OA16917A - Novel benzopyran compounds, compositions and uses thereof. - Google Patents

Novel benzopyran compounds, compositions and uses thereof. Download PDF

Info

Publication number
OA16917A
OA16917A OA1201400252 OA16917A OA 16917 A OA16917 A OA 16917A OA 1201400252 OA1201400252 OA 1201400252 OA 16917 A OA16917 A OA 16917A
Authority
OA
OAPI
Prior art keywords
compound
substituted
unsubstituted
alkyl
administration
Prior art date
Application number
OA1201400252
Inventor
Peter J. Kushner
David C. Myles
Cyrus L. HARMON
Leslie Carol Hodges GALLAGHER
Original Assignee
Olema Pharmaceuticals
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 filed Critical Olema Pharmaceuticals
Publication of OA16917A publication Critical patent/OA16917A/en

Links

Abstract

Benzopyran compounds with strong antiestrogenic activity and essentially no estrogenic activity are provided, which are OP- 1038, which is 3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3methylpyrrolidin-1-y1]ethoxy}phenyl)-2H-chromen7-ol, and OP-1074, which is (2S)-3-(4hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3methylpyrrolidin-1-yl]ethoxy}phenyl)-2H-chromen7-ol. OP-1074 is a pure anti-estrogen when tested in the agonist mode and a complete anti-estrogen when tested in the antagonist mode. These compounds are useful for the treatment or prevention of a variety of conditions that are modulated through the estrogen receptor in mammals including humans.

Description

NOVEL BENZOPYRAN COMPOUNDS, COMPOSITIONS AND USES THEREOF
CROSS REFERENCE TO RELATED APPLICATIONS
This application daims priority to U.S. Provisional Application No.
61/576,890, filed on December 16,2011, the contents of which are incorporated herein by reference.
FIELD [0001] This invention is in the field of pharmaceuticals, and ls in particular novel benzopyran compounds, and salts, prodrugs and dérivatives thereof and their medical uses, including as estrogen receptor modulators and for medical conditions that would bondit from an anti-estrogenic drug, and pharmaceutical compositions thereof.
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 20 antagonists, or mixed agonist-antagonists (sometimes referred to as Sélective Estrogen
Receptor Modulators (SERMs)). For example, estradiol (A) is a pure agonist, fulvestrant (B) îs a complété 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 îs 25 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 stimulâtes tumor growth. In contrast, fulvestrant, a 30 complété anti-estrogen, is free of estrogen-like action on the utérus 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).
-2[0004] Estradiol is n naturally-occuring female estrogenic hormone. Raloxifene was disclosed by Eli Lilly in 198 i (U.S. Patent No. 4,418,068; 5,478,847; 5,393,763; and
5,457,117) for prévention of breast cancer and treatment of osteoporosis. Fulvestrant was disclosed by Impérial 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)).
[0005] The degree of anti-estrogenicity is often assayed by exposing female, immature (preferably ovariéetomized) rodents to test doses of the compound both in the absence (agonist mode) and presence (antagonist mode) of estrogen. Tamoxifen and other partial ami-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 complété 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 complété anti-estrogenicity and correlates well with the rodent weight gain assay.
[0006] Tamoxifen and fulvestrant both inhibit cultured human breast cancer cell prolifération provoked by estrogen. However, fulvestrant more fuily inhibits the prolifération when provoked with growth factors, especially of the insulin/insulin-like growth factor family. Thus the inhibition of growth-factor driven breast cancer cell prolifération and the effect on uterine weight provide two assays which can distinguish between complété 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 dégradation 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, dégradé the receptor but are partial estrogens- that is, not complété antiestrogens. Thus the ability to dégradé the estrogen receptor does not ensure complété antiestrogenicity. The ability to induce dégradation of the receptor is nonetheless a factor that differentiates the behavior of tamoxifen and fulvestrant and may be désirable in u drug to treat breast cancer.
[0008] Fulvestrant, which dégradés the estrogen receptor, incorporâtes a core of 17beta estradiol. It has a long flexible aliphatîc side chain that blocks oral absorption. The 25 estradiol core blocks oral absorption and the long flexible aliphatîc side chain makes the drug very insoluble which worsens the problem. Fulvestrant must be injected because of the poor oral bioavaïlability. 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 30 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:
[0010] Sharma et al. sliowed that the combination of 7-hydroxyl and 4’-hydroxy! groups conferred high-afTinity binding of the benzopyran core to the estrogen receptor (Compound G; sec Compound 25 of Sharma et al. (1990) J Med Chem, 33(12):3222-9 where R1 and R2 are OH).
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 îssued 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.
-5H [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 prodnigs 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 ls (S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-(2-(piperidin-lyl )ethoxy)phenyl )-2H-chromen-7-ol.
EM-652, acolbifene
I [0013] Labrie et al. in WO 01/54699 (sec Figure 4a and 4b) also presented several broad gencric Markush formulae of benzopyran-containing compounds, including acolbifene analogs, in which the side chain terminâtes in various substituted ring Systems including pyrrolidinyl, piperidinyl, and methyl- 1-pyrrolidinyl and dimcthyl-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:
wherein D is -OCHîCHjNÎRjjRj (Rj and Rj either being independently selected from the group consisting of Cj-Cj alkyl, or Rj, Rj and the nitrogen atom to which they are bound together being a ring structure selected from the group
consisting of pyrrolidino, dimethyl-l-pyrrolidino, methyl-l-purroüdinyl, piperidino, hexamethyleneimino and morpholino); and wherein Ri and Ri are independently selected from the group consisting of hydrogen, hydroxyl and a moiety converted in vivo in to hydroxyl, and
wherein Rj and Ri are independently selected from the group consisting of hydroxyl and a moiety converted in vivo in to hydroxyl:
wherein Ri 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, nilrogen-containing cyclic moiety, nitrogen-containing polycyclic moiety, and NRaRb (Ra and Rb being independently hydrogen, straight or branched C|-Ce alkyl, straight or branched Ci-C« alkenyl, and straight of 15 branched Ci-Cô alkynyl.
[0015] Acolbifene binds to the estrogen receptor alpha with three times the affïnity of 17-beta estradiol, the native ligand (Katzenellenbogen (2011) J Med Chem 54(15):5271-82). Since anti-estrogens must compete wîth estradiol for binding to the estrogen receptor, high 20 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 *‘lail'* or R3 position as depicted in Compound F. EM-800, a pivalate prodrug of EM-652, and HCl salts of EM-652 were also described in the *503 patent [0016] Acolbifene was initially thought to be a complété anti-estrogen. However, careful studies with the rodent utérine assay and human uterine cell alkaline phosphatase assays rcvealed that It retajned some estrogen-like action, about 12% that of estradiol (Labrie et al. “The combination of a novel sélective estrogen receptor modulator with an estrogen protects the mammary gland and utérus 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 dégradation of the estrogen receptor, while acolbifene induces either no or modest receptor dégradation. Raloxifene and bazedoxifene don’t dégradé 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 mctastatic estrogen receptor positive breast cancer.
[0018] Starting in 2005, Blizzard and coworkers at Merck published a sériés 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 (Sélective Estrogen Receptor Alpha Modulators) (Blizzard et al. (2005) Bioorg Med Chem Lett. 15(1): 107* 13).
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 pyrrol idinyl and 3,4-methyl pyrrolidinyl as well as the 25 a-methyl (i.e., a methyl on the α-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.
-8[0020] Blizzard et al. also studied a chromane core (Blizzard et al. (2005) Bioorg
Med Chem Lett. 15(6):1675-81) (Compound K).
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-methyl pyrrolidine (but not a3-methyl) with a methyl on the linker created a nearly complété anti-estrogen, (see compound 12 of the Blizzard et al. paper). Blizzard et al. commcnted on the différences among anti-cstrogenic activîties of variousty 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 sériés (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 hâve 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 sériés was determined to hâve 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 structura! modification of an exisîting SERM resulted in a conversion of a SERM to a SERAM/SERD (Sélective 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, bazedoxïfene, 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 transférable to other platforms.
[0024] In yet another 2005 research publication. Gauthier, Labrie and colleagues reported the synthesis and structure-activity relationshîps 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 creatîng analogs in which the terminal piperidine was either replaced by pyrrolidine or substituted in various ways. Ail of these analogs proved to be more estrogenic than acolbifene as revealed by the rodent utérus assay. This expérience suggests that improvement of the anti-estrogenicity of acolbifene will be a challenge and modifications provide unpredictable results.
[0025] Blizzard reviewed lhe Merck research on anti-estrogens in 2008 (Curr Top
Med Chem. 8(9):792-812). He noted that:
“Sélective Estrogen Receptor Modulators (SERMs) hâve been the subject of extensive médicinal chemistry efforts at several pharmaceutical companies, including Merck.....The Merck SERM project involved a large number of talented and dedicated chemists and biologists who workcd for several years to discover novel classes of SERMs with a range of selectivities....no drugs hâve yet reached the market as a resuit 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 încluded 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 3R methyl pyrrolidine terminus, showed anti-estrogenicity, although not as complété as fulvcstrant in lhe rodent utérus assay. A chiral methyl on atom 2 of lhe flexible linker also conferrcd improved anti-estrogenicity. The two featurcs togclher in a doubly substituted side chain conferrcd anti-estrogenicity that was similar to fulvestrant. Unfortunately the Merck core had problematic reactive métabolites 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 (J.S. Provisional Application
- 1061/353,531 titled “Estrogen Receptor Modulators and Uses Thereof' filed in June 2010. Aragon disclosed large genuses of benzopyran dérivatives and at least 71 acolbifene analogs intended for treatment of tamoxifen résistant breast cancer. Aragon appears to hâve 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 dinic for late stage progressive metastatic disease.
[0028] Bazedoxïfene is a SE RM, under development for prévention and treatment of postmenopausal osteoporosis (Biskobing, D. M. (2007) Clinical interventions in aging 2 (3): 299-303). Lasofoxîfene 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).
[0029] U.S. Patent 5,254,568 discloses benzopyrans as anti-estrogenic agents.
[0030] WO2010/145010 discloses a combination of SERM and sex steroid precursor for treating hot flash es and other symptoms.
[0031] W02004/091488 discloses benzopyrans as estrogen receptor modulators.
[0032] U.S. Patent 5,840,735 discloses benzopyrans as sex steroid activity înhibitors.
[0033] U.S. Patent 6,262,270 discloses a method for the enantiomeric synthesis of acolbifene dérivatives.
[0034] The object of the présent invention is to provide a new improved antiestrogenic compound for the treatment of medical disorders that are mediated or affectcd by an estrogen receptor and pharmaceutical compositions and uses thereof.
SUMMARY OF THE INVENTION [0035] The présent invention is based on the discovery that a spécifie benzopyran (in the form of a mixture of S-C2 and R-C2 diastereomers and also its pure S-diastereomer) has an unexpected combination of improved properties for the treatment of medical disorders that are modulated or affected by an estrogen receptor. It was surprisi ngly found that a benzopyran core with the combination of: i) a mono-substiluted 3-methylpyrrolidyl in the side chain, ii) wherein the 3’methylpyrrodinyl is in the R-stereoconfiguration, iii) hydroxyl groups are positioned on the 7 and 4'-position and iv) wîih no methyl substitution in the linker moiety provides an optimal anti-estrogenic effect with almost no estrogenic activity. It is surprising that in none of the publications of the years of research at Merck nor in the Aragon PCT application WO2011/156518 was the optimal species disclosed or taught.
-11[0036] The compounds are 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-lyl]ethoxy|phenyI)-2H-chromen-7-ol. The chemical name for OP-1074 is (2S)-3-(4hydroxypheπyl)-4-methyl-2-(4-^2-[(3R)-3-methylpyIτolidin-l·yl]ethoxy|phenyl)-2Hchromen-7-ol. OP-1074 exhibits essentially no estrogenic activity in the alkaline phosphatase assay in ECC-I cells. In addition, OP-1074 is apureanti-estrogen when tested in theagonist mode and a complété anti-estrogen when tested in the antagonist mode.
[0037] The general structure methyl-1 -pyrrolidinyl can refer to 2-methyl or 3-methyl pyrrolidinyl (wherein the methyl group is attached to the second or third carbon in the pyrrolidine ring) and in each, because the carbon attached to the methyl is chiral, there are possible R and S stereoconfigurations for each. The spécifie benzopyrans, OP-1038 and pure
S-form OP-1074 hâve a 3-R-methyl-pyrrolidinyl. It has been discovered, contraiy to the Merck and Aragon teachings, that a single substitution in the side chain of a benzopyran, in the very spécifie position of a methyl in the 3-position of a pyrrolidinyl ring, and with R stereochemistry in combination with an unsubstituted linker group has excellent antiestrogenic properties with the minimal estrogenic effect. As any degree of estrogenic activity may provide risk to a patient with estrogen-receptor positive cancer, a decrease in estrogenic effect can be therapeutically important and represents an advancc in the art.
[0038] The active compound can be provided if desired as a phannaceutically acceptable sait, solvaté, hydrate, prodrug, stereoisomer, tautomer, N-oxide or R1 and/or R2substituted dérivative optionally in a phannaceutically acceptable composition to treat a disorder that is modulated or affected by an estrogen receptor, including those treatable with an anti-estrogenic compound optimally with virtually no estrogenic effect.
[0039] OP-1038 and OP-I074 hâve two chiral carbons and thus there are four possible diastereomers. The chiral carbon at the C2 position is in the S-configuration in OP1074 (the same configuration in EM-652, acolbifene) and is a mixture of R and S ίη ΟΡΙΟ I038.
[0040] As Merck has previously reported, mînor modifications in the side chain or linker of anti-estrogenic agents hâve been proven to resuit in signîficant and unpredictable effects on biological activity, especially in uterine tissue. The inventors hâve unexpectedly discovered a single orally available compound that provides optimal anti-estrogenic activity 15 with minimal or essentîally no estrogenic effect.
[0041] The prior art, and notably the Merck results, suggest that achieving fui I antiestrogenic properties requires two features in the molécule. First, the core must be modified from the original acolbifene core by moving the hydroxyl from position 7 to position 6 to resemble the two cores in which the Merck team had success - even if that might confer a 20 diminution of binding affinity for the estrogen receptor. Second, the prior art taught that optimal anti-estrogenicity would be achieved by appending a side chain with a 2- or 3-methyl pyrrolidîne terminus and a chiral methyl on the linker. That a single substitution would be
-13însufficient (as opposed to a significant improvement, as seen here) is strongly suggested by the expérience of the Merck team both with the benzoxathiin core and especially with the chromane core. When a 3-mcthyl pyrrolidine is appended to the chromane core the subséquent compound (number 6, in Blizzard 10) is a partial anti-estrogen with substantial 5 estrogen-like activity, 31 % of that of estradiol, in the rat uterine weight gain assay.
[0042] Further, the Aragon PCT WO2011/156518 follows the teachings of Merck wherein of its 71 disclosed benzopyran species, 68 hâve the hydroxyls in the 6 and 4’positions. Of the others, only one Aragon species has hydroxyls located at the 7 and 4'posilions, and that compound has a methyl in lhe linker as well as on the pyrrolidine ring 10 (again following the Merck teaching) (Aragon*s Compound 28). Figure 3 illustrâtes that the compound of the présent invention has less estrogenic activity than Aragon Compound 28. [0043] OP-1038 and OP-1074 also induce substantial dégradation of the estrogen receptor, comparable to that of fulvestrant and Aragon Compound 28.
[0044] The addition of a methyl modification to the linker of a compound with lhe 315 methyl pyrrolidine, contrary to expectations raised by the Merck expérience, actually make s the doubly modified molécule (Compound 28 of WO2011/156518) more estrogenic.
[0045] OP-1038, OP-1074 and their prodrugs (including esters, carbonates and phosphates), dérivatives and their salts are complété anti-estrogens useful to treat locally advanced or metastatic breast cancer that is positive for expression of estrogen receptors, 20 progestérone receptors or both (receptor positive advanced breast cancer). In an alternative embodiment, the compound is used to treat estrogen or progestérone receptor négative breast cancer. The compound can be used as the initial treatment of advanced breast cancer în patients who hâve never received previous hormonal therapy for advanced breast cancer, either by itsclf or in combination with one or more other anti-cancer agents, including 25 targeted thérapies. 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 anticanccr agent, for example, a targeted therapy such as an mTOR inhibitor such as everolimus.
[0046] The compounds of the invention are also useful as adjuvante therapy after surgery to preveni rucurrance. Such adjuvant use is often administered for several years, for 30 instance 5 years, or even up to 10 years after surgery and associated chemotherapy and radiotherapy hâve been concluded.
[0047] The compounds of the invention are also useful for lhe prévention of breast cancer in women al high risk and can be taken for any desired lime period, including indefinitely. For example, a patient, typically a woman, witha family history of breast
cancer, or who has been determined to carry a mutation in the BRACAI or BRACA2 gene or other genes that prédisposé a patient to breast cancer may choose to use such preventative treatment instead of a mastectomy or other intervention. The compounds described herein are also useful as neoadjuvants to shrink large tutnors prior to surgical removal, both to enable 5 breast conservative surgery and to reduce the risk of récurrence. In addition to breast cancer these compounds also are useful to treat other cancers and other overgrowth diseases of the female reproductive tract including ovarian, endométrial, and vaginal cancer and endometriosis. Besides these reproductive tissues the compounds are useful in treating lung cancers that are positive for estrogen or progestérone receptors.
[0048] Sélective estrogen receptor tnodulators (SERMS) are useful for hormonal therapy for postmenopausal women in particular to treat or prevent osteoporosis. In one embodiment, a compound of the présent invention is used in combination with an estrogen, SERM or partial anti-estrogen such that the complété anti-estrogen prevents adverse action of the total or partial estrogen on the utérus and other tissues.
