol, 1.0 equiv.) in anhydrous THF (150 mL) was added dropwise to the cold suspension via a pressure equalizing addition funnel over 20 minutes. The reaction was kept at -78 °C for 1 hour and stirred at room temperature for 1 hours. The reaction was carefully quenched with 6 N HC1 solution (100 mL) and stirred for 17 h to give grey suspension. A solution 6 N NaOH (216 mL) was added to the mixture to give a white suspension after stirring for 30 minutes. The mixture was filtered through a pad of Celite (4 cm). The solids were washed with DCM (5 x 500 mL). The filtrate was poured into a separatory funnel and the layers separated (-200 mL aqueous layer recovered). The aqueous layer was extracted with DCM (3 x 100 mL). The organic layers were combined and washed with brine (500 mL), dried over anhydrous sodium sulfate, filtered and concentrated to a yellow liquid (33.43 g). This liquid was loaded onto silica gel (25 g) and chromatographed through silica gel (2 x 100 g cartridge) with 50-100 % ethyl acetate in hexanes followed by 10-40 % methanol in dichloromethane to give a yellow oil (29.17 g, quant).
[00211] Gradient method: 50 % EA/Hex 4 minutes, gradient to 100 % EA over 6 minutes, hold at 100 % EA for 5 minutes, Solvent change to 10 % MeOH in DCM hold for 0 minutes, gradient to 40 % MeOH in DCM over 1 minute, hold at 40 % MeOH in DCM for 8 minutes. The fractions were pooled and concentrated to a yellow oil (29.17 g, quant).
Step 11:
[00212] (R)-l-(2-(benzyloxy)ethyl)-3-methylpyrrolidine (10.000 g, 45.6 mmol, 1.0 equiv.)
(0.4822 g; 0.7137 g) was added to a 400 mL Parr flask, methanol (60 mL) was added and the solution cooled in an ice bath for 10 minutes. 20% Pd(OH)2 on Carbon, 50 % H20 (6.403 g, 45.6 mmol, 1.0 equiv.) was added to the cooled solution and flushed with nitrogen. Hydrochloric acid (6 M, 7.6 mL) was added to mixture. The flask was pressurized with hydrogen to 30 psi shaken for 1 minute and the hydrogen released. This was repeated twice more and pressurized to 100 psi with hydrogen. This suspension was shaken for 16 hours. A sample was taken and the TLC (10 % MeOH in DCM) indicated the reaction was incomplete and additional catalyst (2.0 g) was added to the mixture. The reaction was treated in a similar manner described above and shaken on the hydrogenator for an additional 30 hours. Celite (5 g) was added to the Parr flask and the mixture filtered through a pad of Celite (2 cm). The solid was washed wth methanol (2 x 250 mL). The filtrate was concentrated on a rotovap to dryness to give a red oil (7.81 g). The oil was taken up in methanol (50 mL) and 25 % sodium methoxide in methanol (9.9 mL, 45.5 mmol, 1 equiv) was added to the methanolic solution to give a white suspension. The mixture was concentrated to dryness and taken up into anhydrous DCM (35 mL). The suspension was centrifuged at 3K rpm for 5 minutes. The clear solution was collected and the solid resuspended into DCM (35 mL). This process was repeated a total of 4 times. The combined solution was concentrated to a yellow liquid (5.6341 g, 95.6 %).
[00213] Following the general method above and using the appropriate reagents and starting materials, OP- 1046 and OP- 1047 were synthesized.
Synthesis of HC1 Salts of OP-1038 and OP-1074
[00214] 3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3-methylpyrrolidin-l- yl]ethoxy}phenyl)-2H-chromen-7-ol (OP-1038; 0.020 g, 0.0 mmol, 1.0 equiv.) or (2S)-3-(4- hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3 -methyl pyrrolidin-1 -yl]ethoxy}phenyl)-2H-chromen- 7-ol (OP-1074; 0.020 g, 0.0 mmol, 1.0 equiv.) (Compound 33) was placed into a 1 dram vial and dissolved into methanol (0.2 mL). 4 M HC1 in methanol (200 μΐ.) was added to the solution and stirred for 15 minutes. The yellow solution was concentrated a yellow orange solid (0.022 g and 0.0206 respectively).
Synthesis of OP-1083
[00215] OP-1083 was prepared by air oxidation on OP-1074, followed by
chromatographic separation of OP-1083 and OP-1074. The mixture of OP-1074 and OP-1083 (560 mg) was dissolved in methanol (15 mL) and mixed with silica gel (3 g). The mixture was dried to give a dark red powder. This powder was loaded into a cartridge and chromatographed on silica gel (4 g cartridge) with 0-25 % methanol in dichlorom ethane to give OP-1074 as an orange solid (0.261 g, 46.6 %) and OP-1083 as an orange solid (41.1 mg, 15 %)
[00216] Method: 0 % MeOH for 4 min, gradient to 5 % MeOH/DCM over 5 minutes, hold at 5 % for 6 minutes, gradient to 10 % MeOH/DCM over 2 minutes, hold at 10 % MeOH/DCM for 8 minutes, gradient to 25 % MeOH/DCM over 0 minutes, hold at 25 % for 5 minutes.
Synthesis of OP-1084
[00217] (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3-methylpyrrolidin-l- yl]ethoxy}phenyl)-2H-chromen-7-ol (0.020 g, 0.0 mmol, 1.0 equiv.) was added to a 30 mL vial and suspended into anhydrous ethyl acetate (20 mL). Diisopropylethylamine (19 ul, 0.1 mmol, 2.5 equiv.) was added to the suspension and the solution was cooled in an ice bath for 5 minutes. Ethyl chloroformate (10 ul, 0.1 mmol, 2.3 equiv.) was added to the reaction via a gas tight syringe. The reaction immediately became a cloudy white suspension. The reaction was removed from the ice bath and stirred at room temperature for 16 h. The reaction was concentrated to dryness and dissolved into a minimum of DCM to load onto a 4 g silica gel cartridge. The crude material was eluted with 0-15 % MeOH/DCM to give the desired product as a pale yellow film (0.006.8 g, 27 %).
[00218] TLC (5 % MeOH/DCM): 4 spots, 0.84, 0.42, 0.26 (Product), 0.16 (Mono carbonate). Gradient method: 0 % MeOH/DCM hold for 2 minutes, gradient to 5 % MeOH/DCM over 5 minutes, hold at 5 % MeOH/DCM for 3 minutes, gradient to 15 % MeOH/DCM over 3 minutes, hold at 15 % MeOH/DCM for 2 minutes. Fractions 16-19 pooled: 6.8 mg LCMS (m/z): 602; HPLC (254 nm): 95.65 %.
Synthesis of OP-1085
[00219] (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3-methylpyrrolidin-l- yl]ethoxy}phenyl)-2H-chromen-7-ol (0.035 g, 0.1 mmol, 1.0 equiv.) was added to a dry 1 dram vial equipped with a stir bar under a stream of N2. The vial was sealed with a septum and freshly distilled Pyridine (400 ul, 5.0 mmol, 113.6 equiv.) was added to the vial via syringe to give pink red solution. The vial was cooled in a 0 °C ice/water bath for 10 minutes and Trimethylacetyl Chloride (100 ul, 0.8 mmol, 10.6 equiv.) was added via a GC syringe in one portion to the solution. The solution immediately became a light yellow color and was stirred for 30 minutes at 0 °C. The reaction was allowed to reach room temperature over 30 min and stirred at room temperature for 1 h. A sample was analyzed by LCMS to show the presence of a mixture of mono ester and diester. The reaction mixture was concentrated to dryness and dissolved into a minimum amount of DCM and chromatographed on silica gel (4 g cartridge) with 0-25
MeOH/DCM. The fractions containing product were pooled and concentrated to a give a pale yellow film (9.2 mg, 33 %).
[00220] Gradient: 0 % MeOH/DCM for 2 minutes, gradient to 2.5 % MeOH/DCM over 4 minutes, hold at 2.5 % MeOH/DCM for 5 minutes, gradient to 10 % MeOH/DCM over 1 minute, hold at 10 % MeOH/DCM for 4 minutes, gradient to 25 % MeOH/DCM over 2 minutes, hold at 25 % for 7 minutes. Fractions 11-17 and 19-28 pooled. LCMS (m/z): M+l, 626; HPLC (254 nm): 98.0 %. Synthesis of OP-1086 and OP-1088