[0049] Other objects and advantages will become apparent to those skilled in the art from a considération of the ensuing detailed description. Ail variations and modifications of the disclosed invention are considered within the scope of this invention.
BRIEF DESCRIPTION OF THE FIGURES [0050] Figure 1. Représentative compounds of the présent invention (Figure I A) as well as représentative prodnigs and salts of OP-1038 and OP-1074 (Figure IB). The IUPAC naines of the compounds are;
OP-1038 3-(4-hydroxyphcnyl)-4-methyl-2-(4-(2-[(3R)-3-methylpynOlidin-ly I ]e thoxy ] pheny l)-2H-ch rome n -7-ol
OP-1039 3-(4-hydroxyphcnyl)-4-rnclhyl-2-(4-( 2-[(2R)-2-rnethylpynOlidin-lyl Jethoxy ] phenyl )-2H-chromen-7-ol
OP-1040 3-(4-hydroxyphenyl)-^4-rnelhyl-2-(4-[2-(pyrrolidin-l-yl)ethoxy]phcnyl )2H-chromen-7-o]
OP-1042 3-(4-hydroxyphenyl)-4-niethyl-2-(4-[(2R)-2-((3R)-3-methylpyrrolÎdin-ly I ] propoxy] phenyl ] -2H-chromen-7-ol
OP-1046 3-(4-hydroxyphenyl)-4-niethyl-2-(4-(2-[(3S)-3-methylpyrrolidin-l yljethoxy ] phenyl )-2H-chromen-7-ol
OP· 1047 3-(4-hydroxyphenyl)-4-methy 1-2-(4- ( 2-[(2S)-2-methylpynOlidin-1 yljethoxy | phenyl)-2H-chromen-7-ol
OP-1049 3-(4-hydroxyphenyl)-4-methyl-2-(4-[(2S)-2-(pyrrolidin-lyl)propoxy]phenyl )-2H-chromen-7-ol
- 15OP-1050 3-(4-hydroxyphenyl)-4-methyl-2-{4-[(2S)-2-(piperidin-1yl)propoxy]phenyl]-2H-chromen-7-ol
OP-1053 3-(4-hydroxyphenyl)-4-methyl-2-[4-[(2S)-2-[(2R)-2-methylpyrrolidin-lyl]propoxy]phenyl J -2H-chromcn-7-ol
OP-1056 3-(4-hydroxyphenyl)-2-(4-{ 2-[(3R)-3-methy!pyrTolidin-1 yl]ethoxy) phenyl )-4-(trifluoromethyl)-2H-chromen-7-ol
OP-1060 5-fluoro-3-(4-hydroxyphenyl)-4-methyl-2-{4-[(2S)-2-I(3R)-3methylpyrrolidin-1 -yl]propoxy]phenyl |-2H-chromen-7-ol
OP-1061 5-fluoro-3-(4-hydroxyphenyl)-4-methyl-2-(4- {2-[(3R)-3-methylpyrrolidinl-yl]ethoxy]pheny!)-2H-chromen-7-ol
O P-1074 ( 2S )-3-(4-hydroxy phenyl )-4-meth y! -2-(4- {2-[( 3 R)-3- met hyl pyrro I idin-1 yljethoxy ] phenyl)-2H-chromen-7-ol
OP-1075 (2R)-3-(4-hydroxyphenyl)-4-methyl-2-(4- ( 2-[(3R)-3-methy!pyrro!idin-1yljethoxy | phenyl)-2H-chromen-7-ol [0051] Figure 2. Alkaline phosphatase (AP) activity in ECC-1 cells correlates with the uterine wet weight of ovariectomized rats, as reported prevïously in Scafonas, et al. (*; Scafonas et ai. “Agonist-like SERM effects on ER alpha-mediated repression of MMPI promoter activity predict in vivo effects on bone and utérus. J Steroid Biochem Mol Biol. 2008 110(3-5): 197-206) and Labrie, et ai. (t; Labrie et al. “The combination of a novel sélective estrogen receptor modulator with an estrogen protects the mammary gland and utérus in a rodent mode!: the future of postmenopausal women's health? Endocrinology. 2003 144(11):4700-6). ECC-1 cells were treated with 500pM 17P-estradiol (E2) or 1-5 nM anti-estrogens in hormone-depleted medium for 3 days. Note that the racemic mixture of acolbifene, EM-343, was used in the in vitro AP assay. AP activity was measured by incubating a chromogenic AP substrate, p-nitrophenyl phosphate, at 40*C for 40 minutes followed by measuring the absorbance at 405nm. Results were from a single représentative experiment and reported as the mean percent induction relative to E2 from triplîcate treatments, with error bars representing SEM.
[0052] Figure 3. OP-1038 and OP-1074 lack estrogen ic activity in the alkaline phosphatase (AP) assay in ECC-I cells. Comparison of estrogenic-like AP activity of various benzopyran compounds (right) compared to published reference compounds (left). ECC-I cells were treated with ΙΟΟηΜ anti-estrogens and AP activity was measured as described in Figure 2. Results were from a single représentative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing SEM. Note that OP-1038 was statistically different from Aragon Compound 28 (WO2011/156518), EM-343 and Gauthiercompound la (p<0.0l (exact p values stated in graph), calculated using Student’s t-test).
-16[0053] Figure 4. OP-1038 is less estrogenic than Aragon Compound 28 at three different doses in the AP assay in ECC-I cells. AP activity was measured as described in Figure 2. Results were from a single représentative experiment and reported as the mean percent induction relative to E2 from sextuplicate treatments, with error bars representing 5 SEM. Note that OP-1038 was statistically different from Aragon Compound 28 at ail three doses tested ((p< 0.001 (exact p values stated in graph), calculated using Student’s t-test). [0054] Figure 5. OP-1038 lacks estrogenic activity in the AP assay in ECC-1 cells, in contrast to other mono-methyl substituted pyrrolidines. AP activity was measured as described in Figure 2. Results were from a single représentative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing SEM.
[0055] Figure 6. OP-1038 and OP-1074 inhibit E2-stimulated AP activity in ECC-l cells. A) Comparison of inhibition of E2-stimulated AP activity of various benzopyran compounds(right)compared to published référencécompounds(left). ECC-l cells were treated with ΙΟΟηΜ anti-estrogens in the presence of 500 pM E2 and AP activity was measured as described in Figure 2. Note that OP-1038 was statistically different from EM343 and Gauthier compound la (p< 0.0001 (exact p values stated in graph), calculated using Student’s t-test). Results were from a single représentative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing 20 SEM. B) Comparison of potency and efficacy of OP-1038 to Aragon Compound 28 in inhibiting E2-stimulated AP activity in ECC-l cells. Results were from a single représentative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing SEM. IC50's were calculated using the least squares fit method. *Aragon Compound 28 was statistically different from équivalent dose of 25 OP-1038 (p< 0.01, calculated using Student’s t-test). C) Detail of 100 nM dose in B indicating a statistical différence between Aragon Compound 28 at saturatîngdose.
[0056] Figure 7. Comparison of potency and efficacy of OP-1074 to EM-652 in the APassay in ECC-l cells. A) EM-343 and ils active isomer EM-652 are more estrogenic than OP-1038 and OP-1074 in the AP assay. AP activity was measured as described in Figure 2.
Results were from a single représentative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representing SEM. B) OP1038 and OP-1074 are more anti-estrogenic than EM-343 and EM-652 in the presence of 500 pM E2. Results were from a single représentative experiment and reported as the mean
-I7percent induction relative to E2 from triplicate treatments, with errer bars représentât!g SEM. ICSO’s were calculated using the least squares fit method.
[0057] Figure 8. OP-1074 is a pure anti-estrogen in the agonist mode (without E2) and a complété anti-estrogen in the antagonist mode in the ECC-l AP assay. A) and B) OP5 1074, similar to fulvestrant, did not stimulate AP activity and inhibited E2-stimulated AP activity in a dose dépendent manner. AP activity was measured as described in Figure 2. Results were from a single représentative experiment and rcported as the mean percent induction relative to E2 from triplicate treatments, with error bars reprcsentlng SEM. C) and D) OP-1074 was confirmed to be the active diastereomer of the equal mix of two
I0 diastereomers OP-I038, while the other diastereomer, OP-I075, had reduced activity. Results were from a single représentative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with error bars representîng SEM. [0058] Figure 9. OP-1074 is an essentially pure anti-estrogen when tested in the agonist mode and a complété anti-estrogen when tested in the in the antagonist mode on the mouse utérus. Uterine wet weight was measured at the end of three days after treating ovariectomized BALB/c mice q.d. with vehicle or one of the following treatments (10 mice per group): 50mg/kg tamoxifen p.o. in 0.5% carboxymethylcellulose (CMC); 50mg/kg fulvestrant sc in 5% éthanol; lOOmg/ml OP-I038 p.o. in 0.5% CMC. Half the animais in each group were co-treated with 0.1 pg/ml E2 sc in cotlonseed oil-ethanol (95:5), or vehicle alone.
Animal experiments were conducted at the University of California, San Francisco following institutional animal care and use committee protocol s. OP-1038 was not significantly different from control or fulvestrant (determined by one way ANOVA at p>0.05) in the agonist mode. OP-1038 +E2 was not different from fulvestrant in the antagonist mode or from the control without E2.
[0059] Figure 10. OP-I038 and OP-1074 are potent antagonists of E2-stîmulated estrogen response element (ERE)-regulated reporter gene activity. A) OP-1074 had similar potency to model anti-estrogens in the ERE reporter gene assay. MCF-7 cells were transiently transfccted with ERE-tklO9-Luc and treated with anti-estrogens in hormone-depleted medium in the presence of 100 pM E2 for 22 hours, Results were from a single représentative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, witherrorbars representîng SEM. B) OP-1074 was confirmed to be the active diastereomer of the equal mix of two diastereomers OP-1038, while the other diastereomer, OP-I075, had reduced activity in the ERE reporter gene assay. Results were from a single représentative experiment and reported as the mean percent induction relative to E2 from
triplicate treatments, with errer bars representing SEM. IC50's were calculated using the least squares fit method.
[0060] Figure 11. OP-1038 and OP-1074 are potent antagonïsts of E2-stimulated prolifération in human MCF-7 breast cancer cells. A) OP-1074 had similar potency to model anti-estrogens in inhibiting E2-stimulated prolifération of breast cells in vitro. MCF-7 cells were treated with anti-estrogens in hormone-depleted medium for 6-8 days in the presence of 100 pM E2. Prolifération was measured using Cyquant fluorescent DNA-binding dye (Invitrogen, Grand Island, NY). Results were from a single représentative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with errer bars representing SEM. B) OP-1074 was confirmed to be the active enantiomer of the dïastereomer OP-1038, while the other diastereomer, OP-1075, had reduced activity in inhibiting E2-stimulated prolifération of breast cells in vitro. As shown, OP-1075 is more estrogenic than OP-1074. Results were from a single représentative experiment and reported as the mean percent induction relative to E2 from triplicate treatments, with errer bars representing SEM, lC50’s were calculated using the least squares fit method.
[0061] Figure 12. OP-1074 and OP-1038 induce dégradation of estrogen receptor alpha (ERa) in human breast and endométrial cells. A) ERa levels in breast cells after treatment with OP-1074 compared to treatment with model anti-estrogens. MCF-7 cells were treated with 100 nM anti-estrogen for 24 hours in serum-free medium and protein extracts 20 immunoblotted with an antibody to ERa (DI 2, Santa Cruz Biotechnology, Santa Cruz, CA).
Image is from a single représentative experiment and number on top dénotés the mean percent ERa expression relative to vehicle from quadruplicate treatments. B) OP-1038 induccs dégradation of ERa in both MCF-7 breast cells and ECC-1 endométrial cells.
[0062] Figure 13. OP-1074 induces rapid and complété régression of MCF-7 clone 18
HER2/neu xenografts stimulated by estrogen. A) Percent change in tumor volume of human
MCF-7 HER2/neu clone 18 xenografts in ovariectomized athymie nude mice implanted with estrogen pellets and treated with either tamoxifen (by oral gavage), fulvestrant (Faslodex) by s.c. injection, or OP-1074 (by oral gavage). B) Weîghts of the animais treated as above. C) Waterfall plot of final tumor volumes compared with volumes at the start of treatment.
-19DETAILED DESCRIPTION OF THE INVENTION [0063] The présent invention is based on the discovery that a spécifie benzopyran (in the form of a C2 equal mix of diastereomers OP-1038 and its pure S-diastereomer OP-1074) has unexpectedly improved properties for the treatment of medical disorders that are mediated, modulated or affected by an estrogen receptor, including breast cancer.
[0064] The compound can be provided if desired as a pharmaceutically acceptable sait, solvaté, hydrate, prodrug, stereoisomer, tautomer, N-oxide or R1 and/or R^substituted dérivative 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.
»
OP-1074 .or
wherein R1 and R2 are independently either:
(i) R9 which is independently selected from H, halogen (Cl, Br, I or F), natural or nonnaturally occurring amino acid (bound through either the OC(O)- or C(O)O- (an ester) or the amino (through either -C(O)-N- or-N-C(O)- (an amide linkage)), R10, -OR10, or -SR10 where R10 is -C(=O)Rcl, -C(=O)ORcl, -C(=O)SRCI, -C(=O)N(Rc,)2; or polyethylene glycol, substituted or unsubstituted alkyi, 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(=O)2RCI, -S(=O)iORcl, -S(=O)RCI, -S(=O)ORCI, -P(=O)2RCI, -P(=O)2ORCI, -P(=O)(ORCI )2, -P(=O)(RC,)21 or-P(Rcl)(ORcl); 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 -NH- on administration);
and Ra can be independently selected from hydrogen. polyethylene glycol, substituted or unsubstituted alkyi, 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 RC1 groups are Joined to form an substituted or unsubstituted heterocyclic ring.
In certain embodiments either or both of R1 or R2 is an ester, amide, carbonate or phosphate.
[0065] Spécifie examples of prodrugs of the described compounds are:
S80l*dO
7801'dO
980L-dO
-tz16917
[0066] Examples of useful metabolically cleavable prodrug groups inciude acetyl, methoxycarbonyl, benzoyl, methoxymethyl and tri methyl sil y 1 groups [0067] The compounds of the invention can be administered in a pharmaceutical composition suitable for oral delivery to the patient, typically a human. Alternatively, the compounds can be delivered in u carrier suitable for topical, transdermal (including by patch), intravenous, parentéral, 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.
[0068] ln yet another aspect, the compounds of the invention can be used to prevent a disorder modulated through the estrogen receptor, which comprises administering to a patient in need of such prévention, a prophylactically effective amount of a compound or pharmaceutical composition.
[0069] The compounds of the invention can be ln the form of a sait. They can be administered as a pharmaceutically acceptable sait, for example, a pharmaceutically acceptable acid addition sait, including a hydrochloride, hydroiodide, hydrobromide, nitrate, sulfate, bisulfate, phosphate, acetate, lactate, citrate, tartrate, succinate, malcate, fumarate, benzoate, para-loluenesulfonate and the like.
-23[0070] The compounds are used to treat or prevent a disorder modulated by the estrogen receptor in an animal, typicaliy a mammal, and most typicaliy a human.
[0071] In yet another aspect, the présent invention provides a combination of a compound of the instant invention, and another pharmacologîcally active agent.
[0072] The compounds can also be used as adjunctive therapy or combination therapy with another active agent. For example, a therapeutically effective amount of the compound can bc used in combination with another anti-cancer agent, especially for estrogen receptor positive breast cancer, but in some embodiments, for estrogen receptor négative breast cancer.
[0073] Additional embodiments within the scope provided herein are set forth in non-limiting fashion elsc where herein and in the examples. It should bc understood that these examples are for illustrative purposes only and are not to be construed as limiting in any manner.
PHARMACEUTICAL COMPOSITIONS [0074] In one aspect, the invention provides a pharmaceutical composition comprising a pharmaceutically effective amount of a compound of the présent invention and a pharmaceutically acceptable carrier.
[0075] The compounds provided herein are administered for medical therapy in a therapeutically effective amount. The amount of the compound administered will typicaliy be determined by a physician, In the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the compound administered, the âge, weight, and response of the individual patient, the severity of the patient’s symptoms, and the like.
[0076] The pharmaceutical compositions provided herein can be administered by a variety of routes including oral, topical, parentéral, 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.
[0077] The compositions for oral administration can take the form of bulk liquid solutions or suspensions, or bulk powders. Typicaliy, the compositions are presented in unit dosage forms to facîlitate accurate dosing. The term “unit dosage forms” refers to physically discrète units suitable as unitaiy dosages for human subjects and other mammals, each unit containing a predetermïned quantity of active material calculated to produce the desired
-24therapeutic effect, in association with a suitable pharmaceutical excipient. Typical unit dosage forms include prefilled, pre measured 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 examplc, 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 processîng aids hclpful for forming the desired dosing form.
[0078] 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 ingrédients, 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 alginîc acid. Primogel, or com starch; a lubricant such as magnésium stéarate; a glîdant such as colloïdal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyi salicylate, or orange flavoring, [0079] Injectable compositions are typically based upon injectable stérile saline or phosphate-buffered saline or other injectable carriers known in the art.
[0080] Transdermal compositions arc typically formulated as a topical oîntment 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 oîntment, the active ingrédients will typically be combined with either a suitable delivery polymeric composition, or a paraffinic or a water-miscible oîntment base. Altematively, lhe active ingrédients 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 ingrédients to enhance the dermal pénétration of stability of the active ingrédients or the formulation. Ail such known transdermal formulations and ingrédients are ïncluded within the scope provided herein.
[0081] The compounds provided herein can be administered by a transdermal device.
Transdermal administration can be accomplished using a patch either of the réservoir or porous membrane type, or of a solid matrix variety.
[0082] The abovc-described components for orally administrable, injectable or topically administrable compositions are merely représentative. Other materials as well as processîng techniques and lhe like are set forth in Part 8 of Remington's Pharmaceutical ί
I
-25Sciences, 17th édition, 1985, Mack Publîshing Company, Easton, Pennsylvania, which is incorporated herein by reference.
[0083] The compounds of this invention can also be administered în sustained release forms or from sustained release drug delivery Systems. A description of représentative sustained release materials can be found in Remington's PharmaceuticalSciences. [0084] In certain embodiments, the formulation comprises water. In another embodiment, the formulation comprises a cyclodextrin dérivative, b certain embodiments, the formulation comprises hexapropyl-0-cyclodextrin. In a more particular embodiment, the formulation comprises hexapropyl-0-cyelodextrin (10-50% în water).
[0085] The présent invention also includes pharmaceutically acceptable acid addition salts of compounds of the compounds of the invention. The acids which are used to préparé the pharmaceutically acceptable salts are those which form non-toxic acid addition salts, i.e. salts containing pharmacologically acceptable anîons such as the hydrochloride, hydroiodidc, hydrobromide, nitrate, sulfate, bisulfate, phosphate, acetate, lactate, citrate, tartrate, succinate, 15 maleate, fumarate, benzoate, para-toluenesulfonate, and the like.
[0086] The following formulation examples illustraie non-limiting représentative pharmaceutical compositions that may be prepared in accordance with this invention for the purpose of illustration only. The présent invention is specifically not limited to the following pharmaceutical compositions.
Formulation 1 - Tablets [0087] A compound of the invention may be admixed as a dry powder with a dry gelatin binder în an approximate 1:2 weight ratio. A minor amount of magnésium stéarate is added as a lubricant. The mixture is formed into 240-270 mg tablets (80-90 mg of active compound per tablet) in a tablet press.
Formulation 2 - Capsules [0088] A compound of the invention may be admixed as a dry powder with a starch diluent in an approximate 1:1 weight ratio. The mixture is filled into 250 mg capsules (125 mg of active compound per capsule).
Formulation 3 - Liquid [0089] A compound of the invention ( 125 mg) may be admixed with sucrose ( 1.75 g) and xanthan gum (4 mg) and the résultant mixture may be blended, passed through a No. 10 mesh U.S. sieve, and then mixed with a previously made solution of microcrystailine cellulose and sodium carboxymethyl cellulose (11:89,50 mg) in water. Sodium benzoate (10
mg), flavor, and color arc diluted with water and added with stirring. Sufficient water may then be added to produce a total volume of 5 mL.
Formulation 4 * Tablets [0090] A compound of the invention can be admixed as a dry powder with a dry gelatin binder in an approximate 1:2 weight ratio. A minor amount of magnésium stéarate is added as a lubricant. The mixture îs formed into 450-900 mg tablets (150-300 mg of active compound) in a tablet press. In other embodiments, there is between 10 and 500 mg of active compound in the oral tablet.
Formulation 5 - Injection [0091] A compound of the invention can be dissolved or suspended în a buffered stérile saline injectable aqueous medium to a concentration of approximately 5, or 10, or 15, or 20, or 30 or 50 mg/mL.
Formulation 6 - Tablets [0092] A compound of the invention may be admixed as a dry powder with a dry gelatin binder in an approximate 1:2 weight ratio. A minor amount of magnésium stéarate is added as a lubricant. The mixture îs formed into 90-150 mg tablets (30-50 mg of active compound per tablet) in a tablet press.
Formulation 7 * Tablets [0093] A compound of the invention may be admixed as a dry powder with a dry gelatin binder in an approximate 1:2 weight ratio. A minor amount of magnésium stéarate is added as a lubricant. The mixture is formed into 30-90 mg tablets (10-30 mg of active compound per tablet) in a tablet press.
Formulation 8 - Tablets [0094] A compound of the invention may be admixed as a dry powder with a dry gelatin binder in an approximate 1:2 weight ratio. A minor amount of magnésium stéarate is added as a lubricant. The mixture is formed Into 0.3-30 mg tablets (0.1-10 mg of active compound per tablet) in a tablet press.
Formulation 9 - Tablets [0095] A compound of the invention may be admixed as a dry powder with a dry gelatin binder in an approximate 1:2 weight ratio. A minor amount of magnésium stéarate is added as a lubricant. The mixture is formed into 150-240 mg tablets (50-80 mg of active ' compound per tablet) in a tablet press. f
I f
-27Formulation 10 - Tablets [0096] A compound of the invention may be admixed as a dry powder with a dry gelatin binder in an approxîmate 1:2 weight ratio. A miner amount of magnésium stéarate is added as a lubricant. The mixture is formed into tablets (5-1000 mg of active compound per 5 tablet) in a tablet press.
USE OF COMPOUNDS IN MEDICAL THERAPY [0097] OP-1038, OP-1074 and their prodrugs (including esters, carbonates and phosphates), dérivatives and their salts as described herein are complété anti-estrogens useful to treat any disorder modulated, mediated or affected by the estrogen receptor.
[0098] In one embodiment, the compound is used in combination or alternation with another anti-cancer agent for the treatment of cancer, as described more fully below. In another embodiment, the compound in combination or alternation with estrogen or a partial 15 estrogen receptor angatonîst for the treatment of a postmenopausal disorder, also described below.
[0099] ln one embodiment, local, a compound of the présent invention is use to treat local, advanced or metastatic breast cancer that is positive for expression of estrogen receptors, progestérone receptors or both (receptor positive advanced breast cancer). In an 20 alternative embodiment, the compound is used to treat estrogen or progestérone receptor négative breast cancer. The compound can be used as the initial treatment of advanced breast cancer in patients who hâve never received previous hormonal therapy for advanced breast cancer, either by Itself or 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 anticancer agent, for example, a targeted therapy such as an mTOR inhibitor such as everolimus.
[00100] The compounds 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 30 years, perhaps 5 years, after chemotherapy or other thérapies hâve been conduded, but may optimallybe continued for additional years.
[00101] The compounds of the invention are also useful for the prévention of breast cancer in women at high risk and can be taken for any desired time period, including indefïnitely. For example, a patient, typically a woman, with a family historyof breast
-28cancer, or who has been determined to carry a mutation in the B RAC Al or BRACA2 gene gene or other genes that prédisposé a patient to breast cancer may choose to use such preventative treatment instead of a mastectomy or other intervention. The compounds 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 récurrence. In addition to breast cancer these compounds also are useful in treating other cancers and other overgrowth diseases of the female reproductive tract including ovarian, endométrial, and vaginal cancer and endometriosis. Besides these reproductive tissues the compounds are useful în treating lung cancers that are positive for estrogen or progestérone receptors.
[00102] Sélective estrogen receptor modulators (SERMS) are useful for hormonal therapy for postmenopausal women in particular to treat or prevent osteoporosis. In one embodiment, a compound of the présent invention is used in combination with an estrogen, SERM or partial anti-estrogen whcreby the complété anti-estrogen prevents adverse action of the total or partial estrogen on the utérus and other tissues.
[00103] The présent compounds are used as therapeutic or prophylactic agents for the treatment of conditions in mammals, particularly humans that are moduiated by estrogen receptors.
[00104] An oral complété anti-estrogen is useful for treating locally advanced or metastatic breast cancer, preventing récurrence or early breast cancer after surgery, and 20 preventing breast cancer in women at high risk. Il is useful for treating all estrogen-dependent cancers of the reproductive tract including endométrial and ovarian cancers. It has potential uses in the treatment of lung and bronchial cancers that express estrogen receptors.
Sélective estrogen receptor modulators (SERMS) such as tamoxifen, raloxifene, lasofoxifene, and bazedoxifene additionally hâve application as hormone replacement therapy to prevent 25 osteoporosis and other disorders such as hot flashes, etc. in post-menopausal women, a use that dépends on their partial estrogen like action, for example, on bone. The compounds described herein can be employed in combination with an estrogen or a sélective estrogen receptor modulator to block the unwanted estrogen ic activity of the therapy. The complété anti-estrogen is dosed in the amount to prevent the adverse action of the estrogen or estrogen 30 receptor modulator on the utérus and mammary gland yet allowing the bénéficiai action of estrogen on bone and vasomotor symptoms.
[00105] The compounds of the présent invention can be administered for the treatment of cancer, and in particular breast cancer in combination or association with Herceptin, Tykerb, CDK4/6 inhibitor such as PD-0332991, mTOR inhibitor such as Novartis'
-29everolimus and other rapamycin analogs such rapamycin and temsirolimus, Millennium’s MLN0128 TORC1/2 inhibitor, an EFGR-family inhibitor such as trastuzumab, pertuzumab, trastuzumab, emtansine, erlotinib, gefitinib. neratinib and similar compounds, a PI3 Kinase Inhibitor such as perifosene, CAL101, BEZ235, XL147, XL765, GDC-0941, and IPI-145, a histone deacetylase inhibitor such as vorinostat, romidepsin, panobinostat, valproic acid, etinostat, and bclinostat.
[00106] In another method of treatment aspect, provided herein is a method of treating a mammal susceptible to or afflicted with a condition related to estrogen receptor.
[00107] In another embodiment, the compounds of the présent invention are provided for use in medical therapy, including for any of the conditions described herein. The use of the présent compounds in the manufacture of a médicament for the treatment or prévention of one of the aforementioned conditions and diseases is also provided.
[00108] Injection dose levels range are provided in any desired dosage, for example, from about 0.1 mg/kg/hour to at least 10 mg/kg/hour, ail for from about 1 to about 120 hours 15 und 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 adéquate steady state levels. The maximum total dose is not expected to exceed about 2 g/day for a 40 to 80 kg human patient.
[00109] For oral dosing, any dose is appropriate that achieved the desired goals. In one example, suitabie daily dosages 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 o rail y oncedaily, 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, 25 200.250, 1000 or up to about 2000 mg per day.
[00110] For the prévention and/or treatment of long-term conditions, such as neurodegenerative and autoimmune conditions, the regimen for treatment usually stretches over many months or years. Oral dosing may be preferred for patient convenience and tolérance. With oral dosing, one to five and especially two to four and typically three oral 30 doses per day are représentative regimens. Using these dosing patterns, nonlîmiting dosages might range from about 0.01 to about 20 mg/kg of the compound provided herein, with preferred doses each providing from about 0.1 to about 10 mg/kg and especially about 1 to about 5 mg/kg.
-30[00111] Transdermal doses are generally selected to provide similar or lower blood levels than are achieved using injection doses.
[00112] When used to prevent the onset of cancer, a neurodegenerative, autoimmune or inflammatory condition, the compounds provided herein will be administered to a patient 5 at risk for developing the condition, typically on the advice and under the supervision of a physician, at the dosage levels described above. Patients at risk for developing a particular condition generally include those that hâve a family history of the condition, or those who hâve been identified by genetic testïng or screening to be particularly susceptible to developing the condition.
[00113] The compounds provided herein can be administered as the sole active agent or they can be administered in combination with other agents. Administration in combination can proceed by any technique apparent to those of ski 11 in the art including, for example, separate, sequential, concurrent and altemating administration.
GENERAL SYNTHETIC PROCEDURES [00114] 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 (f.e., 20 reaction températures, times, mole ratios of reactants, solvents, pressures, etc.) arc given, other process conditions can also be used unless otherwise stated. Optimum réaction 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.
[00115] 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 deproteclion 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 référencés cited lherein.
[00116] 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 préparation of représentative substituted benzopyrans that hâve been listed herein. The
compounds provided herein may be prepared from known orcommercially available starting materials and reagents by one skilled in the art of organic synthesis.
[00117] 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 race mate or mixture of diastereomers by any conventional technique, for example, by chromatographie resolution using a chiral column, TLC or by the préparation of diastereoisomers, séparation thereof and régénération of the desired enantiomer or diastereomer. See, e.g., Enantiomers, Racemates and Résolutions, 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 Opticai Résolutions 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,
Mihâly Nôgrâdi (1995 VCH Publishers, Inc., NY, NY).
[00118] In certain embodiments, a diastereomerically pure compound of formula (1) 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, 20 Sait Forms of Drugs and Adsorptlon, 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. Ani. Chem. Soc. 95:512; and CRC Handbook of Opticai Resolution via Dlastereomeric Sait Formation, the contents of which are hereby incorporated by reference in their entireties.
[00119] Enantiomerically or diastereomerically pure compounds can also be recovered either from the crystallized diastereomer or from the molher liquor, depending on the sol ubility properties of the particular acid resolving agent employed and the particular acid enantiomer or diastereomer used. The identity and opticai purity of the particular compound so recovered can be determined by polarimetry or other analytical methods known in the art.
The dïasteroisomers 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 séparation.
-32[00120] 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 séparation of the enantiomers or diastereomers are available and suitable conditions for the séparation can be empirically determined by methods known to one of ski II in the art Exemplary chiral columns available for use in the séparation 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.
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 INTERMEDIATES [00121] The various intermediates useful for préparation of the compounds of the invention can be prepared in accordance with methods described in the art and using the appropriate reagents, starting materials, and purification methods known to those skilled in the art.
Représentative Intermediate Synthesis 1
Représentative synthesis of 4-(2-(methylpyrrolidln-l-yt)ethyl)benza!dehyde analogs
[00122] The intermediate methylpyrrolidine dérivatives can be prepared by following the représentative method described below.
-33Scheme 1
4-hydroxybenzaldehyde (5')
PPh3. DIAD
PBr3, Pyrldine ’
K2CO3. ch3cn. ethartolamine
(R)-l,4-Dîbromo-2-methylbutane (2’) [00123] To a solution of R-2-methylbutane-l,4-diol (1*) (5 g, 48 mmol) tn pyridinc (5 mL) at 0 °C is added PBr3 (9 g, 33 mmol) and the resulting yellow paste is stirred at room température for 30 minutes. The reaction mixture is heated at 100 °C for two hours. Tire cooled mixture is treated with water (50 mL) and extracted with hexanes (3 x50 mL). The combined organic extracts are washed with 5% sodium hydroxide, concentrated suIfurie acid and water and concentrated to yield a yellow oil y residue. This residue is distilled at 75-85 °C (3 mm Hg) to yield 2' as a clear colorless oil (4.5 g 42% yield).
(R)-2-(3-Methylpyrrolidin-l-yl)ethanol (4*) [00124] To a solution of 2’ (4.5 g, 19 mmol) tn acetonitrile (200 mL) and potassium carbonate (5.5 g, 30.5 mmol) is addedethanolamine (1.2 mL, 19 mmol) and the resulting suspension is heated at reflux for 48 hours. The cooled solution is filtered and concentrated. This residue is dissolved in DCM ( 100 mL) and washed with 5% aqueous sodium hydroxide (2 x 50 mL), brine, dried over anhydrous sodium sulfate, filtered, and concentrated to yield a pale yellow oil. This oil is distilled (100-110,3 mm Hg) to yield 4 as a clear colorless oil (1.4 g, 30% yield).
(R )-4-(2-(3 -Me thy Ipyrrol idin-1 -y l)c thoxy) benzaldehyde (6*a) [00125] Toasolutionof4’(l,4 g, 10.9 mmol), p-hydroxybenzaldehyde,5’, (2.0g,
16.3 mmol) and triphenylphosplùne (4.3 g, 16.3 mmol) in dichloromethane (20 mL) is added diisopropylazodicarboxylatc (2.3 mL, 16.3 mmol) dropwise at 0 °C over 30 minutes and then allowed to warm to room température and stirred for an additional two hours. The solution is washed with water, brine, dried over anhydrous sodium sulfate, filtered, and concentrated to
-34yield a pale yellow oil. This oil is purified by silica gel column chromatography using a gradient of 0 to 5% methanol in dicloromethane to yield 6’a as a clear colorless oîl (1.0 g, 39% yield).
MS Calculated C|4H|9NO3 + H+ = 235; Observed 234.
[00126] The following intermediates are or can be prepared following the method described for 6’a and using the appropriate reagents, and starting materials.
Représentative Intermediate Synthesis 2 Représentative synthesis of l-((2-Chloro-l-methyl)ethyl)-3-alkyl pyrrolidine analogs
Ak
Me
Ak - alkyl [00127] The intermediate chloroetliylalkylpyrrolidine dérivatives can be prepared by following the représentative method described in Bioorganic ά Médicinal Chemistry Letters (2005) 15 3912-3916.
[00128] Tlie synthesis of a représentative pyrrolidine dérivative, (R)-l-((S)-2-chloro-l methyl-ethyO-S-methyl-pyrrolidine is given below.
-35Intermedlate A
Scheme 2
Step-lAî [00129] A solution of (R)-2-methyl succinic acid (lg, 7.57 mmol) in 40 mLof toluene was heated at 100 °C and (S)-2-Amino-propan-l-oi (0.59 mL, 7.57 mmol) was added slowly <o the réaction mixture. After the completion of addition, the reaction mixture was further heated at 130°C for 16 h. The réaction mixture was cooled to room température, concentrated under vacuum to get the crude product, which was purified by chiral préparative HPLC to get pure IA. Yield: 0.6 g, 46%.