[00221] Pyridine (2000 ul, 24.8 mmol, 113.6 equiv.) was added to a dry 1 dram vial with a stir bar. The vial was cooled in a -15 °C dry ice methanol/water bath. Phosphorus oxychloride (71 ul, 0.8 mmol, 3.5 equiv.) was added via a GC syringe in one portion to the solution. The solution was stirred for 5 minutes then the solid 3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3- methylpyrrolidin-l-yl]ethoxy}phenyl)-2H-chromen-7-ol (OP-1038; 0.100 g, 0.2 mmol, 1.0 equiv.) or (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3-methylpyrrolidin-l- yl]ethoxy}phenyl)-2H-chromen-7-ol (OP- 1074; 0.100 g, 0.2 mmol, 1.0 equiv.) was added in one portion under a stream of N2. The reaction stirred at this temperature for 1.5 h. The solution became slurry after -45 minutes. The reaction was allowed to reach room temperature over 45 min and stirred at room temperature for 2 h. A sample was quenched with water and analysis showed the mass of the desired product. The reaction mixture was concentrated to dryness. The crude mixture was suspended into 2 N HC1 (5 mL). The suspension was sonicated and centrifuged at 5000 rpm for 6 minutes. The supernatant was decanted and the solid resuspended into 5 mL of 2 N HC1 and the process was repeated. The solid was dried under vacuum to give 129 mg of the crude product. The solid was suspended into water (2 mL) and 6 N sodium hydroxide solution (174 μL, 1 mmol, 5 equiv) was added to give an orange solution. This was purified by Preparative LC with acetonitrile and water to give the product as a tan solid.
Synthesis of OP-1087
[00222] (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3-methylpyrrolidin-l- yl]ethoxy}phenyl)-2H-chromen-7-ol (0.020 g, 0.0 mmol, 1.0 equiv.) was added to a 30 mL vial and suspended into anhydrous ethyl acetate (20 mL). Diisopropylethylamine (19 ul, 0.1 mmol, 2.5 equiv.) was added to the suspension and the solution was cooled in an ice bath for 5 minutes. Methyl chloroformate (7 ul, 0.1 mmol, 2.2 equiv.) was added to the reaction via a gas tight syringe. The reaction immediately became a cloudy white suspension. The reaction was removed from the ice bath and stirred at room temperature for 16 h. The reaction was concentrated to dryness and dissolved into a minimum of DCM to load onto a 4 g silica gel cartridge. The crude material was eluted with 0-15 % MeOH/DCM to give the desired product as a pale yellow film (0.0096 g, 40 %). [00223] TLC (5 % MeOH/DCM): 4 spots, 0.95, 0.55, 0.38 (Product), 0.25 (Mono carbonate).
[00224] Gradient method: 0 % MeOH/DCM hold for 2 minutes, gradient to 5 %
MeOH/DCM over 5 minutes, hold at 5 % MeOH/DCM for 3 minutes, gradient to 15 %
MeOH/DCM over 3 minutes, hold at 15 % MeOH/DCM for 2 minutes. Fractions 11-15 pooled: 9.6 mg. LCMS (m/z): 574; HPLC (254 nm): 95.08 %.
ASSAYS
[00225] Compounds provided herein can be evaluated using various in vitro and in vivo assays; examples of which are described below.
[00226] The following biological examples are offered to illustrate the compounds, pharmaceutical compositions and methods provided herein and are not to be construed in any way as limiting the scope thereof.
Demonstration of the Superiority of OP-1074 Combined With Chemotherapeutics Using
Sensitive In vitro Proliferation Assays
[00227] Proliferation in MCF-7 was measured using a fluorescent DNA binding dye 6-8 days after treatment in triplicate with anti-estrogens in the presence of 100 pM E2. Specifically, MCF-7 cells were treated with anti-estrogens alone or in combination with other
chemotherapeutics in hormone-depleted medium for 6-8 days in the presence of 100 pM E2. Proliferation was measured using Cyquant fluorescent DNA-binding dye (Invitrogen, Grand Island, NY). Results were from a single representative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing SEM.
[00228] As shown in Figure 1, OP-1074 increases the potency and efficacy of the CDK
4/6 inhibitor PD-0332991 in inhibiting E2-stimulated MCF-7 breast cell proliferation. The antiestrogen OP-1074 enhances the efficacy and potency of PD-0332991 in inhibiting E2- stimulated breast cell proliferation in vitro. MCF-7 cells were treated with the indicated amount of antiestrogen and PD-0332991 in hormone-depleted medium for 6-7 days in the presence of 100 pM 17p-estradiol (E2). Fig. 1 A plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or ΙΟΟηΜ OP-1074 in combination with increasing amounts of PD-0332991. Fig. IB plots the effects on MCF-7 cell proliferation of vehicle or ΙΟΟηΜ PD-0332991 in combination with increasing amounts of OP-1074. Fig. 1C plots the effects on MCF-7 cell proliferation of vehicle or 3nM OP-1074, tamoxifen, or fulvestrant in combination with increasing amounts of PD- 0332991. As can be seen from these experiments, the combination of OP-1074 with PD- 0332991 at several concentrations leads to a lower percentage of proliferation of MCF-7 breast cells than either agent by themselves.
[00229] As shown in Figure 2, OP-1074 increases the potency of the mTOR inhibitor everolimus in inhibiting E2-stimulated MCF-7 breast cell proliferation. The antiestrogen OP- 1074 enhances the efficacy and potency of the mTOR inhibitor everolimus in inhibiting E2- stimulated breast cell proliferation in vitro. MCF-7 cells were treated with the indicated amount of OP-1074 and everolimus in hormone-depleted medium for 6 days in the presence of 100 pM 17P-estradiol (E2). Fig. 2 A plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or ΙΟΟηΜ OP-1074 in combination with increasing amounts of everolimus. Fig. 2B plots the effects on MCF-7 cell proliferation of vehicle, InM or lOnM everolimus in combination with increasing amounts of OP-1074. Fig. 2C plots the effects on MCF-7 cell proliferation of vehicle or 3nM OP- 1074, tamoxifen, or fulvestrant in combination with increasing amounts of everolimus. As can be seen from these experiments, the combination of OP-1074 with everolimus at several concentrations leads to a lower percentage of proliferation of MCF-7 breast cells than either agent by themselves.
[00230] As shown in Figure 3, OP-1074 increases the potency of the pan PI3K inhibitor
BKM-120 in inhibiting E2-stimulated MCF-7 breast cell proliferation. The antiestrogen OP- 1074 enhances the efficacy and potency of the PI3K inhibitor BKM-120 in inhibiting E2- stimulated breast cell proliferation in vitro. MCF-7 cells were treated with the indicated amount of OP-1074 and BKM-120 in hormone-depleted medium for 7 days in the presence of 100 pM 17P-estradiol (E2). Fig. 3 A plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or ΙΟΟηΜ OP-1074 in combination with increasing amounts of BKM-120. Fig. 3B plots the effects on MCF-7 cell proliferation of vehicle, 32nM or ΙΟΟηΜ BKM-120 in combination with increasing amounts of OP-1074. As can be seen from these experiments, the combination of OP- 1074 with BKM-120 at several concentrations leads to a lower percentage of proliferation of MCF-7 breast cells than either agent by themselves.
[00231] As shown in Figure 4, OP-1074 increases the potency of 5-fluorouracil in inhibiting E2-stimulated MCF-7 breast cell proliferation. The antiestrogen OP-1074 enhances the efficacy and potency of the thymidylate synthase inhibitor 5-fluorouracil in inhibiting E2- stimulated breast cell proliferation in vitro. MCF-7 cells were treated with the indicated amount of OP-1074 and 5-fluorouracil in hormone-depleted medium for 6 days in the presence of 100 pM 17p-estradiol (E2). The effects on MCF-7 cell proliferation of vehicle, 1, 10 or ΙΟΟηΜ OP- 1074 in combination with increasing amounts of 5-fluorouracil are plotted. As can be seen from these experiments, the combination of OP- 1074 with 5-fluorouracil at several concentrations leads to a lower percentage of proliferation of MCF-7 breast cells than either agent by themselves.
[00232] As shown in Figure 5, OP-1074 increases the potency of taxanes in inhibiting El- stimulated MCF-7 breast cell proliferation. The antiestrogen OP-1074 enhances the efficacy and potency of paclitaxel and docetaxel in inhibiting E2-stimulated breast cell proliferation in vitro. MCF-7 cells were treated with the indicated amount of OP-1074 and taxane in hormone-depleted medium for 6 days in the presence of 100 pM 17P-estradiol (E2). Fig. 5 A plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or ΙΟΟηΜ OP-1074 in combination with increasing amounts of paclitaxel. Fig. 5B plots the effects on MCF-7 cell proliferation of vehicle, 1, 10 or ΙΟΟηΜ OP-1074 in combination with increasing amounts of docetaxel. As can be seen from these experiments, the combination of OP-1074 with paclitaxel or docetaxel at several concentrations leads to a lower percentage of proliferation of MCF-7 breast cells than either agent by themselves.