Step-2 A:
[00130] A solution of Intermediate IA (0.5 g, 2.92 mmol) in dry ether (10 mL) was added in drops to a cooled suspension of LAH (0.433 g, 1L7 mmol) in dry diethyl ether at 0 °C. After completionof addition, the reaction mixture was gradually allowed to reach room température and stirred for 12 h. The reaction was monitored by TLC (20% MeOH in DCM). After completion of reaction, the reaction mixture was cooled to 0 °C, quenched successively with water (0.5 mL), 10%> NaOH (1 mL) and water (IJ mL). The solid precipitated was filtered through celite and the filtrate was concentrated under vacuum to get the product which was used as such for next step. Yield: 290 mg, 70%.
Step-3A:
[00131] To a solution of Intermediate 2A (0.3 g, 2.1 mmol) in 1,2 dichloroethane (10 mL), thionyl chloride (0.18 mL, 2.5 mmol) was added drop wise at 0 “CThen the reaction mixture was gradually heated to 80 °C for 2 h. The excess solvent and thionyl chloride was concentrated under reduced pressure to get the crude Intermediate A, which was taken as such for next step (product formation was confirmed by LCMS). Yield: 300 mg, 88%.
[00132] Intermediate B The synthesis of a représentative pyrrolidine dérivative, (R)-l-((R)-2-chloro-l-
methyl-ethyl)-3-methyl-pyrrolidine (Intermediate B) is given below (Bioorganîc &
Médicinal Chemistry Letters 15 (2005) 3912-3916).
Scheme 3
Step-1 B:
[00133] A solution of (R)-2-methy! succinic acid (ig, 7.5 mmol) in 40 mL of toluene was heated at 100 °C and (R)-2-amino-propan-l-ol (0.59 ml, 7 J mmol) was added slowly to the reaction mixture. After complété addition, the reaction mixture was further heated at 130 °C for 16 hrs. After the completion of reaction, the réaction mixture was cooled to room température, concentrated under vacuum to get the crude product, which was purified by chiral préparative HPLC to get pure IB (600 mg, 60%).
Step-2B:
[00134] A solution of Intermediate IB (0.5 g. 2.9 mmol) in dry ether (10 mL) was added in drops to a cooled suspension of LAH (0.433 g, 11.6 mmol) in dry diethyl ether at 20 0nC. After completion of addition, the reaction mixture was gradually allowed to reach room température and stirred for 12 h. The reaction was monitored by TLC (20% MeOH ln DCM). After completion of reaction, the reaction mixture was cooled to 0 °C, quenched successively with water (0.5 mL), 10% NaOH (l mL) and water (1.5 mL). The precipitated solid was filtered through celite and the nitrate was concentrated under vacuum to get the product (2B) 25 which was used as such for next step (product formation confïrmed by *H NMR). Yield: 300 mg, 70%.
Step-3B:
[00135] To a solution of Intermediate 2B (0.3 g, 2 mmol) ln 1,2 dichloroethane (10 mL). thionyl chloride (0.18 mL, 2.5 mmol) was added drop wise at 0 °C. The reaction
-37mixture was grndually heated to 80 °C for 2 h. After completion of reaction, the excess solvent and thionyl chloride were removed under reduced pressure to get the crude Intermediate B, which was taken as such for next step (product formation was confirmed by LCMS). Yield: 300 mg, 88%.
[00136] Tlie following intermediates are or can be prepared following the method described for Intermediate A and Intermediate B, and using the appropriate reagents, and starting materials.
Me Me r f
Me * Me
A' B*
Représentative Intermediate Synthesis 3
Représentative synthesis of l-((2-Chloro-l-methyI)ethyl) pyrrolidine analogs
Ma [00137] The intermediate chloroethylpyrrolidine dérivatives can be prepared by following the représentative method described in Bioorganic & Médicinal Chemistry Letters (2005) 15 3912-3916.
Intermediate C [00138] Tlie synthesis of a représentative pynolidine dérivative, I -((S)-2-Chloro-Imethyl-ethyl)-pyrrolidine (Intermediate C) is given below.
-38Scheme 4
Br'
Step>1C
1,4-Dlbromo-butane (S}-2-Amlno-propan-1-ol
IC
Intermediate C
Step-lC:
[00139] To a solution of 1,4 dibromo butane (500 mg, 2.31 mmol) in acetonitrile (8 mL) was added KjCOj (0.64 g, 4.62 mmol) followed by (S)-2-amino propanol-1 (0.18 g, 2.31 mmol) in acetonitrile (2 mL) at room température. The reaction mixture was refluxed for 18 h. After completion of reaction (by TLC, 20% MeOH in DCM), the reaction mixture was allowed to reach room température and then filtered. The filtrate was concentrated under vacuum to get the crude IC (300 mg) which was lakcn for next step without any further purification (product formation confirmed by *H NMR).
Step-2C:
[00140] To a solution of lntermediate IC (300 mg, 2.3 mmol) in 1,2 dichloroethane 15 (10 mL) was added S OC h (0.2 mL, 2.7 mmol) drop wise and then slowly heated to 80 °C for
h. After completion of reaction (by TLC), excess solvent and thionyl chloride were removed under vacuum to get Intermediate C (300 mg, 87%), which was taken as such for next step (product formation was confirmed by LCMS). Yield: 300 mg, 87%.
[00141] The following intermediate is or can be prepared following the method described for Intermediate C, and using the appropriate reagents, and starting materials.
Me c
Représentative Intermediate Synthesis 4
Représentative synthesis of l-((2-Chioro-l-methyl)ethyl) pyrrolidine analogs
CI* Y
Me
[00142] The intermediate chloroethylpyrrolidine dérivatives can be prepared by following the représentative method described below.
Intermediate D [00143] The synthesis of a représentative pyrrolidine dérivative, 1 -((S)-2-Chloro-1 met hy 1 -et liyl)-pi péri dine (Intermediate D) is given below.
Scheme 5
Step-tD
Slep-2D
(S}-2-Amino- propan-1 *ol
1D
Intarmediale D
Step-1D:
[00144] To a solution of 1,5 dibromo pentane (500 mg, 2.18 mmol) in acetonitriie (8 mL), was added K2CO3 (0.9 g, 6.55 moi) followed by (S)-2-amino propanol-1 (163 mg, 2.18 mmol) in acetonitriie (2 mL) at room température. The reaction mixture was heated to reflux 20 for 18 hrs. After completion of reaction (by TLC), the reaction mixture was cooled to room température, filtered and the nitrate was concentrated to get the crude product (ID) which was taken as such for next step. Yield: 300 mg.
Step-2D:
[00145] To a solution of Intermediate ID (300 mg, 2.1 mmol) in 1,2 dichloroethane 25 (10 mL) was added SOCh (0.2 mL, 2.5 mmol) drop wise and then slowly heated to 80 °C for
h. After completion of reaction (by TLC), excess solvent and thionyl chloride were
-40removed under vacuum to get Intermediate D (300 mg, 88%), which was taken as such for next step (product formation confïrmed by LCMS).
[00146] The following intermediate is or can be prepared following the method described for Intermediate C, and using the appropriate reagents, and starting materials.
Me
Représentative Intermediate Synthesis 5 [00147] The intermediate benzopyran dérivatives can be prepared by following the 10 représentative method described below.
Intermediate E
Représentative synthesis of 2-(4-hydroxyphenyl)-benzopyranone analogs
Stcp-1:
[00148] A suspension of resorcinol (6-1) (40 g, 0.363 mol) and 4-hydroxyphenylacetic acid (49 g, 0.326 mol) in BF3.etherate (1.09 mol, 138 mL) was refluxed for 15 min under
-41argon atmosphère (the reaction mixture became clear solution). The reaction was monitored by TLC (30% pet ether in ethyl acetate was used as eluting solvent). After completion of the reaction, the reaction mixture was cooled in an ice bath and then poured into an excess of icewater. The resulting yellow precipitate obtained was collected by filtration and washed with 20 % Pet ether/ ethyl acetate followed by 25% ethanol-water to give Intermediate 6-2 as offwhite solid. Yield 45 g, 51%.
Step-2:
[00149] To asuspension of Intermediate 6-2 (15 g, 0.061 mol) in 3,4-dihydro-2Hpyran (52 g, 0.61 mol) was added p-toluenesulfonîc acid monohydrate (584 mg, 3 mmol) al 10 °C. The reaction mixture was stirred for 45 min at 0°C. The reaction was monitored by TLC (30 % pet ether in ethyl acetate as eluting solvent). After completion of reaction, the reaction mixture was treated with saturated sodium bicarbonate and diethyl ether. The organic phase was separated, washed with saturated sodium bicarbonate, brine and dried over sodium sulfate. The solvent was concentrated under reduced pressure to get crude compound, which was purified by flash column chromatography (silica gel, pet ether / ethyl acetate as solvents) to get pure Intermediate 6-3 as white solid. Yield: 17 g, 67 %.
Step-3:
[00150] A solution of Intermediate 6-3 (6 g, 0.015 mol), 4-hydroxybenzaldehyde (1.59 g, 0.013 mol) and piperidine (370 mg, 4.0 mmol) in benzene (100 mL) was refluxed using Dean-Stark apparatus for 16 h. The reaction was monitored byTLC (using 25 % ethyl acetate in Pet ether as eluting solvent). After completion of reaction, the reaction mixture was cooled to room température and the solvent was removed under reduced pressure to get crude product. The crude product was purified by flash column chromatography (silica gel, Pet ether / ethyl acetate as solvent system) to get Intermediate 6-4 (Intermediate E) as white solid. Yield: 1.6 g, 21 %.
-42SYNTIIESIS OF REPRESENTATIVE COMPOUNDS
Synthesis of QP-1038 (alkylation method)
Scheme 5
Step-4:
Intermedlate-B [00151] To the stirred solution of Intermediate-4 (0.25 g 0.48 mmol) in dry acetone (10 mL) was added césium carbonate (0.47 g, 1.4 mmol) at 0 °C and stirred for 10 min. A solution of (R)-l-(2-chloro-ethyl)-3-methyl-pyrrolidine hydrochloride (Intermedlate-B) (90 mg, 0.58 mmol) in 2 mL acetone was added at 0 °C. After completion of addition, the reaction mixture was gradually heated to reflux and maintained for 18 h. The reaction mixture was filtered, washed with acetone and concentrated to get Intermediate-5B as yellow oil. The crude product was taken as such for next step (product formation was confirmed by LCMS). Yield: 0.210 g (crude) 68%.
Scheme 6
Step-5:
[00152] To a cooled (0 °C) solution of Intermediate-5B (200 mg, 0.31 mmol) in dry THF (5 mL), CHjMgI (1.2 mL, 1.5 M solution in THF, 1.55 mmol) was added drop wise. After completion of addition, the réaction mixture was slowly allowed to reach room température and stirred for 6 h. Then reaction mixture was cooled to 0 °C, quenched with ammonium chloride solution (10 mL) and extracted with EtOAc (3 x 10 mL). The combined
-43organic layer was dried over anhydrous sodium sulphate, filtered and concentrated under vacuum to get crude intermediate 6B as yellow oil. This was taken în acetic acid (9 mL) and water (1 mL) and heated to 90 flC for 2 h. The reaction mixture was cooled to room température and concentrated under vacuum to remove the solvents. The residue was taken in
EtOAc and quenched with saturated NaHCOj solution at 0 °C. The organic layer was separated and aqueous layer extracted with EtOAc (2 x 10 mL). The combined organic layer was dried over anhydrous sodium sulphate, filtered and concentrated to get the crude product. The crude product was purified by flash column chromatography (silica gel, EtOAc / Pet ether) followed by préparative HPLC to get pure OP-1038 as beige colored solid. Yield: 25 10 mg, 18 %.
[00153] Following the general method above and using the appropriate reagents and starting materials OP-1039, OP-1042, OP-1049, OP-1050, and OP-1053 were synthesized.
Synthesis of QP-1060 and QP-1061
A—A—
I. Z—/Y»
Step-1:
[00154] A solution ofdimethoxy fluorobenzene (1) (iO g, 64 mmol) in dry dichloromethane (40 mL) was cooled to -30 °C. To the cooled solution was added a solution of BBrj (35.93 mL, 384 mmol ) in 60 mL of DCM slowly (in drops) over 30 min. After completion of addition, the reaction mixture was allowed to reach 25 °C and stirred for 12 h.
-44After completion of reaction (by TLC using 30% ethyl acetate and pet ether as eluting solvent), reaction mixture was cooled to 0 °C and then slowly quenched with water and stirred for 30 min at 25 °C. The réaction mixture was extracted with dlchloromethane (3 x 100 mL) and the combined organic layer was washed with sodium bicarbonate, dried over Na^SOi and concentrated to get the product (2) as light brown solid. Yield: 8g (97%)
Siep-2:
[00155] A mixture of Fluoro resorcinol (2) (9 g. 70 mmol ) and 4hydroxyphenylaceticacid (9.62 g, 63 mmol) in B Fj-EfiO (26.7 ml, 210 mmol ) were stirred under reflux for 15 min T. After completion of the reaction (monitored by TLC, using 30% ethyl acetate and pet ether as eluting solvent) the reaction mixture was cooled in an ice bath and poured into excess of ice-water and extracted with ethyl acetate (3 x 100 mL). The combined ethyl acetate layer was dried over anhyd Na^SO^ and concentrated under vacuum to get the crude product as mixture of regioisomers. Préparative HPLC purification afforded the require isomer (3) as beige colored solid. Yield: 4.5 g (25%).
[00156] A suspension of Intermediate 3 (6 g, 23 mmol) in 3,4-dihydro-2H-pyran (23.28 mL, 270 mmol) was treated with catalytic amount (2 drops) of conc.HCl at -10°C. The reaction mixture was stirred for 45 min at 0°C. After completion of reaction ( by TLC using 30% pet ether and ethyl acetate as eluting solvent),the reaction mixture was treated with saturated sodium bicarbonate and dicthyl ether (100 mL). The organic layer was separated.
-45dried over sodium sulfate and evaporated under reduced pressure to get the crude product as yellow oil. This was recrystallized from with hexane and diethy! ether to get compound 4 as white solid. Yield: 3.4 g (34 %).
Step-4:
[00157] A solution of bis-THP ether Intermediate 4 (5 g, 11.6 mmol ), 410 hydroxybenzaldehyde ( 1.27 g, 10.4 mmol ) and piperidîne (0.114 mL, 11.6m ) in benzene (50 mL) was stirred and refluxed using a Dean-Stark apparatus for 16 h. After cooting to room température, the solvent was removed under reduced pressure to get crude product The crude material was purified by column chromatography (silica get, pet ether / ethyl acetate) to get Intermediate 5 as a pale yellow solid. Yield: 2 g (32%).
[00158] To the stirred solution of Intermediate 5 (500 mg, 0.93 mmol) in dry acetone (15 mL), césium carbonate (91 Img, 2.8 mmol) was added at 0°C. The reaction mixture was stirred for 10 min at 0°C, and (R)-l-((S)-2-Chloro· l-methy!-ethyl)-3-methylpyrrolidinehydrochloride (3A) (185 mg, 0.93 mmol ) was added and heated to 60 eC for 19 h.
After completion of reaction (by TLC using ethyl acetate and 10% methanol as elutîng solvent), the reaction mixture was filtered, washed with acetone and concentrated to get the crude product. The crude product was purified by column chromatography (silica gel, pet
-46ether / ethyl acetate) to get pure Intermediate 6A as yellow solid. The product formation was confirmed by LCMS. Yield: 200 mg (30%)
[00159] To a cooled solution (-5 °C) of Intermediate 6A (300 mg, 0.45 mmol, 1 eq) 10 in dry THF (5 mL) was added MeMgBr ( 1.4M în THF, 3.20 mL, 4.50 mmol) slowly in drops. After completion of addition, the reaction mixture was allowed to reach 25 °C and stirred for 6 h. Aller completion of réaction (monitored by LCMS), the reaction mixture was quenched with satd ammonium chloride solution and extracted with ethylacetate (3 x 2o mL). The combined organic layer was dried over anliydrous sodium sulphate and concentrated under vacuum to get the crude Intermediate 7 as yellow oîl (the product was confirmed by LCMS). The crude Intermediate 7A was dissolved în acetic acid (9 mL) and water (1 mL) and heated to 90 cC for 3h. After completion of reaction (by TLC, using 20% MeOH and ethyl acetate as eluting solvent), the reaction mixture was cooled to 25 °C and concentrated under vacuum. The residue obtained was taken in EtOAc and quenched with saturated
NaHCOj solution at 0 °C. The organic layer was separated and aqueous layer extracted with EtOAc (2 x 10 mL). The combined organic layer was dried over anhydrous sodium sulphate, filtered and concentrated to get the crude product. The crude product was purified by flash column chromatography (silica gel, EtOAc / Pet ether) followed by préparative HPLC to get pure OP-1060 as beige colored solid. Yield: 60 mg (27%).
[00160] Following the above procedure and using the appropriate reagents and starting materials, OP-1061 was also synthesized.
-47Synthesis of QP-1056
3-(4-Hydroxyphenyl)-2-(4-(2-((R)-3-methylpyrrolidin-l-yl)ethoxy)phenyl)-4- (trifluoromethyl)-2H-chromen-7-ol
Scheme 16
2- (4-(2-((R)-3-Methylpyrrolidin-l-yi)ethoxy)phenyl)-7-((tetrahydro-2H-pyran-2-yl)oxy)-
3- (4-((tetrahydro-2H-pyran-2-yl)oxy)phenyi)chroman-4-one (8*) [00161] To a solution of 6* (1.0 g, 4.3 mmol), resorcinoi dérivative T (2.1 g, 5.1 mmol) and piperdine (0.17 mL, 1.7 mmol) in toluene (150 mL) is heated at reflux with azeotæpic removal of water for 24 hours. The solution is concentrated and the residue is purified by silica ge! column chromatography using a gradient of 0 to 8% methanoi in dichloromethane to yield 8’ as a brown viscous oii (1.4 g, 52% yield).