[00233] As shown in Figure 6, OP-1074 increases the potency of the anthracycline doxorubicin in inhibiting E2-stimulated MCF-7 breast cell proliferation. The antiestrogen OP- 1074 enhances the efficacy and potency of the anthracycline doxorubicin in inhibiting El- stimulated breast cell proliferation in vitro. MCF-7 cells were treated with the indicated amount of OP-1074 and Doxorubicin in hormone-depleted medium for 7 days in the presence of 100 pM 17p-estradiol (E2). The effects on MCF-7 cell proliferation of vehicle, 1, 10 or ΙΟΟηΜ OP-1074 in combination with increasing amounts of doxorubicin are plotted. As can be seen from these experiments, the combination of OP-1074 with doxorubicin at several concentrations leads to a lower percentage of proliferation of MCF-7 breast cells than either agent by themselves.
DEFINITIONS
[00234] Compounds described herein can comprise one or more asymmetric centers, and thus can exist in various isomeric forms, e.g., enantiomers and/or diastereomers. For example, the compounds described herein can be in the form of an individual enantiomer, diastereomer or geometric isomer, or can be in the form of a mixture of stereoisomers, including racemic mixtures and mixtures enriched in one or more stereoisomer. Isomers can be isolated from mixtures by methods known to those skilled in the art, including chiral high pressure liquid chromatography (HPLC) and the formation and crystallization of chiral salts; or preferred isomers can be prepared by asymmetric syntheses. See, for example, Jacques et al, Enantiomers, Racemates and Resolutions (Wiley Interscience, New York, 1981); Wilen et al, Tetrahedron 33 :2725 (1977); Eliel, Stereochemistry of Carbon Compounds (McGraw-Hill, NY, 1962); and Wilen, Tables of Resolving Agents and Optical Resolutions p. 268 (E.L. Eliel, Ed., Univ. of Notre Dame Press, Notre Dame, IN 1972). Unless otherwise stated, the invention encompasses compounds described herein as individual isomers substantially free of other isomers, and alternatively, as mixtures of various isomers.
[00235] The articles "a" and "an" may be used herein to refer to one or to more than one {i.e. at least one) of the grammatical objects of the article. By way of example "an analogue" means one analogue or more than one analogue.
[00236] "Pharmaceutically acceptable salt" refers to a salt of a compound of the invention that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound. In particular, such salts are non-toxic may be inorganic or organic acid addition salts and base addition salts. Specifically, such salts include: (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethane-disulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 4-methylbicyclo[2.2.2]-oct-2-ene-l-carboxylic acid, glucoheptonic acid, 3 -phenyl propionic acid, trimethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, and the like; or (2) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion, or an aluminum ion; or coordinates with an organic base such as ethanolamine, diethanolamine, triethanolamine, N- methylglucamine and the like. Salts further include, by way of example only, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like; and when the compound contains a basic functionality, salts of non toxic organic or inorganic acids, such as
hydrochloride, hydrobromide, tartrate, mesylate, acetate, maleate, oxalate and the like. The term "pharmaceutically acceptable cation" refers to an acceptable cationic counter-ion of an acidic functional group. Such cations are exemplified by sodium, potassium, calcium, magnesium, ammonium, tetraalkyl ammonium cations, and the like (see, e.g., Berge, et al., J. Pharm. Sci. 66(1): 1-79 (Jan.'77).
[00237] "Pharmaceutically acceptable vehicle" refers to a diluent, adjuvant, excipient or carrier with which a compound of the invention is administered.
"Pharmaceutically acceptable metabolically cleavable group" refers to a group which is cleaved in vivo to yield the parent molecule indicated herein.
[00238] "Prodrugs" refers to compounds, including derivatives of the compounds of the invention, which have cleavable groups and become by solvolysis or under physiological conditions a compound of the invention that are pharmaceutically active in vivo.
[00239] "Solvate" refers to forms of the compound that are associated with a solvent or water (also referred to as "hydrate"), usually by a solvolysis reaction. This physical association includes hydrogen bonding. Conventional solvents include water, ethanol, acetic acid and the like. The compounds of the invention may be prepared e.g. in crystalline or liquid form and may be solvated or hydrated. Suitable solvates include pharmaceutically acceptable solvates, such as hydrates, and further include both stoichiometric solvates and non-stoichiometric solvates. In certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid. "Solvate" encompasses both solution-phase and isolable solvates. Representative solvates include hydrates, ethanolates and methanolates.
[00240] A "subject" to which administration is contemplated includes, but is not limited to, humans {i.e., a male or female of any age group, e.g., a pediatric subject (e.g, infant, child, adolescent) or adult subject (e.g., young adult, middle-aged adult or senior adult)) and/or a non- human animal, e.g., a mammal such as primates (e.g., cynomolgus monkeys, rhesus monkeys), cattle, pigs, horses, sheep, goats, rodents, cats, and/or dogs. In certain embodiments, the subject is a human. In certain embodiments, the subject is a non-human animal. The terms "human", "patient" and "subject" are used interchangeably herein.
[00241] As used herein the term "enantiomerically pure" or "pure enantiomer" denotes that the compound comprises more than 95% by weight. In alternative embodiments, when specified, the term may refer to more than 96% by weight, more than 97% by weight, more than 98%) by weight, more than 98.5%> by weight, more than 99% by weight, more than 99.2% by weight, more than 99.5% by weight, more than 99.6% by weight, more than 99.7% by weight, more than 99.8% by weight or more than 99.9% by weight, of the enantiomer. The weights are based upon total weight of all enantiomers or stereoisomers of the compound.
[00242] As used herein and unless otherwise indicated, the term "enantiomerically pure R- compound" refers to at least 95% by weight R-compound and at most about 5% by weight S- compound. In alternative embodiments, when specified, the term can refer to at least about 99% by weight R-compound and at most about 1% by weight S-compound or at least about 99.9% by weight R-compound or at most about 0.1% by weight S-compound. In certain embodiments, the weights are based upon total weight of compound.
[00243] As used herein and unless otherwise indicated, the term "enantiomerically pure S- compound" or "S-compound" refers to at least about 95% by weight S-compound and at most about 5% by weight R-compound. In alternative embodiments, when specified, the term can refer to at least about 99% by weight S-compound and at most about 1% by weight R-compound or at least about 99.9% by weight S-compound and at most about 0.1% by weight R-compound. In certain embodiments, the weights are based upon total weight of compound.
[00244] These and other exemplary substituents are described in more detail in the Detailed Description, Examples, and claims. The invention is not intended to be limited in any manner by the above exemplary listing of substituents.

Claims

What is claimed is:
Figure imgf000071_0001
wherein Ri and R2 are independently either:
(i) OH or OR9, (ii) wherein R9 is independently selected from H, halogen (CI, Br, I or F), natural or non- naturally occurring amino acid (bound through either the OC(O)- or C(0)0- (an ester) or the amino (through either -C(0)-N- or -N-C(O)- (an amide linkage)), R10, -OR10, or -SR10 where R10 is -C(=0)RC1, -C(=0)0RC1, -C(=0)SRC1, -C(=0)N(RC1)2; or polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;
-S(=0)2RC1, -S(=0)2ORcl, -S(=0)RC1, -S(=0)ORcl, -P(=0)2RC1, -P(=0)2ORcl, -P(=0)(ORcl)2, -P(=0)(RC1)2, or -P(RC1)(0RC1); or oxygen attached to an oxygen protecting group (to produce OH on administration), sulfur attached to a sulfur protecting group (to produce SH or a disulfide on administration), or nitrogen attached to a nitrogen protecting group (to produce - H- on administration);
and R can be independently selected from hydrogen, polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or two R groups are joined to form an substituted or unsubstituted heterocyclic ring;
or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, stereoisomer, tautomer, or N- oxide thereof,
in combination with an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic.
2. The method of claim 1, wherein the compound is in the form of a pharmaceutically acceptable salt.
3. The method of claim 1, wherein the compound is (2S)-3-(4-hydroxyphenyl)-4-methyl-2- (4-{2-[(3R)-3-methylpyrrolidin-l-yl]ethoxy}phenyl)-2H-chromen-7-ol, which has the chemical structure:
Figure imgf000073_0001
OP-1074
or a pharmaceutically acceptable salt, solvate, hydrate, or N-oxide thereof.
4. A method of treating a disorder modulated, mediated or affected by the estrogen receptor in a subject comprising administering to the subject a compound, which is (2S)-3-(4- hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3 -methyl pyrrolidin-1 -yl]ethoxy}phenyl)-2H-chromen- 7-ol, which has the chemical structure:
Figure imgf000073_0002
OP-1074
or a pharmaceutically acceptable salt thereof,
in combination with an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic.
5. The method of claim 1 or 4, wherein the disorder is mediated by the estrogen receptor.
6. The method of claim 1 or 4, wherein the disorder is breast cancer.
7. The method of claim 6, wherein the breast cancer is local, advanced or metastatic estrogen or progesterone receptor positive advanced breast cancer.
8. The method of claim 6, wherein the breast cancer is estrogen or progesterone receptor positive advanced breast cancer.
9. The method of claim 6, wherein the breast cancer is estrogen or progesterone receptor negative breast cancer.
10. The method of claim 1 or 4, wherein the disorder is selected from the group consisting of ovarian, endometrial, and vaginal cancer and endometriosis.
11. The method of claim 1 or 4, wherein the disorder is retinoblastoma positive breast cancer.
12. The method of claim 1 or 4, wherein the disorder is selected from the group consisting of retinoblastoma positive endometrial, vaginal and ovarian cancers and lung and bronchial cancers.
13. The method of claim 1 or 4, wherein the disorder is lung cancer or bronchial cancer.
14. The method of claim 1 or 4, wherein the agent is a CDK 4/6 inhibitor.
15. The method of claim 14, wherein the CDK 4/6 inhibitor is PD-0332991, LY2835219, or LEE011.
16. The method of claim 14, wherein the CDK 4/6 inhibitor is PD-0332991, or a
pharmaceutically acceptable salt thereof.
17. The method of claim 14, wherein the CDK 4/6 inhibitor is PD-0332991.
18. The method of claim 1 or 4, wherein the agent is an mTOR inhibitor.
19. The method of claim 18, wherein the mTOR inhibitor is everolimus.
20. The method of claim 18, wherein the mTOR inhibitor is rapamycin, everolimus, temsirolimus, AP23573, AZD8055, WYE-354, WYE-600, WYE-687, or Ppl21.
21. The method of claim 1 or 4, wherein the agent is a PI3 Kinase inhibitor.
22. The method of claim 21, wherein the PI3 Kinase inhibitor is BKM-120, XL-147, RG- 7321, CH-5132799 and BAY-80-6946. .
23. The method of claim 21, wherein the PI3 Kinase inhibitor is BKM-120.
24. The method of claim 1 or 4, wherein the agent is a taxane.
25. The method of claim 24, wherein the taxane is paclitaxel or docetaxel.
26. The method of claim 1 or 4, wherein the agent is an antimetabolite.
27. The method of claim 26, wherein the antimetabolite is 5-fluorouracil.
28. The method of claim 1 or 4, wherein the agent is an antitumor antiobiotic.
29. The method of claim 28, wherein the antitumor antibiotic is doxorubicin.
30. A pharmaceutical composition comprising a pharmaceutically effective amount of a compound, which is
Figure imgf000075_0001
OP-1038
Figure imgf000075_0002
Figure imgf000075_0003
OP-1074 or
Figure imgf000075_0004
wherein Ri and R2 are independently either:
(i) OH or OR9, (ii) wherein R9 is independently selected from H, halogen (CI, Br, I or F), natural or non- naturally occurring amino acid (bound through either the OC(O)- or C(0)0- (an ester) or the amino (through either -C(0)-N- or -N-C(O)- (an amide linkage)), R10, -OR10, or -SR10 where R10 is -C(=0)RC1, -C(=0)0RC1, -C(=0)SRC1, -C(=0)N(RC1)2; or polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;
-S(=0)2RC1, -S(=0)2ORcl, -S(=0)RC1, -S(=0)ORcl, -P(=0)2RC1, -P(=0)2ORcl, -P(=0)(ORcl)2, -P(=0)(RC1)2, or -P(RC1)(0RC1); or oxygen attached to an oxygen protecting group (to produce OH on administration), sulfur attached to a sulfur protecting group (to produce SH or a disulfide on administration), or nitrogen attached to a nitrogen protecting group (to produce - H- on administration);
and R can be independently selected from hydrogen, polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or two R groups are joined to form an substituted or unsubstituted heterocyclic ring;
or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, stereoisomer, tautomer, or N- oxide thereof,
in combination with an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic, and a pharmaceutically acceptable carrier.
31. The pharmaceutical composition of claim 30, wherein the compound is in the form of a pharmaceutically acceptable salt.
32. The pharmaceutical composition of claim 31, wherein the compound is (2S)-3-(4- hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3 -methyl pyrrolidin-1 -yl]ethoxy}phenyl)-2H-chromen- 7-ol, which has the chemical structure:
Figure imgf000077_0001
OP-1074
or a pharmaceutically acceptable salt, solvate, hydrate, or N-oxide thereof.
33. A pharmaceutical composition comprising a pharmaceutically effective amount of a compound, which is (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3-methylpyrrolidin-l- yl]ethoxy}phenyl)-2H-chromen-7-ol, which has the chemical structure:
Figure imgf000077_0002
OP-1074
or a pharmaceutically acceptable salt thereof,
in combination with an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic, and a pharmaceutically acceptable carrier.
Figure imgf000077_0003
OP-1038
Figure imgf000078_0001
Figure imgf000078_0002
Figure imgf000078_0003
wherein Ri and R2 are independently either:
(i) OH or OR9,
(ii) wherein R9 is independently selected from H, halogen (CI, Br, I or F), natural or non- naturally occurring amino acid (bound through either the OC(O)- or C(0)0- (an ester) or the amino (through either -C(0)-N- or -N-C(O)- (an amide linkage)), R10, -OR10, or -SR10 where R10 is -C(=0)RC1, -C(=0)0RC1, -C(=0)SRC1, -C(=0)N(RC1)2; or polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl;
-S(=0)2RC1, -S(=0)2ORcl, -S(=0)RC1, -S(=0)ORcl, -P(=0)2RC1, -P(=0)2ORcl, -P(=0)(ORcl)2, -P(=0)(RC1)2, or -P(RC1)(0RC1); or oxygen attached to an oxygen protecting group (to produce OH on administration), sulfur attached to a sulfur protecting group (to produce SH or a disulfide on administration), or nitrogen attached to a nitrogen protecting group (to produce - H- on administration); and R can be independently selected from hydrogen, polyethylene glycol, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, substituted or unsubstituted carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl, or two R groups are joined to form an substituted or unsubstituted heterocyclic ring;
or a pharmaceutically acceptable salt, solvate, hydrate, prodrug, stereoisomer, tautomer, or N- oxide thereof,
and an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic, for the treatment of a disorder modulated, mediated or affected by the estrogen receptor.
35. The combination of claim 34, wherein the compound is in the form of a pharmaceutically acceptable salt.
36. The combination of claim 34, wherein the compound is (2S)-3-(4-hydroxyphenyl)-4- methyl-2-(4-{2-[(3R)-3-methylpyrrolidin-l-yl]ethoxy}phenyl)-2H-chromen-7-ol, which has the chemical structure:
Figure imgf000079_0001
OP-1074
or a pharmaceutically acceptable salt, solvate, hydrate, or N-oxide thereof.
37. A combination of (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3-methylpyrrolidin- l-yl]ethoxy}phenyl)-2H-chromen-7-ol, which has the chemical structure:
Figure imgf000079_0002
OP-1074 or a pharmaceutically acceptable salt thereof,
and an agent selected from the group consisting of an mTOR inhibitor, a CDK 4/6 inhibitor, a PI3 Kinase inhibitor, a taxane, an antimetabolite, and an antitumor antibiotic for the treatment of a disorder modulated, mediated or affected by the estrogen receptor.
PCT/IB2014/062176 2013-06-19 2014-06-12 Combinations of benzopyran compounds, compositions and uses thereof WO2014203129A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361837130P 2013-06-19 2013-06-19
US61/837,130 2013-06-19

Publications (1)

Publication Number Publication Date
WO2014203129A1 true WO2014203129A1 (en) 2014-12-24

Family

ID=51022942

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2014/062176 WO2014203129A1 (en) 2013-06-19 2014-06-12 Combinations of benzopyran compounds, compositions and uses thereof

Country Status (4)

Country Link
JP (1) JP2015003909A (en)
AR (1) AR096645A1 (en)
TW (1) TW201511755A (en)
WO (1) WO2014203129A1 (en)