MS Calculated C3BH45NO7 + H+ = 628; Observed 628.
3-(4-Hydroxyphenyl)-2-(4-(2-((R)-3-methyipyrro!ldIn-l-yl)ethoxy)phenyl)-4(trifluoromethyl)chroman-4,7-diol (9’) [00162] To a solution of 8* (1.0 g, 1.6 mmol), trimethyl(trifluoromethyl)silanc (1.1 mL, 7.5 mmol) is added césium fluoride (0.045 g, 0.29 mmol) and the solution is stirred at room température for 48 hours. The reaction mixture is concentrated and the residue is dissolved in ethyl acetate (100 mL) and washed with brine, dried over anhydrous sodium sulfate, fïitered, and concentrated to yield a brown residue that is purified by silica gei column chromatography with using a gradient of 0 to 8% methanoi in dichloromethane to yield 9’ as a yellow solid (0.5 g, 59% yield).
-48MS Calculated C29H30F3NO5 + H* = 530. Observed 530.
3-(4-Hydroxyphenyl)-2-(4-(2-((R)-3-methylpyrrolidin-l-yl)ethoxy)phenyl)-4(trinuoromethyl)-2H-chromen-7-ol (10*) (OP-1056) [00163] To a solution of 9’ (0.5 g, 0.94 mmol) in tetrahydrofuran (2 mL) is added triethylamine (6.6 mmol, 0.91 mL) and trifluoroacetic anhydride (2.8 mmol, 039 mL) simultaneously at 0 °C and the solution is allowed to warm to room température with stirring for 16 hours. The solution is concentrated and the residue is dissolved in ethyl acetate (100 mL) and washed with brine, dried over anhydrous sodium sulfate, filtered, and concentrated to yield a brown residue that is purified by silica gel column chromatography with using a gradient of 0 to 10% méthanol in dichloromethane to yield a yellow solid. This solid was further purified by préparative HPLC to yield, after lyophyl izatîon (0.024 g, 4.9% yield). MS Calculated CioHkFjNOî + H* = 512. Observed 512.
[00164] The synthèses of représentative compounds of this invention can be carried out in accordance with the représentative methods set forth below and using the appropriate reagents, starting materials, and purification methods known to those skilled in the art.
-49Synthesisor3-(4-hydroxyphenyi)-4-methyI-2-(4-{2-[(3R)-3-methylpyrrolidin*l· yl]ethoxy)phenyl)-2H-chromen-7-ol (OP-1038) and Séparation and Purification of Stereoisomers (OP-1074 and OP-1075)
FF .yxr
Reaction to produce l*(2,4-Dihydroxyphenyl)-2*(4-hydroxyphenyl)ethanone [00165] Rcsorcinol (1,3-dihydroxybenzene) (62.000 g, 563.1 mmol, 1.0 equiv.) and 4Hydroxyphenylacetic acid (94.237 g, 619.4 mmol. 1.1 equiv.) were added to a 3 neck 2 L round bottomcd flask fitted with a paddle, a pressure cqualizing addition funnel and a thermometer and a heating mantle. Toluene (350 mL) was added to the flask to give a suspension. The réaction purged with nitrogen and the addition funnel fil led 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 internai température rose to 100 °C. The reaction went through various changes in color from yellow to dark red. After complété addition of boron trifluoride etherate the addition funnel was removed and replaced with a condenser. The reaction was stirred for 1.5 h at on internai température of 108 °C. A sample was taken and
-50HPLC analysis indicated the reaction was complété. 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 5 on a vacuum oven for 16 h to give the product as a white powder ( 119.67 g, 87.0 %).
Slep 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 [00166] 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 mi, 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 température rose from 20 to 33 °C over 5 minutes. The yellow suspension became a red solution within I minute of PTS A addition.
The reaction was stirred for 66 h at room température. The reaction was monitored by HPLC at 4,5 and 6 hours. The chromatograms indicated the reaction was 74 %, 90 % and 100 % complété 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 1PA (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 an orange oil (38.09 g).
Step 3:
Reaction to produce 2-(44odophenyl)-7*((tetrahydro-2H-pyran-2-yl)oxy)-3-(4· ((tetrahydro-2II'pyran-2-yl)oxy)phenyl)chroman-4-one [00167] l-(2-hydroxy-4-((tetrahydro-2H-pyran-2-yl)oxy)phenyl)-2-(4-((tetrahydro-2Hpyran-2-yl)oxy)phenyl)ethanone (16.228 g, 39.34 mmol) was added to a 3 neck I L RB flask.
-512-Butanol (380 mL, 0.197 M) and 4-iodobenzaldehyde (51.700 g, 222.8 mmol, LO equiv.) was added to the flask to give a suspension. Pîperidine (7300 ml, 73.9 mmol, 0.3 equiv.) and l,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, 5 a stirrer shaft and heated in an oil bath at 130 °C to give an orange solution (became a solution when the internai température 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 redîssolved 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 al 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 température. 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 sïntered 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 % IPA/water ( 1.2 L) and the solution allowed to cool to room température and stand ovemight 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 %).
-52Step 4a:
Reaction to produce 2-(4-Iodophenyl)-4-methyl-7-((tetrahydro-2H«pyran-2-yl)oxy)-3-(4· ((tetrahyd ro-2 H-pyran-2-y l)oxy)pheny I) chroman-4-ol [00168] To a solution of 90.0% 2-(4-îodophenyl)-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 Méthylmagnésium chloride 3.0 M solution in THF (160.000 ml, 480.0 mmol, 3.2 equiv.) by addition funnel over 30 minutes. The température did not rise about 8 °C during the addition. The reaction was removed from the ice bath and stirred at room température and stirred for another hour. TLC (20% ethyl acetate in hexanes) showed the réaction 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 réaction 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 NaiSOj, 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-oI [00169] 2-(4-iodophcnyl)-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% acetîc acid in HjO (686 mL) was added to a 2 L RB flask. The suspension was degassed, fiushed 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 présent. 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 I 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
-53pink 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 solidîfy. The oil was not combined with previous batches but reprotected in a separate reaction.
[00170] 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:
Réaction to produce 2-(4-lodophenyl)-4-methyl-7-((tetraliydro-2H-pyran-2-yl)oxy)-3-(4((tetrahydro-2II-pyran-2-yl)oxy)phenyl)-21I-chromene [00171] To a solution of 3-(4-hydroxyphenyl)-2-(4-iodophenyl)-4-methyl-2Hchromen-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 température ovemight (17 h). TLC showed major desired product. The reaction was diluted with DCM (200 mL), washed with saturated NaHCO3 (200 mL), water (200 mL), brine (200 mL), dried over Na2SO4, filtered and concentrated to give a red viscous residue. The residue adsorbed onto silica gel (75 g) was purifîed 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 oC for 16 h to afford the titled compound as a white powder (51.67 g 90.2 %).
[00172] 1H NMR (300 MHz, CDCI3 ): 6 7.53 (d, J - 5.4 Hz, 2H)), 7.18 (d, J = 8.7 Hz, 1H), 7.06 (aprent t, J = 7.8 Hz, 4H), 6.71 (s, IH), 6.59 (d, J = 2.4 Hz, 1H), 6.45 (d, J = 2.4 Hz, IH), 5.15 (s, 2H), 4.59 (s, 2H), 4.63 (d, J = 5.7 Hz, 2H), 3.98 (s, 3H), 3.84 (s, 3H).
-54Step 6:
Reaction to produce -(3R)-3-methyl-I-(2-(4-(4-methyl-7-((tetrahydro-2II-pyran-2yl)oxy)-3-(4-((tetrahydro-2H-pyran-2-yl)oxy)phenyl)-2II-chromen-2yl)phenoxy)ethy!)pyrrolidine [00173] A mixture of 2-(4-iodophenyl)-4-methyl-7-((tetrahydro-2H-pyran-2-yl)oxy)-3(4-((tetrahydro-2H-pyran-2-yl)oxy)phenyI)-2H-chromene (16.800 g, 26.9 mmol, 1.0 equiv.), (R)—2-(3-methy!pyrrolidin-l-y!)ethanol (10.416 g, 80.6 mmol, 3.0 equiv.), 1,10Phenanthroline (0.970 g, 5.4 mmol, 0.2 equiv.), and Césium 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 hcating the reaction was cooled to room température and the mixture filtered through a pad of Cclitc (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 ge! (25 g) purified with silica ge! ( 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 %).
[00174] Gradient method 0 % MeOH 4 minutes, gradient to 1 % MeOH/DCM over 3 minutes, hold at I % 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 zéro minutes, hold at 25 % MeOH/DCM for i5 minutes. Many fractions contained a mixture of the starting material and product ail 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 I % MeOH/DCM over 3 minutes, hold at I % 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 %) (Laie eluting fractions from the first column contained a spot which corresponded to the amînoalcohol. These fractions were pooled and concentrated to give a red black liquid (6.38 g).
Step 7.·
Reaction to produce 3-(4-hydroxyphenyl)-4-inethyl*2-(4-{2-{(3R)-3-inethylpyrrolidin*lyl]ethoxy]phenyl)-2H*chromen*7-ol (OP-1038) [00175] (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-y!)phenoxy)ethyl)pyrro!idine ( 15.130 g, 24.2 mmol, 1.0 equiv.) was dissolvcd into 80% acetic acîd/water (150 mL). The solution was heated in an oil bath at 90 ’C for 1 hour. HPLC analysis of the réaction mixture indicated the reaction was complété. The dark red solution was concentrated to a dark red oil.
The oil was suspended into ethyl acetate (600 mL) and washed with saturated NaHCO3 (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 magnésium sulfate, filtered and concentrated to give a red oil (14.03 g, crude). The oil was adsorbcd onto silica gel (30 g) and chromatgraphed on silica gel (2 x 100 g cartridge) with ΟΙ 5 10 % MeOH în 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 %.
[00176] 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]+ e 458.3.
Step 8:
[00177] OP-1038 was separated into its diastereomers (2S)-3-(4-hydroxyphenyl)-4methyl-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-o! (OP-1075) using a Diacel, Chiralpak IC column at room 30 température 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 préparative scale.
-56Step 9:
Reaction to produce (R)-2-(benzyloxy)-l-(3-methylpyrrolldin-l-yl)ethanone [00178] (RJ-3-methylpyn-olidine 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 distîlled Diisopropylethylamine (60.157 ml, 345.4 mmol, 2.1 equiv.) and freshly activated 4 A molecular sïeves (— 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 température dropwise via syringe over 20 minutes with a room température water bath for cooling. After complété 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 pourcd into a separatory funnel and the organic layer washed successively with 1 M HCI (2 x 200 mL), saturated sodium bicarbonate (200 mL) and brine (200 mL). The organic layer was dried over anhydrous MgSO4, filtered and concentrated to an orange oil (42.40 g). The oil was loaded onto silica gel (30 g) and the mixture split into — 18g 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 oïl (34.02 g, 88.7 %).
[00179] 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 [00180] 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)-1-(3methylpyrrolidin-l-yl)ethanone (33.980 g, 136.3 mmol, 1.0equiv.) in anhydrous THF(150 mL) was added dropwise to the cold suspension via a pressure equalizing addition funnel
-57over 20 minutes. The reaction was kept at -78 °C for 1 hour and stirred at room température for 1 hours. The reaction was carefully quenchcd with 6 N HCl 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 se para tory funnel and the layers separatcd (-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 hexancs followed by 10-40 % methanol in dichloromethane to give a yellow oil (29.17 g, quant).
[00181] 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:
[00182] (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 % H2O (6.403 g, 45.6 mmol, 1.0 equiv.) was added to the cooled solution and flushcd with nitrogen. Hydrochloric acid (6 M, 7.6 mL) was added to mixture. The flask was pressurized with hydrogen to 30 psi shaken for I 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 incomplète 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 ccntrifuged 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 %).
[00183] Following the general method above and using the appropriate reagents and starting materials, OP-1046 and OP-1047 were synthesized.
Synthesis of HCl Salts of QP-1038 and QP-1074 [00184] 3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3-methylpynolidin-lyl]ethoxy)phenyl)-2H-chromen-7-ol (OP-1038; 0.020 g, 0.0 mmol, 1.0 equiv.) or (2S)-3-(4hydroxyphenyl)-4-methyl-2-(4-(2-[(3R)-3-methylpyrrolidin-l-yl]ethoxy|phenyl)-2Hchromen-7-ol (OP-1074; 0.020 g, 0.0 mmol, 1.0 equiv.) (Compound 33) was placed into a I dram vial and dissolved into methanol (0.2 mL). 4 M HCl in methanol (200 pL) 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 QP-1083 [00185] OP-1083 was prepared by air oxidation on OP-1074, followed by chromatographie séparation of OP-1083 and OP-1074. The mixture of OP-1074 and OP-1083 (560 mg) was dissolved in methanol (15 mL) andmixed 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 dichloromethane to give OP-1074 as an orange solid (0.261 g, 46.6 %) and OP-1083 as an orange solid (41.1 mg. 15%) [00186] Method: 0 % MeOH for 4 min, gradient to 5 % McOH/DCM over 5 minutes, hold at 5 % for 6 minutes, gradient to 10 % McOH/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 30 minutes.
-59Synthesis of OP-1084 [00187] (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-(2-[(3R)-3-methylpyrrolidin-lyl]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). Diisopropylcthylamine ( 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 light syringe. The réaction immedîately became a cloudy white suspension. The reaction was removed from the ice bath and stirred at room température 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 %).
[00188] 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 QP-1085 [00189] (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-{2-[(3R)-3-methylpynOlidin-lyl]ethoxy)phenyl)-2H-chromen-7-ol (0.035 g, 0.1 mmol, 1.0 equiv.) was added to a dry I 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, i 13.6 equiv.) was added to the vial via syringe to give pink red solution. The vial was cooled in a 0 °C icc/water bath for 10 minutes andTrimethylacetyl Chloride (100 ul, 0.8 mmol, 10.6 equiv.) was added viaa GC syringe in one portion to the solution. The solution immedîately became a light yellow color and was stirred for 30 minutes at 0 °C. The reaction was allowed to reach room température over 30 min and stirred at room température for i 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 %).
·60· [00190] 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, 5 626; HPLC (254 nm): 98.0 %.
Synthesis οΓΟΡ-1086 and QP-1088 [00191] Pyridine (2000 ul, 24.8 mmol, 113.6 equiv.) was added to a dry I dram vial with a stir bar. The vial was cooled in a -15 °C dry icc methanol/water bath. Phosphores 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-methyl2-(4-{2-[(3R)-3-methylpyrro1idin-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)-315 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 température for 1.5 h. The solution became slurry after -45 minutes. The reaction was allowed to reach room température over 45 min and stirred at room température 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
HCl (5 mL). The suspension was sonicated and centrifuged at 5000 rpm for 6 minutes. The supematant was decanted and the solid resuspended into 5 mL of 2 N HCl and the process was repeated. The solid was dried under vacuum to give 129 mgof the crude product. The solid was suspended into water (2 mL) and 6 N sodium hydroxide solution (174 pL, l mmol,
5 equiv) was added to give an orange solution. This was purified by Préparative LC with acetonitrile and water to give the product as a tan solid.
Synthesis of QP-1087 [00192] (2S)-3-(4-hydroxypheny!)-4-methyl-2-(4-{2-[(3R)-3-methylpyrrolidin-lyl]ethoxy]phenyl)-2H-chromen-7-ol (0.020 g, 0.0 mmol, 1.0 equiv.) was added to a30 mL via! and suspended into anhydrous ethyl acetate (20 mL). Diisopropylethy lamine (19 ul, 0.1 mmol, 2.5 equiv.) was added to the suspension and the solution was cooled in an ice bath for minutes. Methyl chloroformate (7 ul, 0. J mmol, 2.2 equiv.) was added to the reaction via a
-61gas light syringe. The reaction immediately became a cloudy white suspension. The reaction was removed from the ice bath and stirred at room température for t6 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 %).
[00193] TLC (5 % MeOH/DCM): 4 spots, 0.95,0.55,0.38 (Product), 0.25 (Mono carbonate).
[00194] 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 % 10 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 %.
Exemplary Compounds of the Invention i5 [00195] Figures lA and IB depicts the structureof compounds, with stereochemistry as defined herein, which hâve been or can be prepared according to the synthetic methods described herein. Figure ! A also shows the structure of représentative reference compounds used for comparison, including Aragon 28 (Example 28 of WO2011/156518); Gauthier iA 20 (Gauthier et al. Synthesis and structure-activity rclatlonships of analogs of EM652(acolbifene), a pure sélective estrogen receptor modulator. Study of nitrogen substitution” Journal of Enzyme Inhibition and Médicinal Chemistry, 2005; 20(2): 165—177); EM-343, EM-651 andEM-652 (Acolbifene).Figure IB depicts représentative prodrugsandsaltsof OP-1038and OP-1074.
ASSAYS [00196] Compounds provided herein can be evaluated using various in vitro and in vivo assays; exemples of which are described below.
[00197] The following biological examples are offered to ilJustrate the compounds, pharmaceutical compositions and methods provided herein and are not to be construed in any way as limiting the scope thereof.