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104529904A (en) * 2015-01-09 2015-04-22 苏州明锐医药科技有限公司 Preparing method for bemaciclib
WO2015136016A3 (en) * 2014-03-13 2015-12-10 F. Hoffmann-La Roche Ag Therapeutic combinations with estrogen receptor modulators
WO2016176666A1 (en) * 2015-04-29 2016-11-03 Radius Health, Inc. Methods of treating cancer
WO2017024318A1 (en) 2015-08-06 2017-02-09 Dana-Farber Cancer Institute, Inc. Targeted protein degradation to attenuate adoptive t-cell therapy associated adverse inflammatory responses
WO2017059139A1 (en) 2015-10-01 2017-04-06 Olema Pharmaceuticals, Inc. TETRAHYDRO-1H-PYRIDO[3,4-b]INDOLE ANTI-ESTROGENIC DRUGS
WO2017197036A1 (en) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. Spirocyclic degronimers for target protein degradation
WO2017197046A1 (en) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. C3-carbon linked glutarimide degronimers for target protein degradation
WO2017197055A1 (en) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. Heterocyclic degronimers for target protein degradation
WO2018129419A1 (en) * 2017-01-05 2018-07-12 Radius Pharmaceuticals, Inc. Polymorphic forms of rad1901-2hcl
US10071066B2 (en) 2014-03-28 2018-09-11 Duke University Method of treating cancer using selective estrogen receptor modulators
US10118910B2 (en) 2015-12-09 2018-11-06 The Board Of Trustees Of The University Of Illinois Benzothiophene-based selective estrogen receptor downregulators
WO2019006393A1 (en) 2017-06-29 2019-01-03 G1 Therapeutics, Inc. Morphic forms of git38 and methods of manufacture thereof
US10208011B2 (en) 2017-02-10 2019-02-19 G1 Therapeutics, Inc. Benzothiophene estrogen receptor modulators
WO2019079569A1 (en) 2017-10-18 2019-04-25 Novartis Ag Compositions and methods for selective protein degradation
US10420734B2 (en) 2014-03-28 2019-09-24 Duke University Method of treating cancer using selective estrogen receptor modulators
WO2020132561A1 (en) 2018-12-20 2020-06-25 C4 Therapeutics, Inc. Targeted protein degradation
US10703747B2 (en) 2016-10-24 2020-07-07 The Board of Directors of the University of Illinois Benzothiophene-based selective mixed estrogen receptor downregulators
WO2020219742A1 (en) 2019-04-24 2020-10-29 Novartis Ag Compositions and methods for selective protein degradation
CN111918656A (en) * 2018-03-29 2020-11-10 柏林化学股份有限公司 Anti-cancer pharmaceutical composition for combination therapy
CN113045546A (en) * 2021-03-25 2021-06-29 浙江天宇药业股份有限公司 Azolidinib impurity, preparation method and application thereof
EP3858835A1 (en) 2016-07-01 2021-08-04 G1 Therapeutics, Inc. Pyrimidine-based antiproliferative agents
US11311609B2 (en) 2017-02-08 2022-04-26 Dana-Farber Cancer Institute, Inc. Regulating chimeric antigen receptors
US11364222B2 (en) 2017-01-06 2022-06-21 G1 Therapeutics, Inc. Combination therapy for treatment of cancer
WO2022199656A1 (en) * 2021-03-24 2022-09-29 贝达药业股份有限公司 Pharmaceutical combination, kit containing same, and use thereof
US11643385B2 (en) 2018-07-04 2023-05-09 Radius Pharmaceuticals, Inc. Polymorphic forms of RAD1901-2HCl
WO2024030968A1 (en) 2022-08-03 2024-02-08 Brystol-Myers Squibb Company Compounds for modulating ret protein

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AR104068A1 (en) * 2015-03-26 2017-06-21 Hoffmann La Roche COMBINATIONS OF A 3-KINASE PHOSFOINOSYTIDE INHIBITOR COMPOSITE AND A CDK4 / 6 INHIBITOR COMPOUND FOR CANCER TREATMENT
WO2019097426A1 (en) * 2017-11-16 2019-05-23 Novartis Ag Pharmaceutical combination comprising lsz102 and ribociclib

Citations (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3668222A (en) 1969-05-14 1972-06-06 Sandoz Ltd 11-desacetoxy-wortmannin
US4418068A (en) 1981-04-03 1983-11-29 Eli Lilly And Company Antiestrogenic and antiandrugenic benzothiophenes
US4659516A (en) 1983-10-12 1987-04-21 Imperial Chemical Industries Plc Steroid derivatives
US5254568A (en) 1990-08-09 1993-10-19 Council Of Scientific & Industrial Research Benzopyrans as antiestrogenic agents
US5378725A (en) 1993-07-19 1995-01-03 The Arizona Board Of Regents Inhibition of phosphatidylinositol 3-kinase with wortmannin and analogs thereof
US5393763A (en) 1992-07-28 1995-02-28 Eli Lilly And Company Methods for inhibiting bone loss
US5395842A (en) 1988-10-31 1995-03-07 Endorecherche Inc. Anti-estrogenic compounds and compositions
US5441947A (en) 1993-08-25 1995-08-15 Eli Lilly And Company Methods of inhibiting vascular restenosis
US5468773A (en) 1993-08-25 1995-11-21 Eli Lilly And Company Methods for inhibiting bone loss and cartilage degradation using wortmannin and its analogs
US5478847A (en) 1994-03-02 1995-12-26 Eli Lilly And Company Methods of use for inhibiting bone loss and lowering serum cholesterol
US5480906A (en) 1994-07-01 1996-01-02 Eli Lilly And Company Stereochemical Wortmannin derivatives
US5504103A (en) 1993-08-25 1996-04-02 Eli Lilly And Company Inhibition of phosphatidylinositol 3-kinase with 17 β-hydroxywortmannin and analogs thereof
US5726167A (en) 1993-10-12 1998-03-10 Eli Lilly And Company Inhibition of phosphatidylinositol 3-kinase with viridin, demethoxyviridin, viridiol, demethoxyviridiol, virone, wortmannolone, and analogs thereof
US6060503A (en) 1991-12-02 2000-05-09 Endorecherche, Inc. Benzopyran-containing compounds and method for their use
US6262270B1 (en) 1998-08-14 2001-07-17 Schering Corporation Enantioselective synthesis
WO2001054699A1 (en) 2000-01-28 2001-08-02 Endorecherche, Inc. Selective estrogen receptor modulators in combination with estrogens
US20020037276A1 (en) 1998-06-01 2002-03-28 Andrzej Ptasznik Phosphatidylinositol 3-kinase inhibitors as stimulators of endocrine differentiation
US6403588B1 (en) 2000-04-27 2002-06-11 Yamanouchi Pharmaceutical Co., Ltd. Imidazopyridine derivatives
US6465445B1 (en) 1998-06-11 2002-10-15 Endorecherche, Inc. Medical uses of a selective estrogen receptor modulator in combination with sex steroid precursors
US20020161014A1 (en) 2000-04-25 2002-10-31 Chanchal Sadhu Inhibitors of human phosphatidylinositol 3-kinase delta
WO2003062236A1 (en) 2002-01-22 2003-07-31 Warner-Lambert Company Llc 2-(PYRIDIN-2-YLAMINO)-PYRIDO[2,3d]PYRIMIDIN-7-ONES
US6608056B1 (en) 2000-04-27 2003-08-19 Yamanouchi Pharmaceutical Co., Ltd Fused heteroaryl derivatives
US6774122B2 (en) 2000-01-10 2004-08-10 Astrazeneca Ab Formulation
US20040192770A1 (en) 1998-06-26 2004-09-30 Kozikowski Alan P. Inhibitors of phosphatidyl myo-inositol cycle
WO2004091488A2 (en) 2003-04-14 2004-10-28 Merck & Co., Inc. Estrogen receptor modulators
WO2004108713A1 (en) 2003-06-05 2004-12-16 Warner-Lambert Company Llc Cycloalkyl and heterocycloalkyl substituted benzothiophenes as therapeutic agents
WO2004108715A1 (en) 2003-06-05 2004-12-16 Warner-Lambert Company Llc 3-aryloxy and 3-heteroaryloxy substituted benzo(b) thiophenes as therapeutic agents with pi3k activity
WO2005005426A1 (en) 2003-07-11 2005-01-20 Warner-Lambert Company Llc Isethionate salt of a selective cdk4 inhibitor
US7005428B1 (en) 1998-06-11 2006-02-28 Endorecherche, Inc. Medical uses of a selective estrogen receptor modulator in combination with sex steroid precursors
WO2007127183A1 (en) 2006-04-26 2007-11-08 Genentech, Inc. Phosphoinositide 3-kinase inhibitor compounds and pharmaceutical compositions containing them
WO2008032157A2 (en) 2006-09-08 2008-03-20 Pfizer Products Inc. Synthesis of 2-(pyridin-2-ylamino)-pyrido[2,3-d]pyrimidin-7-ones
WO2008070740A1 (en) 2006-12-07 2008-06-12 F.Hoffmann-La Roche Ag Phosphoinositide 3-kinase inhibitor compounds and methods of use
WO2008073785A2 (en) 2006-12-07 2008-06-19 Genentech, Inc. Phosphoinositide 3-kinase inhibitor compounds and methods of use
WO2010145010A1 (en) 2009-06-16 2010-12-23 Endorecherche, Inc. Treatment of hot flushes, vasomotor symptoms, and night sweats with sex steroid precursors in combination with selective estrogen receptor modulators
WO2011156518A2 (en) 2010-06-10 2011-12-15 Aragon Pharmaceuticals, Inc. Estrogen receptor modulators and uses thereof
WO2013090921A1 (en) 2011-12-16 2013-06-20 Olema Pharmaceuticals, Inc. Novel benzopyran compounds, compositions and uses thereof

Patent Citations (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3668222A (en) 1969-05-14 1972-06-06 Sandoz Ltd 11-desacetoxy-wortmannin
US4418068A (en) 1981-04-03 1983-11-29 Eli Lilly And Company Antiestrogenic and antiandrugenic benzothiophenes
US4659516A (en) 1983-10-12 1987-04-21 Imperial Chemical Industries Plc Steroid derivatives
US5840735A (en) 1988-10-31 1998-11-24 Endorecherche Inc. Sex steroid activity inhibitors
US5395842A (en) 1988-10-31 1995-03-07 Endorecherche Inc. Anti-estrogenic compounds and compositions
US5254568A (en) 1990-08-09 1993-10-19 Council Of Scientific & Industrial Research Benzopyrans as antiestrogenic agents
US6060503A (en) 1991-12-02 2000-05-09 Endorecherche, Inc. Benzopyran-containing compounds and method for their use
US5393763A (en) 1992-07-28 1995-02-28 Eli Lilly And Company Methods for inhibiting bone loss
US5457117A (en) 1992-07-28 1995-10-10 Eli Lilly And Company Method for inhibiting bone loss using 6-hydroxy-2-(4-hydroxyphenyl)-benzo[B][2-(piperidin-1-yl)ethoxyphenylimethanone hydrochloride
US5378725A (en) 1993-07-19 1995-01-03 The Arizona Board Of Regents Inhibition of phosphatidylinositol 3-kinase with wortmannin and analogs thereof
US5504103A (en) 1993-08-25 1996-04-02 Eli Lilly And Company Inhibition of phosphatidylinositol 3-kinase with 17 β-hydroxywortmannin and analogs thereof
US5441947A (en) 1993-08-25 1995-08-15 Eli Lilly And Company Methods of inhibiting vascular restenosis
US5468773A (en) 1993-08-25 1995-11-21 Eli Lilly And Company Methods for inhibiting bone loss and cartilage degradation using wortmannin and its analogs
US5726167A (en) 1993-10-12 1998-03-10 Eli Lilly And Company Inhibition of phosphatidylinositol 3-kinase with viridin, demethoxyviridin, viridiol, demethoxyviridiol, virone, wortmannolone, and analogs thereof
US5478847A (en) 1994-03-02 1995-12-26 Eli Lilly And Company Methods of use for inhibiting bone loss and lowering serum cholesterol
US5480906A (en) 1994-07-01 1996-01-02 Eli Lilly And Company Stereochemical Wortmannin derivatives
US20020037276A1 (en) 1998-06-01 2002-03-28 Andrzej Ptasznik Phosphatidylinositol 3-kinase inhibitors as stimulators of endocrine differentiation
US6465445B1 (en) 1998-06-11 2002-10-15 Endorecherche, Inc. Medical uses of a selective estrogen receptor modulator in combination with sex steroid precursors
US7005428B1 (en) 1998-06-11 2006-02-28 Endorecherche, Inc. Medical uses of a selective estrogen receptor modulator in combination with sex steroid precursors
US20040192770A1 (en) 1998-06-26 2004-09-30 Kozikowski Alan P. Inhibitors of phosphatidyl myo-inositol cycle
US6262270B1 (en) 1998-08-14 2001-07-17 Schering Corporation Enantioselective synthesis
US7456160B2 (en) 2000-01-10 2008-11-25 Astrazeneca Ab Formulation
US6774122B2 (en) 2000-01-10 2004-08-10 Astrazeneca Ab Formulation
WO2001054699A1 (en) 2000-01-28 2001-08-02 Endorecherche, Inc. Selective estrogen receptor modulators in combination with estrogens
US20020161014A1 (en) 2000-04-25 2002-10-31 Chanchal Sadhu Inhibitors of human phosphatidylinositol 3-kinase delta
US6653320B2 (en) 2000-04-27 2003-11-25 Yamanouchi Pharmaceutical Co. Ltd. Imidazopyridine derivatives
US6403588B1 (en) 2000-04-27 2002-06-11 Yamanouchi Pharmaceutical Co., Ltd. Imidazopyridine derivatives
US6608056B1 (en) 2000-04-27 2003-08-19 Yamanouchi Pharmaceutical Co., Ltd Fused heteroaryl derivatives
US7456168B2 (en) 2002-01-22 2008-11-25 Warner-Lambert Company 2-(pyridin-2-ylamino)-pyrido[2,3, d]pyrimidin-7-ones
US6936612B2 (en) 2002-01-22 2005-08-30 Warner-Lambert Company 2-(Pyridin-2-ylamino)-pyrido[2,3-d]pyrimidin-7-ones
WO2003062236A1 (en) 2002-01-22 2003-07-31 Warner-Lambert Company Llc 2-(PYRIDIN-2-YLAMINO)-PYRIDO[2,3d]PYRIMIDIN-7-ONES
US7208489B2 (en) 2002-01-22 2007-04-24 Warner-Lambert Company 2-(pyridin-2-ylamino)-pyrido [2,3-d]pyrimidin-7-ones
WO2004091488A2 (en) 2003-04-14 2004-10-28 Merck & Co., Inc. Estrogen receptor modulators
WO2004108713A1 (en) 2003-06-05 2004-12-16 Warner-Lambert Company Llc Cycloalkyl and heterocycloalkyl substituted benzothiophenes as therapeutic agents
WO2004108715A1 (en) 2003-06-05 2004-12-16 Warner-Lambert Company Llc 3-aryloxy and 3-heteroaryloxy substituted benzo(b) thiophenes as therapeutic agents with pi3k activity
WO2005005426A1 (en) 2003-07-11 2005-01-20 Warner-Lambert Company Llc Isethionate salt of a selective cdk4 inhibitor
US7345171B2 (en) 2003-07-11 2008-03-18 Warner-Lambert Company Llc Isethionate salt of a selective CKD4 inhibitor
US7863278B2 (en) 2003-07-11 2011-01-04 Warner-Lambert CompanyLLC Isethionate salt of a selective CDK4 inhibitor
WO2007127183A1 (en) 2006-04-26 2007-11-08 Genentech, Inc. Phosphoinositide 3-kinase inhibitor compounds and pharmaceutical compositions containing them
US7781583B2 (en) 2006-09-08 2010-08-24 Pfizer Inc Synthesis of 2-(pyridin-2-ylamino)-pyrido[2,3-d] pryimidin-7-ones
WO2008032157A2 (en) 2006-09-08 2008-03-20 Pfizer Products Inc. Synthesis of 2-(pyridin-2-ylamino)-pyrido[2,3-d]pyrimidin-7-ones
US20080242665A1 (en) 2006-12-07 2008-10-02 Tracy Bayliss Phosphoinositide 3-kinase inhibitor compounds and methods of use
WO2008073785A2 (en) 2006-12-07 2008-06-19 Genentech, Inc. Phosphoinositide 3-kinase inhibitor compounds and methods of use
WO2008070740A1 (en) 2006-12-07 2008-06-12 F.Hoffmann-La Roche Ag Phosphoinositide 3-kinase inhibitor compounds and methods of use
WO2010145010A1 (en) 2009-06-16 2010-12-23 Endorecherche, Inc. Treatment of hot flushes, vasomotor symptoms, and night sweats with sex steroid precursors in combination with selective estrogen receptor modulators
WO2011156518A2 (en) 2010-06-10 2011-12-15 Aragon Pharmaceuticals, Inc. Estrogen receptor modulators and uses thereof
US20130116232A1 (en) * 2010-06-10 2013-05-09 Aragon Pharmaceuticals, Inc. Estrogen receptor modulators and uses thereof
WO2013090921A1 (en) 2011-12-16 2013-06-20 Olema Pharmaceuticals, Inc. Novel benzopyran compounds, compositions and uses thereof
US20130178445A1 (en) 2011-12-16 2013-07-11 Olema Pharmaceuticals, Inc. Novel Benzopyran Compounds, Compositions and Uses Thereof

Non-Patent Citations (48)