-62Démonstration of the Superiority of OP-1038 and OP-1074 Using
Sensitive In vitro Estrogenicity Assays [00198] Method for performing the alkaline phosphatase (AP) assay. ECC-i cells (American Type Culture Collection, Manassus, VA) were maintained in RPMI medium plus 10% fêtai bovine sérum at 37'C. At the beginning of the assay trypsinized cells were resuspended in RPMI medium plus 5% charcoal dextran stripped sérum (CDSS, (Hyclone, Logan, UT)) and plated at a density of 25-50k cells per well Into a 96-weli plate for at least 6 hours. Compounds were diluted in sérum-free medium and added 1:1 to plated cells in replicate wells (2.5% CDSS final). Plates were incubated for 3 days at 37*C and subsequently frozen at -80*C to lyse cells after removing the medium. Thawed plates were incubated with a chromogenic substrate of AP, p-nitrophenyl phosphate (Invitrogen, Grand island, NY), for 40 minutes at 40'C. Absorbance was read al 405nm using a plate spectophotometer. This assay is shown to correlate with lhe in vivo studies comparing uterine wet weight in ovariectomized rats following treatment with a number of anti-estrogens as shown in Figure 2. This AP assay is used herein for a number of studies as described in Figures 3 through 8. Specifically, Figure 3 demonstrates that OP-1038 and OP-1074 lack estrogenic activity in the alkaline phosphatase assay in ECC1 cells as compared to a number of other anti-estrogens. Figure 4 demonstrates that OP-1038 is less estrogenic than the Aragon Compound 28 at a number of doses tested in the AP assay. Figure 5 demonstrates that OP-1038 lacks estrogenic activity in the AP assay, in conlrast to other mono-methyl substituted pyrrolidines. Figure 6A demonstrates that OP-1038 and OP-1074 inhibit estrogen-stimulated AP in ECC-I cells. Figures 6B and 6C demonstrates that OP-1038 is more potent than Aragon Cpd. 28 in inhibitionof E2-stimulated APactivity in ECC-i cells. Figure7demonstrates that OP-1038 is more potent than EM-652 in lhe AP assay in ECC-l cells. In vivo activity comparing uterine wet weight in ovariectomized rats was then confirmed for OP-1038 and OP-1074 as shown in Figure 9.
[00199] Compounds of invention are tested for their inhibitory activity of estrogen according to the assay methods described in Hodges-Gallagher, L., Valentine, C.V., El Badcr,
S. and Kushner, P.J. (2007) “Histone Dcacetylase Inhibitors Enhance the Effïcacy of Hormonal Thcrapy Agents on Breast Cancer Cells and Blocks Anti-estrogen-Driven Uterine Cell Prolifération Breast Cancer Res Treat, Nov; 105(3):297-309. Specifically, an estrogenresponsive reporter gene (ERE-tklO9-Luc) was transiently transfected into MCF-7 cells and treated with anti-estrogens in triplicate in the presence of 100 pM I7p-estradiol (E2) for 1816917
-6322 hours. Luciferase activity was normalized to activity of E2 alone and IC50’s were calculated using the least squares fit method. This method is also described in legend for Figure 10. OP-1038 and OP-1074 are potent antagoniste of estrogen-stimulated EREregulated reporter gene activity. OP-1038 and OP-1074 hâve improved potency for inhibition 5 of E2 induced transcription compared to tamoxifen, EM-343 and raloxïfene.
[00200] Prolifération in MCF-7 was measured using a fluorescent DNA binding dye 6- days after treatment in triplicatc with anti-estrogens in the presence of 100 pM E2 and as described in the legend to, and depicted in Figure 11. OP-1038 and OP-1074 are potent antagoniste of estrogen in MCF-7 cells. OP-1038 and OP-1074 hâve improved potency for inhibition of E2 stimulated prolifération over tamoxifen, EM-343, and raloxïfene.
[00201] ERa expression was detected in MCF-7 cell lysâtes treated with 100 nM anti* eslrogens in serum-free medium for 22-24 hours and immunoblotted with an antibody spécifie to ERa as described in the legend to, and depicted >n Figure 12. OP-1074 and OP1038 induce dégradation of estrogen receptor-alpha in human breast and endométrial cells in 15 a manner comparable to fulvestrant.
[00202] OP-1038 and OP-1074 inhibit E2 induced transcription, E2 stimulated prolifération and they induce dégradation of the estrogen receptor-alpha in a manner comparable to Aragon Compound 28.
Mammary Tumor Xenografï Study [00203] The purpose of this study is to examine the abîlity of OP-1074, to slow or shrink a tamoxifen résistant tumor (MCF-7 HER2/neu Clone 18) xenograft growing on ovariectomized athymie nude mice under stimulation from exogenaus estrogen. Clone 18 cells grown in culture are implanted along with 0.18 mg estradiol/ 90 day release pellets (lnnovative Research, Sarasota Florida) into 55 mice to ïnîtïate the experiment. When the tumors hâve reached 250 cubic mïllimeters the mice are divided into four groups of 10 mice each and dosing initiated. The four groups are:
[00204] I ) No hormonal treatment- this group receîves daily gavage with vehicle.
[00205] 2) Tamoxifen citrate lOOmg/kg daily by oral gavage in vehicle.
[00206] 3) Faslodex lOOmg/kg delivered daily by subcutaneous injection.
[00207] 4) OP-1074 lOOmg/kg twice daily by oral gavage in vehicle with the exception of two weekends in which dosing was once daily.
-64[00208] Results are shown in Figures 13A, 13B and 13C. OP-1074 induces rapid and complété régression of MCF-7 Clone 18 HER2/neu xenografts growing on nude mice stîmulated by estrogen.
Single Dose Oral Pharmacoklnetics Study ln Femaie Rats [00209] The oral bioavailabiIty in rats of OP- i038 was determined in the following study. 3 rats (femaie Sprague Dawlcy, non-fasted) were dosed by oral gavage (5 mg/kg body weight) in 0.5% CMC in water with a comparison to intravenous dosing (3 mg/kg body
I0 weight). Plasma was collected at the following hourly time points from rats in both groups (0, 0.08, I.0,2.0,4.0, 8.0,I6.0,24.0,48.0 and 96.0 hours post dosing). Plasma concentrations of OP-1038 were determined by HPLC. The results are shown in Table 2 below. OP-I038 is shown to be orally bioavailablc.
I5 Table 2: PK data for OP-1038
Single Dose Oral Pharmacoklnetics Study in Femaie S.D.Rats
Parameters OP-1038
Route of administration Oral
Dose (mg/kg) 5.00
CmM (ng/mL) 24.8 ± 3.00
Tma* (h) 2.33 ±1.53
AUCiast (h*ng/mL) (0 to 96 h) 603 ± 46.7
AUCfnf (h*ng/mL) 671 ±44.8
AUCextrap (%) 10.1 ±6.26
Ti/2 (h) 28.5 ±11.6
MRT|asi 25.9 ±3.37
VM(L/kg) -
CL (mL/min/kg) -
F% (Oral bioavailability) 18.9%
-65DEFTNITIONS [00210] 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 géométrie 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 synthèses. See, for example, Jacques et al., Enantiomers. Racemates and Résolutions (Wiley Interscience, New York, 1981); Wilen et al., Tetrahedron 33:2725 ( 1977); Eliel, Stereochemistry of Carbon Compounds (McGrawHill, NY, 1962); and Wilen, Tables of Resolving Agents and Optical Résolutions p. 268 (E.L. Eliel, Ed., Unîv. 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.
[00211] When a range of values is listed, it îs intended to encompass each value and sub-range within the range. For example Ci_6 alkyl is intended to encompass, Cj, Cj, Cj, Q, Cj, Ce, Cj-6, C|_5, Cj_4, Cj-3, C|-2, Cj_6, Cî-s, Cj_4, C2-3, Cm, C3-3, Cj_i, C|_6, C+-s. and 20 C5-6 alkyl.
[00212] The articles a” and “an” may be used herein to refer to one or to more than one (Le. at least one) of the grammatical objects of the article. By way of example “an analogue” means one analogue or more than one analogue.
[00213] Alkyl refers to a radical of a straight-chain or branched saturated hydrocarbon group which in one embodiment has from 1 to 20 carbon atoms (“Cj-w alkyl”). In some embodiments. an alkyl group has 1 to 12 carbon atoms (Ci-n alkyl). In some embodiments, an alkyl group has I to 10 carbon atoms (”C|_io alkyl). In some embodiments, an alkyl group has 1 to 9 carbon atoms (“C|-$ alkyl”). In some embodiments, an alkyl group has 1 to 8 carbon atoms (Cj-s alkyl”). In some embodiments, an alkyl group has 1 to 7 carbon atoms (“Ci-7 alkyl). In some embodiments, an alkyl group has 1 to 6 carbon atoms (C|_6 alkyl, also referred to herein as “lower alkyl). In some embodiments, an alkyl group has 1 to 5 carbon atoms (Cj-s alkyl). In some embodiments, an alkyl group has 1 to 4 carbon atoms (“Cj_i alkyl). In some embodiments, an alkyl group has I to 3 carbon atoms (“C1.3 alkyl). In some embodiments, an alkyl group has I to 2 carbon atoms (“C1-2 alkyl).
-66In some embodiments, an alkyl group has I carbon atom (“Ci alkyl). In some embodiments, an alkyl group has 2 to 6 carbon atoms (“Cj-e alkyl”). Examples of C|_e alkyl groups include methyl (C|), ethyl (C2), n-propyl (C3), isopropyl (C3), n-butyl (C4), tert-butyl (Cg). sec-butyl (Cg), iso-butyl (C4), n-pentyl (C3), 3-pentanyl (C3), amyl (C3), neopentyl (C3), 3-methyl-25 butanyl (C3), tertiary amyl (C3), and π-hexyl (Q). Additional examples of alkyl groups include n-heptyl (C7), π-octyl (Ce) and the like. Unless otherwise specified, each instance of an alkyl group is independently optionally substituted, i.e., unsubstituted (an unsubstituted alkyl”) or substituted (a “substituted alkyl”) with one or more substituents; e.g., for instance from 1 to 5 substituents, 1 to 3 substituents. or I substituent. In certain embodiments, the alkyl group is unsubstituted Cj_to alkyl (e.g., -CH3). In certain embodiments, the alkyl group is substituted Cmq alkyl.
[00214] “Alkylene” refers to a substituted or unsubstituted alkyl group, as defined above, wherein two hydrogens are removed to provide a divalent radical. Exemplary divalent alkylene groups include, but are not limited to, methylene (-CH2·). ethylene (-CH2CH2-), the 15 propylene isomers (e.g., -CH2CH2CH2' and -CH(CH3)CH2·) and the like.
[00215] “Alkenyl refers to a radical of a straight-chain or branched hydrocarbon group which in one embodiment has from 2 to 20 carbon atoms, one or more carbon-carbon double bonds, and no triple bonds (Cï-m alkenyl”). In some embodiments, an alkenyl group has 2 to 10 carbon atoms (C^-to alkenyl”). In some embodiments, an alkenyl group has 2 to
9 carbon atoms (C2-9 alkenyl”). In some embodiments, an alkenyl group has 2 to 8 carbon atoms (C2-8 alkenyl). In some embodiments, an alkenyl group has 2 to 7 carbon atoms (“C2-7 alkenyl”). In some embodiments, an alkenyl group has 2 to 6 carbon atoms (“C2-6 alkenyl”). In some embodiments, an alkenyl group has 2 to 5 carbon atoms (”C2_3 alkenyl”). In some embodiments, an alkenyl group has 2 to 4 carbon atoms (”C2_g alkenyl”). In some embodiments, an alkenyl group has 2 to 3 carbon atoms (“C2-3 alkenyl”). In some embodiments, an alkenyl group has 2 carbon atoms (“C? alkenyl”). The one or more carboncarbon double bonds can be internai (such as in 2-butenyl) or terminal (such as in 1-butenyl). Examples of C2-1 alkenyl groups include ethenyl (C2), 1-propenyl (C3), 2-propenyl (C3), 1butenyl (C4), 2-butenyl (C4), butadienyl (C4), and the like. Examples of Cj-e alkenyl groups include the aforementioned C2-1 alkenyl groups as well as pentcnyl (C3), pcntadienyl (C3), hexenyl (Ce), and the like. Additional examples of alkenyl include heplenyl (C7), oclenyl (Cg), octatrienyl (Cg), and the like. Unless otherwise specified, each instance of an alkenyl group is independently optionally substituted, i.e., unsubstituted (an “unsubstituted alkenyl”) or substituted (a “substituted alkenyl”) with one or more substituents e.g.. for instance from I
-67to 5 substituents, l to 3 substituents, or t substituent In certain embodiments, the alkenyl group îs unsubstituted C2-10 alkenyl. In certain embodiments, the alkenyl group is substituted Cî-iq alkenyl.
[00216] “Alkenylene refers a substituted or unsubstituted alkenyl group, as defined above, wherein two hydrogens are removed to provide a divalent radical. Exemplary divalent alkenylene groups include, but are not Iimited to, ethenylene (-CH=CH-), propenylenes (e.g., -CH=CHCHî- and -C(CHj)=CH- and -CH=C(CH3}-) and the like.
[00217] “Alkynyl refers to a radical of a straight-chain or branched hydrocarbon group which in one embodiment has from 2 to 20 carbon atoms, one or more carbon-carbon 10 triple bonds, and optionally one or more double bonds (C2-20 alkynyl’’). In some embodiments, an alkynyl group has 210 10 carbon atoms (C2-10 alkynyl). In some embodiments, an alkynyl group has 2 10 9 carbon atoms (C2-9 alkynyl). In some embodiments, an alkynyl group has 2 to 8 carbon atoms (“C2-8 alkynyl). In some embodiments, an alkynyl group has 2 to 7 carbon atoms (C2-7 alkynyl). In some 15 embodiments, an alkynyl group has 2 to 6 carbon atoms (“C2-6 alkynyl). In some embodiments, an alkynyl group has 2 to 5 carbon atoms (C2-J alkynyl). In some embodiments. an alkynyl group has 2 to 4 carbon atoms (C2-1 alkynyl”). In some embodiments, an alkynyl group has 2 to 3 carbon atoms (C2-3 alkynyl”). In some embodiments, an alkynyl group has 2 carbon atoms (“Ci alkynyl’’). The one or more carbon20 carbon triple bonds can be internai (such as in 2-butynyl) or terminal (such as in l-butynyl).
Examples of C2-4 alkynyl groups include, without limitation, ethynyl (C2), l-propynyl (Cj), 2-propynyl (C3), I-butynyl (G), 2-butynyl (Q), and the like. Examples of C2-6 alkenyl groups include the aforementioned C2-i alkynyl groups as well as pentynyl (Q), hexynyl (G), and the like. Additional examples of alkynyl include heptynyl (C?). octynyl (G), and 25 the like. Unless otherwise specified, each instance of an alkynyl group is independently optionally substituted, i.e„ unsubstituted (an “unsubstituted alkynyl) or substituted (a “substituted alkynyl) with one or more substituents; e.g„ for instance from I to 5 substituents, 1 to 3 substituents, or 1 substituent In certain embodiments, the alkynyl group is unsubstituted C2-10 alkynyl. In certain embodiments, the alkynyl group is substituted C2.|q 30 alkynyl.
[00218] “Alkynylene refers a substituted or unsubstituted alkynyl group, as defined above, wherein two hydrogens are removed to provide a divalent radical. Exemplary divalent ulkynylene groups include, but are not Iimited lo, ethynylene, propynylene, and the like.
-68[00219] Naturel ly occurring or non-naturally occurring amino acids” can be used in the préparation of compounds of the invention as described herein. For example, naturel amino acids include valine, leucine, isoleucine, méthionine, phenylalanine, asparagine, glutamic acid, glutamine, histidine, lysine, arginine, aspartîc acid, glycine, alanine, serine, threonine, tyrosine, tryptophan, cysteine, proline, 4-hydrox y proline, g-carboxyglutamic acid, selenocysteine, desmosine, 6-N-methyllysine, e-N.NJ^-trïmethyllysîne, 3-methylhistidine, Ophosphoserine, 5-hydroxy!ysine, e-N-acety!lysine, s-N-methylarginine, N-acetylserine, gaminobutyric acid, citrul line, omithinc, azaserine, homocystéine, b-cyanoalanine and Sadenosylmethionine. Non-limiting examples of non-naturally occurring amino acids include phenyl glycine, meta-tyrosine, para-amino phenylalanine, 3-(3-pyridyl)-L-alanine, 4(trifluoromethyl)-D-phenylalanine, and the like. In one embodiment, an L-amino acid is used. [00220] ‘Aryl refers to a radical of a monocyclic or polycyclic (e.g., bicyclic or tricyclic) 4n+2 aromatic ring system (e.g., having 6, 10, or 14 π électrons shared in a cyclic array) having 6-14 ring carbon atoms and zéro heteroatoms provided in the aromatic ring
System (“C^u aryl”). In some embodiments, an aryl group has six ring carbon atoms (“Cû aryl”; e.g., phenyl). In some embodiments, an aryl group has ten ring carbon atoms (“Cio aryl”; e.g., naphthyl such as l-naphthyl and 2-naphthyl). In some embodiments, an aryl group has fourteen ring carbon atoms (“Ci4 aryl”; e.g., anthracyl). Aryl also includes ring Systems wherein the aryl ring, as defined above, is fused with one or more carbocyclyl or heterocyclyl groups wherein the radical or point of attachment is on the aryl ring, and in such instances, the number of carbon atoms continue to designate the number of carbon atoms in the aryl ring system. Typical aryl groups include, but are not limitée! to, groups derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, coronene. fiuoranthene, iluorenc, hexacene, hexaphene, hexalene, as-indacene, s-indacene, indane, indene, naphthalene, octacene, octaphene, octalenc, ovalene, penta-2,4-diene, pentacene, pentalcne, pentaphcnc, perylene, phenalene, phcnanthrenc, picene, pleiadene, pyrene, pyranthrene, rubicene, triphenylene, and trinaphthalene. Particularly aryl groups include phenyl, naphthyl, indenyl, and tetrahydronaphthyl. Unless otherwise specified, each instance of an aryl group is independently optionally substituted, i.e., unsubstïtuted (an unsubstïtuted aryl”) or substituted (a substituted aryl) with one or more substituents. In certain embodiments, the aryl group is unsubstïtuted Cfr-u aryl. In certain embodiments, the aryl group is substituted Ce-u uryl.
[00221] ln certain embodiments, an aryl group substituted with one or more of groups selected from halo, Ci-Cg alkyl, C|-Ce haloalkyl, cyano, hydroxy, Ci-Cb alkoxy, and amino.
-69[00222] Examples of représentative substituted aryls include the following:
In these Formula one of R36 and R37 may be hydrogen and at least one of R36 and R37 is each independently selected from C|-Cg alkyl, C(-Cg haloalkyl, 4-10 membered heterocyclyl, alkanoyl, Ci-C8 alkoxy, heteroaryloxy, alkylamino, arylamino, heteroarylamino, NR38COR39, NR38SOR39 NR38SO2R39, COOalkyl, COOaryl, CONR38R39, CONR38OR39, NRÎ8R3’, SO2NR38R39, S-alkyl, SOalkyl, SO2alkyl, Saryl, SOaryl, SO2aryl; or Rse and R37 may be joined to form a cyclic ring (saturated or unsaturated) from 5 to 8 atoms, optionaily containing one or more heteroatoms selected from the group N, O, or S. R60 and R61 are independently hydrogen, C|-Cg alkyl, C1-C4 haloalkyl, Cj-Cto cycloalkyl, 4-10 membered heterocyclyl, Ce-Cio aryl, substituted Cé-C(o aryl, 5-10 membered heteroaryl, or substituted 510 membered heteroaryl.
[00223] “Fused aryl” rcfers to an aryl having two of its ring carbon in common with a second aryl ring or with an aliphatîc ring.
[00224] “Aralkyl is a subset of alkyl and aryl. as defined herein, and refers to an optionaily substituted alkyl group substituted by an optionaily substituted aryl group. [00225] Heteroaryl refers to a radical of a 5-10 membered monocyclic or bicyclîc 4n+2 aromatic ring system (e.g., having 6 or 10 π électrons shared in a cyclic array) having ring carbon atoms and 1-4 ring heteroatoms provided in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen and sulfur (“5-10 membered heteroaryl). În heteroaryl groups that contain onc or more nitrogen atoms, the point of attachment can bc a carbon or nitrogen atom, as valency permits. Heteroaryl bicyclîc ring Systems can include one or more heteroatoms in one or both rings. “Heteroaryl includes ring Systems wherein the heteroaryl ring, as defined above, is fused with one or more carbocyclyl or heterocyclyl groups wherein the point of attachment is on the heteroaryl ring, and in such instances, the number of ring members continue to designate the number of ring members in the heteroaryl ring System. “Heteroaryl also includes ring Systems wherein the heteroaryl ring, as defined above, is fused with one or more aryl groups wherein the point of attachment is either on the aryl or heteroaryl ring, and in such instances, the number of ring 30 members désignâtes the number of ring members in the fused (aryl/heteroaryl) ring system.
Bicyclîc heteroaryl groups wherein one ring does not contain a heteroatom (e.g., indolyl,
-70quinolinyl, carbazolyl, and the like) the point of attachment can be on either ring, i.e., either the ring bearing a heteroatom (e.g., 2-indolyl) or the ring that does not contain a heteroatom (e.g., 5-indolyl).
[00226] In some embodiments, a heteroaryl group is a 5-10 membered aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms provided in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur (“5-10 membered heteroaryl). In some embodiments, a heteroaryl group is a 5-8 membered aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms provided in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur (“5-8 membered heteroaryl). In some embodiments, a heteroaryl group is a 5-6 membered aromatic ring System having ring carbon atoms and 1-4 ring heteroatoms provided in the aromatic ring system, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur (“5-6 membered heteroaryl”). In some embodiments. the
5-6 membered heteroaryl has 1-3 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, the 5-6 membered heteroaryl has 1-2 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, the 5-6 membered heteroaryl has 1 ring heteroatom selected from nitrogen, oxygen, and sulfur. Unless otherwise specified, each instance of a heteroaryl group is independently optionally substituted, i.e., unsubstituted (an “unsubstituted heteroaryl**) or substituted (a “substituted heteroaryl”) with one or more substituents. In certain embodiments, the heteroaryl group is unsubstituted 5-14 membered heteroaryl. In certain embodiments, the heteroaryl group is substituted 5-14 membered heteroaryl.
[00227] Exemplary 5-membered heteroaryl groups containing one heteroatom include, without limitation, pyrrolyl, furanyl and thiophenyl. Exemplary 5-membered heteroaryl groups containing two heteroatoms include, without limitation, imidazolyl, pyrazolyl, oxazolyl, isoxazolyl, thiazolyl, and isothiazolyl. Exemplary 5-membered heteroaryl groups containing three heteroatoms include, without limitation, triazolyl, oxadiazolyl, and thiadiazolyl. Exemplary 5-membercd heteroaryl groups containing four heteroatoms include, without limitation, tetrazolyl. Exemplary 6-membered heteroaryl groups containing one heteroatom include, without limitation, pyridinyl. Exemplary 6-membered heteroaryl groups containing two heteroatoms include, without limitation, pyridazinyl, pyrimidinyl, and pyrazinyl. Exemplary 6-membered heteroaryl groups containing three or four heteroatoms include, without limitation, triazinyl and tetrazinyl, respectively. Exemplary 7-membered heteroaryl groups containing one heteroatom include, without limitation, azepinyl, oxepinyl,
-71and thtepinyl. Exemplary 5,6-bicychc heteroaryl groups include, without limitation, indolyl, isoindolyl, indazolyl, benzotriazolyl, benzothiophenyl, isobenzothîophenyl, benzofuranyl, benzoisofuranyl, benzîmidazolyl, benzoxazolyl, benzisoxazolyl, benzoxadiazolyl, benzthîazolyl, benzisothiazolyl, benzthïadiazolyl, indolizinyI, and purinyl. Exemplary 6,6bicyclic heteroaryl groups include, without limitation, naphthyridinyl, pteridinyl, quinolinyl, isoquinolinyl, cinnolinyl, qulnoxalinyl, phthalazinyl, and quinazolinyl.
[00228] Examples of représentative heteroaryls include the following:
Q Q O O O Q
N.
N v T ~ ' wherein each Y is selected from carbonyl, N, NR, O, and S; and R6! is independently hydrogen, Cj-Cg alkyl, C3-Cj0 cycioalkyl, 4-10 membered heterocyclyl, Cô-Cjq aryl, and 5-10 membered heteroaryl.
[00229] Examples of représentative aryl having hetero atoms containing substitution include the following:
« >
Y .
wherein each W is selected from CfR66);!, NR6S, O, and S; and each Y is selected from carbonyl, NRÉS, O and S; and R66 is independently hydrogen, Cj-Cg alkyl, CjC|q cycioalkyl, 4-10 membered heterocyclyl, Cs-Cjo aryl, and 5-10 membcred heteroaryl. [00230] “Heteroaralkyr is a subset of alkyl and heteroaryl, as defined herein, and refers to an optionally substituted alkyl group substituted by an optionally substituted heteroaryl group.
[00231] “Carbocyclyl*’ or '‘carbocyclic” refers to a radical of a non-aromatic cyclic hydrocarbon group having from 3 to 10 ring carbon atoms (“Cj_|0 carbocyclyl”) and zéro heteroatoms in lhe non-aromatic ring system. In some embodiments, a carbocyclyl group has to 8 nng carbon atoms (Cj-e carbocyclyl'*). In some embodiments, a carbocyclyl group has
to 6 ring carbon atoms (C3-6 carbocyclyl). In some embodiments, a carbocyclyl group has 3 to 6 ring carbon atoms (“C3-6 carbocyclyl”). In some embodiments, a carbocyclyl group has 5 to I0 ring carbon atoms (Cj-ιο carbocyclyl”). Exemplary C3-6 carbocyclyl groups include, without limitation, cyclopropyl (C3), cyclopropenyl (C3), cyclobutyl (C4), cyclobutenyl (C4), cyclopentyl (Cj), cyclopentenyl (Cj), cyclohexyl (Ce), cyclohexenyl (Ce), cyclohexadienyl (Cô), and the like. Exemplary C3-8 carbocyclyl groups include, without limitation, the aforementioned Cs-e carbocyclyl groups as well as cycloheptyl (C7), cycloheptenyl (C7), cycloheptadienyl (C7), cycloheptatrienyl (C7), cyclooctyl (Ce), cyclooctenyl (Cg), bicyclo[2.2.1Jheptanyl (C7), bicyclo[2.2.2]octanyl (Cg), and the like. Exemplary C3-10
I0 carbocyclyl groups include, without limitation, the aforementioned C3_g carbocyclyl groups as well as cyclononyl (C9), cydononenyl (C?), cyclodecyl (Cio), cyclodecenyl (Cio), octahydro-IH-indenyl (C9), decahydronaphthalenyl (Cio), spiro[4.5]decanyl (Cio), and the like. As the foregoing examples illustrate, in certain embodiments, the carbocyclyl group is either monocyclic (“monocyclic carbocyclyl”) or contain a fused, bridged or spiro ring system such as a bicyclic system (“bicyclic carbocyclyl) and can be saturated or can be partially unsaturated. “Carbocyclyl” also includes ring Systems wherein the carbocyclyl ring, as defined above, is fused with one or more aryl or heteroaryl groups wherein the point of attachment is on the carbocyclyl ring, and in such instances, the number of carbons continue to desîgnate the number of carbons in the carbocyclic ring system. Unless otherwise specified, each instance of a carbocyclyl group is independently optionally substituted, Le., unsubstituted (an unsubstituted carbocyclyl”) or substituted (a substituted carbocyclyl”) with one or more substituents. In certain embodiments, the carbocyclyl group is unsubstituted C3_io carbocyclyl. In certain embodiments, the carbocyclyl group is a substituted C3-10 carbocyclyl.
[00232] In some embodiments, “carbocyclyl” is a monocyclic, saturated carbocyclyl group having from 3 to I0 ring carbon atoms (“C3-10 cycloalkyl”). In some embodiments, a cycloalkyl group has 3 to 8 ring carbon atoms (Cs-g cycloalkyl”). In some embodiments, a cycloalkyl group has 3 to 6 ring carbon atoms (C3-6 cycloalkyl”). In some embodiments, a cycloalkyl group has 5 to 6 ring carbon atoms (Cj-g cycloalkyl”). In some embodiments, a cycloalkyl group has 5 to I0 ring carbon atoms (“C5-10 cycloalkyl”). Examples of Cj-g cycloalkyl groups include cyclopentyl (Cj) and cyclohexyl (Cj). Examples of C3-6 cycloalkyl groups include the aforementioned Cs_6 cycloalkyl groups as well as cyclopropyl (C3) and cyclobutyl (Q), Examples of C3_g cycloalkyl groups include the aforementioned Cj-g cycloalkyl groups as well as cycloheptyl (C7) and cyclooctyl (Cg). Unless otherwise specified,
-73each instance of a cycloalkyl group is independently unsubstituted (an “unsubstituted cycloalkyl”) or substituted (a “substituted cycloalkyl”) with one or more substituents. In certain embodiments, the cycloalkyl group is unsubstituted C3-10 cycloalkyl. In certain embodiments, the cycloalkyl group is substituted C3.10 cycloalkyl.
[00233] “Heterocyclyl” or heterocyclic” refers to a radical of a 3- to 10-membered non-aromatic ring system having ring carbon atoms and 1 to 4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, sulfur, boron, phosphorus, and silicon (“3-10 membered heterocyclyl”). In heterocyclyl groups that contain one or more nitrogen atoms, the point of attachment can be a carbon or nitrogen atom, as valency permits.
A heterocyclyl group can either be monocyclic (“monocyclic heterocyclyl) or a fused, bridged or spiro ring system such as a bicyclic system (bicyclic heterocyclyl”), and can be saturated or can be partially unsaturated. Heterocyclyl bicyclic ring Systems can include one or more heteroatoms in one or both rings. “Heterocyclyl” also includes ring Systems wherein the heterocyclyl ring, as defined above, is fused with one or more carbocyclyl groups wherein the point of attachment is either on the carbocyclyl or heterocyclyl ring, or ring Systems wherein the heterocyclyl ring, as defined above, is fused with one or more aryl or heteroaryl groups, wherein the point of attachment is on the heterocyclyl ring, and in such instances, the number of ring members continue to designate the number of ring members in the heterocyclyl ring system. Unless otherwise specified, each instance of heterocyclyl is independently optionally substituted. Le., unsubstituted (an “unsubstituted heterocyclyl) or substituted (a substituted heterocyclyl) with one or more substituents. In certain embodiments, the heterocyclyl group is unsubstituted 3-10 membered heterocyclyl. In certain embodiments. the heterocyclyl group is substituted 3-10 membered heterocyclyl. [00234] In some embodiments, a heterocyclyl group îs a 5-10 membered non25 aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen. sulfur, boron, phosphorus. and silicon (5-10 membered heterocyclyl). In some embodiments, a heterocyclyl group is a 5-8 membered non-aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur (5-8 membered heterocyclyl). In some embodiments, a hcterocyclyi group is a 5-6 membered non-aromatic ring system having ring carbon atoms and 1-4 ring heteroatoms, wherein each heteroatom is independently selected from nitrogen, oxygen, and sulfur (5-6 membered heterocyclyl). In some embodiments, the 5-6 membered heterocyclyl has 1-3 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, the 5-6
-74membered heterocyclyl has 1-2 ring heteroatoms selected from nitrogen, oxygen, and sulfur. In some embodiments, tlie 5-6 membered heterocyclyl lias one ring heteroatom selected from nitrogen, oxygen, and sulfur.
[00235] Exemplary 3-membered heterocyclyl groups containing one heteroatom include, without limitation, azirdinyl, oxiranyl, thioranyl. Exemplary 4-membcred heterocyclyl groups containing one heteroatom include, without limitation, azetidinyl, oxetanyl and thietanyl. Exemplary 5-membered heterocyclyl groups containing one heteroatom include, without limitation, tetrahydrofuranyl, dihydrofuranyl, tetrahydrothiophenyl, dihydrothiophenyl, pyrrolidinyl, dihydropyirolyl and pyrrolyl-2,510 dione. Exemplary 5-membered heterocyclyl groups containing two heteroatoms include, without limitation, dioxolanyl, oxasulfuranyl, disulfuranyl, and oxazolidin-2-one. Exemplary
5-membered heterocyclyl groups containing three heteroatoms include, without limitation, triazolinyl, oxadiazolinyl, and thiadiazolinyl. Exemplary 6-membered heterocyclyl groups containing one heteroatom Include, without limitation, piperidinyl, tetrahydropyranyl, dîhydropyridinyl, and thîanyl. Exemplary 6-membered heterocyclyl groups containing two heteroatoms include, without limitation, piperazinyl, morpholinyl, dithianyl, dioxanyl. Exemplary 6-membered heterocyclyl groups containing two heteroatoms include, without limitation, triazinanyl. Exemplary 7-membered heterocyclyl groups containing one heteroatom include, without limitation, azepanyl, oxepanyl and thiepanyl. Exemplary 820 membered heterocyclyl groups containing one heteroatom include, without limitation, azocanyl, oxecanyl and thiocanyl. Exemplary 5-membered heterocyclyl groups fused to a aryl ring (also referred to herein as a 5,6-bicyclic heterocyclic ring) include, without limitation, indolinyl, isoindolinyl, dihydrobenzofuranyl, dihydrobenzothienyl, bcnzoxazolinonyl, and the like. Exemplary 6-membered heterocyclyl groups fused to an aryl ring (also referred to herein as a 6,6-bîcyclic heterocyclic ring) include, without limitation, tctrahydroquinolinyl, tetrahydroisoquinolinyl, and the like.
[00236] Particular examples of heterocyclyl groups are shown in the following illustrative examples:
-75cd
wherein each W is selected from CR , C(R67)2, NR , O, and S; and each Y is selected from NR67,0, and S; and R67 is independently hydrogen, CpCg alkyl, C3-C10 cycloalkyl, 4*10 membered heterocyclyl, Cfi-Cm aryl, 5-10 membered heteroaryl. These heterocyclyl rings may be optionaliy substituted with one or more substituents selected from the group consisting of the group consisting of acyl, acylamino, acyloxy, alkoxy, alkoxycarbonyl, alkoxycarbonylamino, amino, substituted amino, aminocarbonyl (carbamoyl or amido), aminocarbonylamino, aminosulfonyl, sulfonylamino, aryl, aryloxy, azido, carboxyl, cyano, cycloalkyl, halogen, hydroxy, keto, nitro, thial, -S-alkyl, -S-aryl, -S(O)-alkyl,-S(O)-aryl, 10 S(O)r&lkyl, and -S(O)i-aryl. SubstitutIng groups include carbonyl or thiocarbonyl which provide, for example, lactam and urea dérivatives.
[00237] “Hetero” when used to descrïbe a compound or a group présent on a compound means that one or more carbon atoms in the compound or group hâve been replaced by u nitrogen, oxygen, or sulfur heteroatom. Hetero may be applied to any of the hydrocarbyl groups described above such as alkyl, e.g., heteroalkyl, cycloalkyl, e.g., heterocyclyl, aryl, e.g, heteroaryl, cycloalkenyl, e.g,.cycloheteroalkenyl, and the like having from 1 to 5, and particularly from I to 3 heteroatoms.