* Cited by examiner, † Cited by third party
Title
"CRC Handbook of Optical Resolution via Diastereomeric Salt Formation"
"Remington's Pharmaceutical Sciences, 17th edition,", 1985, MACK PUBLISHING COMPANY
BAER ET AL., CALCIFIED TISSUE INT., vol. 55, 1994, pages 338
BERGE ET AL., J. PHARM. SCI., vol. 66, no. 1, January 1977 (1977-01-01), pages 1 - 79
BIGHLEY ET AL.: "Encyclopedia of Pharmaceutical Technology", vol. 13, 1995, MARCEL DEKKER, article "Salt Forms of Drugs and Adsorption"
BISKOBING, D. M., CLINICAL INTERVENTIONS IN AGING, vol. 2, no. 3, 2007, pages 299 - 303
BLIZZARD ET AL., BIOORG MED CHEM LETT., vol. 15, no. 1, 2005, pages 107 - 13
BLIZZARD ET AL., BIOORG MED CHEM LETT., vol. 15, no. 17, 2005, pages 3912 - 6
BLIZZARD ET AL., BIOORG MED CHEM LETT., vol. 15, no. 23, 2005, pages 5214 - 8
BLIZZARD ET AL., BIOORG MED CHEM LETT., vol. 15, no. 6, 2005, pages 1675 - 81
CURR TOP MED CHEM., vol. 8, no. 9, 2008, pages 792 - 812
DALE; MOSHER, JAM. CHEM. SOC., vol. 95, 1973, pages 512
DODGE ET AL., J. BONE MINER. RES., vol. 8, no. 1, 1993, pages S278
E.L. ELIEL: "Stereochemistry of Carbon Compounds", 1962, MCGRAW-HILL
ELIEL: "Stereochemistry of Carbon Compounds", 1962, MCGRAW-HILL
ERNEST L. ELIEL; SAMUEL H. WILEN; LEWIS N. MANDA: "Stereochemistry of Organic Compounds", 1994, JOHN WILEY & SONS, INC.
FOLKES ET AL., JOURNAL OF MEDICINAL CHEMISTRY, vol. 51, 2008, pages 5522 - 5532
GAUTHIER ET AL., J ENZYME INHIB MED CHEM, vol. 20, no. 2, 2005, pages 165 - 77
GENG ET AL., CANCER RES., vol. 64, 2004, pages 4893 - 99
GENNARI ET AL., EXPERT OPIN INVESTIG DRUGS, vol. 15, no. 9, 2006, pages 1091 - 103
HAYAKAWA ET AL., BIOORG MED CHEM, vol. 14, no. 20, 2006, pages 6847 - 6858
HAYAKAWA ET AL., BIOORGANIC & MEDICINAL CHEMISTRY, vol. 14, 2006, pages 6847 - 6858
HORTOBAGYI, DRUGS, vol. 54, no. 4, 1997, pages 1 - 7
IHLE ET AL., MOL CANCER THER., vol. 3, no. 7, 2004, pages 763 - 72
J. JACQUES; A. COLLET; S.H. WILEN: "Enantiomers, Racemates and Resolutions", 1981, WILEY-INTERSCIENCE
JACQUES ET AL.: "Enantiomers, Racemates and Resolutions", 1981, WILEY INTERSCIENCE
JORDAN, J., CELL. BIOCHEM., 1995, pages 51
KATZENELLENBOGEN, J MED CHEM, vol. 54, no. 15, 2011, pages 5271 - 82
LABRIE ET AL.: "The combination of a novel selective estrogen receptor modulator with an estrogen protects the mammary gland and uterus in a rodent model: the future of postmenopausal women's health?", ENDOCRINOLOGY, vol. 144, no. 11, 2003, pages 4700 - 6
LINDMO, JOURNAL OF CELL SCIENCE, vol. 119, 2006, pages 605 - 614
MEUILLET ET AL., ONCOL. RES., vol. 14, 2004, pages 513 - 27
MIHALY NOGRADI: "Stereoselective Synthesis A Practical Approach", 1995, VCH PUBLISHERS, INC.
MINOTTI ET AL., PHARMACOLOGIY. REV., vol. 56, 2004, pages 185 - 229
ROBERTSON ET AL., J CLIN ONCOL, vol. 27, no. 27, 2009, pages 4530 - 5
S.H. WILEN: "Tables o/ResolvingAgents and Optical Resolutions", vol. 268, 1972, UNIV. OF NOTRE DAME PRESS
S.H. WILEN; A. COLLET; J. JACQUES: "Tetrahedron", 1977, pages: 2725
SHARMA ET AL., J MED CHEM, vol. 33, no. 12, 1990, pages 3216 - 22
SHARMA ET AL., J MED CHEM, vol. 33, no. 12, 1990, pages 3222 - 9
STEPHENS, CURRENT OPINION IN PHARMACOLOGY, vol. 5, no. 4, 2005, pages 357 - 365
T. W. GREENE; P. G. M. WUTS: "Protecting Groups in Organic Synthesis, Second Edition,", 1991, WILEY
TABELLINI ET AL., BR. J. HAEMATOL., vol. 126, no. 4, 2004, pages 574 - 82
TEN HOEVE; H. WYNBERG, JOURNAL OF ORGANIC CHEMISTRY, vol. 50, 1985, pages 4508 - 4514
VLAHOS ET AL., J. BIOL., CHEM., vol. 269, no. 7, 1994, pages 5241 - 5248
WAKELING, BREAST CANCER RES. TREAT., vol. 25, 1993, pages 1
WILEN ET AL., TETRAHEDRON, vol. 33, 1977, pages 2725
WILEN: "Tables of Resolving Agents and Optical Resolutions", vol. 268, 1972, UNIV. OF NOTRE DAME PRESS
WU ET AL., MOL CELL, vol. 18, 2005, pages 413
ZHU ET AL., CANCER RES., vol. 64, no. 12, 2004, pages 4309 - 18

Cited By (64)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015136016A3 (en) * 2014-03-13 2015-12-10 F. Hoffmann-La Roche Ag Therapeutic combinations with estrogen receptor modulators
US11951080B2 (en) 2014-03-28 2024-04-09 Duke University Method of treating cancer using selective estrogen receptor modulators
US10420734B2 (en) 2014-03-28 2019-09-24 Duke University Method of treating cancer using selective estrogen receptor modulators
US10071066B2 (en) 2014-03-28 2018-09-11 Duke University Method of treating cancer using selective estrogen receptor modulators
US11779552B2 (en) 2014-03-28 2023-10-10 Duke University Method of treating cancer using selective estrogen receptor modulators
CN104529904A (en) * 2015-01-09 2015-04-22 苏州明锐医药科技有限公司 Preparing method for bemaciclib
CN108024541B (en) * 2015-04-29 2021-07-20 雷迪厄斯制药公司 Methods for treating cancer
CN113288887A (en) * 2015-04-29 2021-08-24 雷迪厄斯制药公司 Methods for treating cancer
WO2016176666A1 (en) * 2015-04-29 2016-11-03 Radius Health, Inc. Methods of treating cancer
US11413258B2 (en) 2015-04-29 2022-08-16 Radius Pharmaceuticals, Inc. Methods for treating cancer
CN108024541A (en) * 2015-04-29 2018-05-11 雷迪厄斯制药公司 Method for treating cancer
CN108135177A (en) * 2015-04-29 2018-06-08 雷迪厄斯制药公司 For the method for the treatment of cancer
RU2737496C2 (en) * 2015-04-29 2020-12-01 Радиус Фармасьютикалз, Инк. Methods of treating cancer
WO2016176664A1 (en) * 2015-04-29 2016-11-03 Radius Health, Inc. Methods for treating cancer
WO2016176665A1 (en) * 2015-04-29 2016-11-03 Radius Health, Inc. Methods of treating cancer
US11819480B2 (en) 2015-04-29 2023-11-21 Radius Pharmaceuticals, Inc. Methods for treating cancer
EP3288383A4 (en) * 2015-04-29 2019-01-23 Radius Pharmaceuticals, Inc. Methods of treating cancer
EP3288382A4 (en) * 2015-04-29 2019-01-30 Radius Pharmaceuticals, Inc. Methods for treating cancer
AU2016256471B2 (en) * 2015-04-29 2020-09-10 Radius Pharmaceuticals, Inc. Methods of treating cancer
AU2016256469B2 (en) * 2015-04-29 2020-12-10 Radius Pharmaceuticals, Inc. Methods for treating cancer
CN108135177B (en) * 2015-04-29 2021-06-01 雷迪厄斯制药公司 Methods for treating cancer
RU2747228C2 (en) * 2015-04-29 2021-04-29 Радиус Фармасьютикалз, Инк. Methods for treating cancer
RU2745678C2 (en) * 2015-04-29 2021-03-30 Радиус Фармасьютикалз, Инк. Methods for treating cancer
US11046954B2 (en) 2015-08-06 2021-06-29 Dana-Farber Cancer Institute, Inc. Targeted protein degradation to attenuate adoptive T-cell therapy associated adverse inflammatory responses
US11293023B2 (en) 2015-08-06 2022-04-05 Dana-Farber Cancer Institute, Inc. Tunable endogenous protein degradation
WO2017024318A1 (en) 2015-08-06 2017-02-09 Dana-Farber Cancer Institute, Inc. Targeted protein degradation to attenuate adoptive t-cell therapy associated adverse inflammatory responses
EP3912680A1 (en) 2015-10-01 2021-11-24 Olema Pharmaceuticals, Inc. Tetrahydro-1h-pyrido[3,4-b]indole anti-estrogenic drugs
WO2017059139A1 (en) 2015-10-01 2017-04-06 Olema Pharmaceuticals, Inc. TETRAHYDRO-1H-PYRIDO[3,4-b]INDOLE ANTI-ESTROGENIC DRUGS
US10624878B2 (en) 2015-10-01 2020-04-21 Olema Pharmaceuticals, Inc. Tetrahydro-1H-pyrido [3,4-b]indole anti-estrogenic drugs
US11229630B2 (en) 2015-10-01 2022-01-25 Olema Pharmaceuticals, Inc. Tetrahydro-1H-pyrido [3,4-b]indole anti-estrogenic drugs
US11672785B2 (en) 2015-10-01 2023-06-13 Olema Pharmaceuticals, Inc. Tetrahydro-1H-pyrido [3,4-b]indole anti-estrogenic drugs
US10292971B2 (en) 2015-10-01 2019-05-21 Olema Pharmaceuticals, Inc. Tetrahydro-1H-pyrido[3,4-b]indole anti-estrogenic drugs
US10377735B2 (en) 2015-12-09 2019-08-13 The Board Of Trustees Of The University Of Illinois Benzothiophene-based selective estrogen receptor downregulators
US11447461B2 (en) 2015-12-09 2022-09-20 The Board Of Trustees Of The University Of Illinois Benzothiophene-based selective estrogen receptor downregulators
US10807964B2 (en) 2015-12-09 2020-10-20 The Board Of Trustees Of The University Of Illinois Benzothiophene-based selective estrogen receptor downregulators
US10118910B2 (en) 2015-12-09 2018-11-06 The Board Of Trustees Of The University Of Illinois Benzothiophene-based selective estrogen receptor downregulators
US11072595B2 (en) 2015-12-09 2021-07-27 The Board of Trustees of lhe University of Illinois Benzothiophene-based selective estrogen receptor downregulator compounds
WO2017197046A1 (en) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. C3-carbon linked glutarimide degronimers for target protein degradation
WO2017197055A1 (en) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. Heterocyclic degronimers for target protein degradation
WO2017197036A1 (en) 2016-05-10 2017-11-16 C4 Therapeutics, Inc. Spirocyclic degronimers for target protein degradation
EP3858835A1 (en) 2016-07-01 2021-08-04 G1 Therapeutics, Inc. Pyrimidine-based antiproliferative agents
US10703747B2 (en) 2016-10-24 2020-07-07 The Board of Directors of the University of Illinois Benzothiophene-based selective mixed estrogen receptor downregulators
WO2018129419A1 (en) * 2017-01-05 2018-07-12 Radius Pharmaceuticals, Inc. Polymorphic forms of rad1901-2hcl
US10385008B2 (en) 2017-01-05 2019-08-20 Radius Pharmaceuticals, Inc. Polymorphic forms of RAD1901-2HCL
US11708318B2 (en) 2017-01-05 2023-07-25 Radius Pharmaceuticals, Inc. Polymorphic forms of RAD1901-2HCL
CN110191707A (en) * 2017-01-05 2019-08-30 雷迪厄斯制药公司 The polymorphic forms of RAD1901-2HCL
KR20190105030A (en) * 2017-01-05 2019-09-11 래디어스 파마슈티컬스, 인코포레이티드 Polymorphic form of RAD1901-2HCL
KR102322802B1 (en) 2017-01-05 2021-11-04 래디어스 파마슈티컬스, 인코포레이티드 Polymorphic form of RAD1901-2HCL
US11364222B2 (en) 2017-01-06 2022-06-21 G1 Therapeutics, Inc. Combination therapy for treatment of cancer
US11311609B2 (en) 2017-02-08 2022-04-26 Dana-Farber Cancer Institute, Inc. Regulating chimeric antigen receptors
EP4119552A1 (en) 2017-02-08 2023-01-18 Dana-Farber Cancer Institute, Inc. Regulating chimeric antigen receptors
US10981887B2 (en) 2017-02-10 2021-04-20 G1 Therapeutics, Inc. Benzothiophene estrogen receptor modulators
US10633362B2 (en) 2017-02-10 2020-04-28 G1 Therapeutics, Inc. Benzothiophene estrogen receptor modulators
US10208011B2 (en) 2017-02-10 2019-02-19 G1 Therapeutics, Inc. Benzothiophene estrogen receptor modulators
WO2019006393A1 (en) 2017-06-29 2019-01-03 G1 Therapeutics, Inc. Morphic forms of git38 and methods of manufacture thereof
WO2019079569A1 (en) 2017-10-18 2019-04-25 Novartis Ag Compositions and methods for selective protein degradation
CN111918656B (en) * 2018-03-29 2023-08-08 柏林化学股份有限公司 Anticancer pharmaceutical composition for combination therapy
CN111918656A (en) * 2018-03-29 2020-11-10 柏林化学股份有限公司 Anti-cancer pharmaceutical composition for combination therapy
US11643385B2 (en) 2018-07-04 2023-05-09 Radius Pharmaceuticals, Inc. Polymorphic forms of RAD1901-2HCl
WO2020132561A1 (en) 2018-12-20 2020-06-25 C4 Therapeutics, Inc. Targeted protein degradation
WO2020219742A1 (en) 2019-04-24 2020-10-29 Novartis Ag Compositions and methods for selective protein degradation
WO2022199656A1 (en) * 2021-03-24 2022-09-29 贝达药业股份有限公司 Pharmaceutical combination, kit containing same, and use thereof
CN113045546A (en) * 2021-03-25 2021-06-29 浙江天宇药业股份有限公司 Azolidinib impurity, preparation method and application thereof
WO2024030968A1 (en) 2022-08-03 2024-02-08 Brystol-Myers Squibb Company Compounds for modulating ret protein

Also Published As

Publication number Publication date
AR096645A1 (en) 2016-01-20
TW201511755A (en) 2015-04-01
JP2015003909A (en) 2015-01-08

Similar Documents

Publication Publication Date Title
WO2014203129A1 (en) Combinations of benzopyran compounds, compositions and uses thereof
JP6880101B2 (en) Benzoxazepine oxazolidinone compound and usage
JP5546636B2 (en) Benzoxepin PI3K inhibitor compounds and methods of use
EP2694072B1 (en) Combination of akt inhibitor compound and abiraterone for use in therapeutic treatments
EP3534903B1 (en) Substituted pyridinone-containing tricyclic compounds, and methods using same
TWI784255B (en) TETRAHYDRO-PYRIDO[3,4-b]INDOLE ESTROGEN RECEPTOR MODULATORS AND USES THEREOF
EP3472162B1 (en) Heteroaryl estrogen receptor modulators and uses thereof
CN107995911B (en) Benzoxazepine compounds and methods of use thereof
JP5511786B2 (en) Benzopyran and benzoxepin PI3K inhibitor compounds and methods of use
JP6858984B2 (en) 6-Heterocyclyl-4-morpholin-4-ylpyridine-2-one compound useful for the treatment of cancer and diabetes
KR20140103335A (en) Novel benzopyran compounds, compositions and uses thereof
CN111527099A (en) Selective inhibitors of protein arginine methyltransferase 5(PRMT5)
WO2017216280A1 (en) TETRAHYDRO-PYRIDO[3,4-b]INDOLE ESTROGEN RECEPTOR MODULATORS AND USES THEREOF
AU2016379290B9 (en) Antitumor effect potentiator comprising pyrrolopyrimidine compound
CA2506308A1 (en) Combination administration of an indolinone with a chemotherapeutic agent for cell proliferation disorders
WO2012118978A1 (en) Methods for treating oncovirus positive cancers
JP7348665B2 (en) Substituted benzothiophene analogs as selective estrogen receptor degraders
JP2014055156A (en) Compound and method for inhibiting interaction of bcl protein with binding partner
KR20210075992A (en) STING working compound
WO2019020559A1 (en) Combination comprising palbociclib and 6-(2,4-dichlorophenyl)-5-[4-[(3s)-1-(3-fluoropropyl)pyrrolidin-3-yl]oxyphenyl]-8,9-dihydro-7h-benzo[7] annulene-2-carboxylic acid and its use for the treatment of cancer
US10654867B2 (en) Heteroaryl estrogen receptor modulators and uses thereof
CN110357858B (en) 5-substituted difluoropiperidine compounds having blood-brain barrier crossing capability

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14733728

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14733728

Country of ref document: EP

Kind code of ref document: A1