[00238] Acyl” refers to a radical -CtOJR20, where R:o is hydrogen, substituted or unsubstitued alkyl, substituted or unsubstitued alkenyl, substituted or unsubstitued alkynyl, substituted or unsubstitued carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstitued heteroaryl, as defined herein. Alkanoyl is an acyl group wherein RM is a group other than hydrogen. Représentative acyl groups include, but are not limited to. formyl (-CHO), acetyl (-C(=O)CH]), cyclohexylcarbonyl, cyclohexylmethylcarbonyl, benzoyl (-C(=O)Ph), benzylcarbonyl (-C(=O)CHiPh), —C(O)25 CrCg alkyl, -QOHCHiMCô-Cjq aryl), -QOHCH?),^-10 membered heteroaryl), -C(O)(CH2X(Cj-C,o cycloalkyl), and -C(O)-(CHi)t(4-10 membered heterocyclyl), wherein t is an integer from 0 to 4. In certain embodiments, R21 is C|-Cg alkyl, substituted with halo or
-76hydroxy; orC3-CI0 cycloalkyl, 4-10 membered heterocyclyl, Cû-Ciq aryl, arylalkyl, 5-10 membered heteroaryl or hcteroarylalkyl, each of which is substituted with unsubstituted C|C4 alkyl, halo, unsubstituted C1-C4 alkoxy, unsubstituted C1-C4 haloalkyl, unsubstituted CiC4 hydroxyalkyl, or unsubstituted C1-C4 haloalkoxy or hydroxy.
[00239] “Acylamino” refers to a radical -NR22C(O)R23, where each instance of R22 and
R23 is independently hydrogen, substituted or unsubstitued alkyl, substituted or unsubstitued alkenyl, substituted or unsubstitued alkynyl, substituted or unsubstitued carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstitued heteroaryl,, as defined herein, or R22 is an amino protecting group. Exemplary “acylamino” groups include, but are not limited to, formylamino, acetylamino, cyclohexylcarbonylamino, cyclohexylmethyl-carbonylamino, benzoylamino and benzylcarbonylamino. Particular exemplary “acylamino” groups are -NR24C(O)-C]-C8 alkyl, -NR24C(O)-(CH2),(C6-C|U aryl), -NRI4C(O)-(CH2)l(5-l0 membered heteroaryl), -NR24C(O)(CH2),(C3-C|0 cycloalkyl), and -NR24C(O)-(CH2)t(4-!0 membered heterocyclyl), wherein t is an integer from 0 to 4, and each R24 independently represents H or C|-Cg alkyl.ïn certain embodiments,
R23 is H, C|-Ca alkyl, substituted with halo or hydroxy,
C3-C]o cycloalkyl, 4-10 membered heterocyclyl, Cô-Cto aryl, arylalkyl, 5-10 membered · heteroaryl or heteroarylalkyl, each of which is substituted with unsubstituted C1-C4 alkyl, halo, unsubstituted C1-C4 alkoxy, unsubstituted C|-C4 haloalkyl, unsubstituted C1-C4 hydroxyalkyl, or unsubstituted Ct-C4 haloalkoxy or hydroxy, and
R26 is H, C|-Cg alkyl, substituted with halo or hydroxy,
C3-C10 cycloalkyl, 4-10 membered heterocyclyl, Ce-Cio aryl, arylalkyl, 5-10 membered heteroaryl or heteroarylalkyl, each of which is substituted with unsubstituted C1-C4 alkyl, halo, unsubstituted C1-C4 alkoxy, unsubstituted C|*C4 haloalkyl, unsubstituted C1-C4 hydroxyalkyl, or unsubstituted C1-C4 haloalkoxy or hydroxyl; provided at least one of R23 and R26 is other than H.
[00240] “Acyloxy” refers to a radical -OC(O)R27, where R27 is hydrogen, substituted or unsubstitued alkyl, substituted or unsubstitued alkenyl, substituted or unsubstitued alkynyl, substituted or unsubstitued carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstitued heteroaryl, as defined herein. Représentative examples include, but are not limited to, formyl, acetyl, cyclohexylcarbonyl, cyclohcxylmethylcarbonyl, benzoyl and benzylcarbonyl. In certain embodiments.
-77R18 is Ci-Cs alkyl, substituted with halo or hydroxy; Cj-Cio cycloalkyl, 4-10 membered heterocyclyl, Q-Cio aryl, arylalkyl, 5-10 membered heteroaryl or heteroarylalkyl,each of which is substituted with unsubstituted C1-C4 alkyl, halo, unsubstituted Cj-C4 alkoxy, unsubstituted Cj-C4 haloalkyl, unsubstituted C1-C4 hydroxyalkyl, or unsubstituted C|-C4 haloalkox y or hydroxy.
[00241] “Alkoxy” refers to the group -OR19 where Rw is substituted or unsubstituted alkyl, substituted or unsubstitued alkenyl, substituted or unsubstitued alkynyl, substituted or unsubstitued carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, or substituted or unsubstitued heteroaryl. Particular alkoxy groups are methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, nhexoxy, and 1,2-dimethylbutoxy. Particular alkoxy groups are lower alkoxy, i.e. with between 1 and 6 carbon atoms. Further particular alkoxy groups hâve between 1 and 4 carbon atoms.
[00242] In certain embodiments, R39 is a group that has 1 or more substituents, for instance from 1 to 5 substituents, and particularly from I to 3 substituents, in particular 1 substituent, selected from the group consisting of amino, substituted amino, Cô-Cio aryl, aryloxy, carboxyl, cyano, C3-C10 cycloalkyl, 4-10 membered heterocyclyl, halogen, 5-10 membered heteroaryl, hydroxyl, nitro, thioalkoxy, thioaryloxy, thiol, alkyl-S(O)·, aryl-S(O)-, alkyl-S(O)2- and aryl-S(O)i-. Exemplary ‘substituted alkoxy’ groups include, but are not limited to, -O-fCHiXtCe-Cio aryl), -O-fCHîMS-10 membered heteroaryl), -0-(CHi),(Cj-Cio cycloalkyl), and -0-(011^(4-10 membered heterocyclyl), wherein t is an integer from 0 to 4 and any aryl, heteroaryl, cycloalkyl or heterocyclyl groups présent, may themselves be substituted by unsubstituted C|-C4 alkyl, halo, unsubstituted CrC4 alkoxy, unsubstituted C|C4 haloalkyl, unsubstituted Ct-Ci hydroxyalkyl, or unsubstituted C|-C4 haloalkoxy or hydroxy. Particular exemplary ‘substituted alkoxy’ groups are -OCF3, -OCH2CF3, -OCH2Ph, -OCHî-cyclopropyl, -OCH2CH2OH, and -OCH2CH2NMe2.
[00243] Amino” refers to the radical -NH2.
[00244] “Substituted amino” refers to an amino group of the formul a -N(R38)2 wherein •Jl)
R is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstitued alkenyl, substituted or unsubstitued alkynyl, substituted or unsubstitued carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, substituted or unsubstitued heteroaryl, or an amino protecting group, wherein at least one of R38 is not a hydrogen. In certain embodiments.
-78each R38 is independently selected from: hydrogen, C|-C8 alkyl, C3-C8 alkenyl, C3-C8 alkynyl, Q-Cio aryl, 5-10 membered heteroaryl, 4-10 membered heterocyclyl, or C3-C10 cycloalkyl; or C|-C8 alkyl, substituted with halo or hydroxy; C3-C8 alkenyl, substituted with halo or hydroxy; C3-C8 alkynyl, substituted with halo or hydroxy, or -(CHi^fCe-Cio aryl), 5 (CHi)t(5-10 membered heteroaryl), -(CH3)i(C3-Cio cycloalkyl), or -<CH2)t(4-10 membered heterocyclyl), wherein t is an integer between 0 and 8, each of which is substituted by unsubstituted C1-C4 alkyl, halo, unsubstituted C1-C4 alkoxy, unsubstituted C1-C4 haloalkyl, unsubstituted C1-C4 hydroxyalkyl, or unsubstituted C1-C4 haloalkoxy or hydroxy; or both R38 groups are joined to form an alkylene group.
[00245] Exemplary ‘substituted amino’ groups are -NR39-C|-C8 alkyl, -NR39(CH2)t(C6-C|o aryl),-NR3’-(CH2)1(5-l0 membered heteroaryl), -NR3’-(CH2)t(C3-C|O cycloalkyl), and-NR39-(CH2)i(4-10 membered heterocyclyl), wherein t is an integer from 0 to 4, for instance 1 or 2, each R39 independently represents H or C|-C8 alkyl; and any alkyl groups présent, may themselves be substituted by halo, substituted or unsubstituted amino, or 15 hydroxy; and any aryl, heteroaryl, cycloalkyl, or heterocyclyl groups présent, may themselves be substituted by unsubstituted C1-C4 alkyl, halo, unsubstituted C1-C4 alkoxy, unsubstituted C1-C4 haloalkyl, unsubstituted C1-C4 hydroxyalkyl, or unsubstituted C1-C4 haloalkoxy or hydroxy. For the avoidance of doubt the term ‘substituted amino’ includes the groups alkylamino, substituted alkylamino, alkylarylamino, substituted alkylarylamino, aryl ami no, 20 substituted arylamino, dialkylamino, and substituted dialkylamino as defined below.
Substituted amino encompasscs both monosubstituted amino and disubsti tu ted amino groups. [00246] “Azido refers to the radical -N3.
[00247] “Carbamoyl or “amido refers to the radical -C(O)NH2.
[00248] Substituted carbamoyl or substituted amido refers to the radical 25 C(O)N(R62)2 wherein each R63 is independently hydrogen, substituted or unsubstituted alkyl, substituted or unsubstitued alkenyl. substituted or unsubstitued alkynyl, substituted or unsubstitued carbocyclyl, substituted or unsubstituted heterocyclyl, substituted or unsubstituted aryl, substituted or unsubstitued heteroaryl, or an amino protecting group, wherein at least one of R61 is not a hydrogen. In certain embodiments, R62 is selected from H, 30 Ci-C8 alkyl, C3-Cio cycloalkyl, 4-10 membered heterocyclyl, Q-C10 aryl, aralkyl, 5-10 membered heteroaryl, and heteroaralkyl; or CpC8 alkyl substituted with halo or hydroxy; or C3-Cio cycloalkyl, 4-10 membered heterocyclyl. Ce-Cio aryl, aralkyl, 5-10 membered heteroaryl, or heteroaralkyl, each of which is substituted by unsubstituted C1-C4 alkyl, halo.
-79unsubstituted C1-C4 alkoxy, unsubstituted C1-C4 haloalkyl, unsubstituted C1-C4 hydroxyalkyl, or unsubstituted Cj-Cj haloalkoxy or hydroxy; provided that at least one R62 is other than H. [00249] Exemplary 'substituted carbamoyl' groups include, but are not limited to, C(O) NRw-CrC8 alkyl. -CiOJNR^CHzJ^Cé-Cæ aryl), -C(O)N^-(CH2)((5-10 membered 5 heteroaryl), -CCOJNR^CHjMCa-Cio cycloalkyl). and -C(O)NRM-(CH2)l(4-10 membered heterocyclyl), wherein t is an integer from O to 4, each R64 independently represents H or C|Cg alkyl and any aryl, heteroaryl, cycloalkyl or heterocyclyl groups présent, may themselves be substituted by unsubstituted C|-C4 alkyl, halo, unsubstituted Ci-C4 alkoxy, unsubstituted C|-C4 haloalkyl, unsubstituted C1-C4 hydroxyalkyl, or unsubstituted Ci-Ci haloalkoxy or 10 hydroxy.
[00250] ‘Carboxy’ refers to the radical -C(O)OH.
[00251] “Cyano refers to lhe radical -CN.
[00252] “Halo or “halogen refers to fluoro (F), chloro (Cl), bromo (Br), and iodo (I). In certain embodiments, the halo group is either fluoro or chloro. In further embodiments, the 15 halo group is iodo.
[00253] “Hydroxy” refers to lhe radical -OH.
[00254] “Nitro” refers to lhe radical -NO2.
[00255] “Cycloalkylalkyl” refers to an alkyl radical in which the alkyl group is substituted with a cycloalkyl group. Typical cycloalkylalkyl groups include, but are not 20 limited lo, cyclopropyl methyl, cyclobuiylmethyl, cyclopenlylmethyl, cyclohexylmeihyl, cycloheptylmethyl, cydooctylmethyl, cyclopropylethyl, cyclobulylethyl, cyclopcniylelhyl, cyclohexyl ethyl, cycloheptylethyl, and cyclooctylelhyl, and the like.
[00256] “Hetcrocyclylalkyl” refers to an alkyl radical in which the alkyl group is substituted with a heterocyclyl group. Typical heterocyclylalkyl groups include, but are not 25 limited to, pyrrolidinylmethyl, piperidinylmethyl, piperazinylmethyl, morpholinylmethyl, pyrrolidinylethyl, piperidinylethy), piperazinylethy), morpholinylethyl, and the like.
[00257] “Cycloalkenyl refers to substituted or unsubstituted carbocydyl group having from 3 to 10 carbon atoms and having a single cyclic ring or multiple condensed rings, including fused and bridged ring Systems and having at least one and particularly from 1 to 2 30 sites of definie unsaturation. Such cycloalkenyl groups include, by way of example, single ring structures such as cyclohexenyl, cyclopentenyl, cyclopropenyl, and the like.
[00258] “Fused cycloalkenyl refers to a cycloalkenyl having two of its ring carbon atoms in common with a second aliphatic or aromatic ring and having its olefinic unsaturation located to impart aromaticity to the cycloalkenyl ring.
-80[00259] Ethenyl refera to substituted or unsubstituted -(C=C)-.
[00260] “Ethylene” refera to substituted or unsubstituted -(C-C)-.
[00261] “Ethynyl refera to -(CsC)-.
[00262] “Nîtrogen-containing heterocyclyr* group means a 4- to 7- membered non5 aromatic cyctic group containing at least one nitrogen atom, for example, but without limitation, morpholine, piperidine (e.g. 2-piperidinyl, 3-piperidinyl and 4-piperidinyl), pyrrotidine (e.g. 2-pyrrolidinyl and 3-pyrrotidinyl), azetidine, pyrrolidone, imidazoline, imidazolidinone, 2-pyrazoline, pyrazolidine, piperazine, and N-alkyl piperazines such as Ν'methyl piperazîne. Particular examples include azetidine, piperidone and piperazone.
[00263] “Thioketo” refera to the group =S.
[00264] Exemplary carbon atom substituents include, but are not limited to, halogen, CN, -NO2, -N3. -SO2H, -SO3H, -OH, -ORaa, -ON(Rbb)2, -N(Rbb)2, -N(Rbb)3+X-, N(ORcc)Rbb, -SH, -SRaa, -SSRcc, -C(=O)Raa, -CO2H, -CHO, -C(ORcc)2, -CO2Raa, OC(=O)Raa, -OCO2Raa, -C(=O)N(Rbb)2, -OC(=O)N(Rbb)2, -NRbbC(=O)Raa, 15 NRbbCO2Raa, -NRbbC(=O)N(Rbb)2, -C(=NRbb)Raa, -C(=NRbb)ORaa, -OC(=NRbb)Raa, -OC(=NRbb)ORaa, -C(=NRbb)N(Rbb)2, -OC(=NRbb)N(Rbb)2, NRbbC(=NRbb)N(Rbb)2, -C(=O)NRbbSO2Raa, -NRbbSO2Raa, -SO2N(Rbb)2, -SO2Raa, -S02ORaa, -OSO2Raa, -S(=O)Raa, -OS(=O)Raa, -Si(Raa)3, -OSÎ(Raa)3 -C(=S)N(Rbb)2. -C(=O)SRaa, -C(=S)SRaa, -SC(=S)SRaa, -SC(=O)SRaa, -OC(=0)SRaa, -SC(=O)ORaa, 20 SC(=O)Raa, -P(=O)2Raa, -OP(=O)2Raa, -P(=O)(Raa)2, -OP(=O)(Raa)2, OP(=O)(ORcc)2, -P(=O)2N(Rbb)2, -OP(=O)2N(Rbb)2, -P(=O)(NRbb)2, OP(=O)(NRbb)2, -NRbbP(=O)(ORcc)2, -NRbbP(=O)(NRbb)2, -P(Rcc)2, -P(Rcc)3, OP(Rcc)2, -OP(Rcc)3, -B(Raa)2, -B(ORcc)2, -BRaa(ORcc), C1-10 alkyl, C1-10 perhaloalkyl, C2-10 alkenyl, C2-10 alkynyl, C3-I0carbocyclyl, 3-14 membered heterocyclyl, C6-14 aryl, and 5-14 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0,1,
2,3,4, or 5 Rdd groups; or two geminal hydrogens on u carbon atom are replaced with the group =0, =S, =NN(Rbb)2, =NNRbbC(=O)Raa, =NNRbbC(=O)ORaa, =NNRbbS(=O)2Raa, =NRbb, or =NORcc;
each instance of Rua is, independently, selected from CI-10 alkyl, Cl-10 perhaloalkyl, C2-I0 alkenyl, C2-10 alkynyl, C3-10 carbocyclyl, 3-14 membered heterocyclyl, C6-I4 aryl, and 5-14 membered heteroaryl, or two Raa groups are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl,
-81alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0,1,2,3,4, or 5 Rdd groups;
each instance of Rbb is, independently, selected from hydrogen, -OH, -ORaa, -N(Rcc)2, -CN, -C(=O)Raa, -C(=O)N(Rcc)2, -CO2Raa, -SO2Raa, -C(-NRcc)ORaa, 5 C(=NRcc)N(Rcc)2, -SO2N(Rcc)2, -SO2Rcc, -SO2ORcc, -SORaa, -C(=S)N(Rcc)2, C(=O)SRcc, -C(=S)SRcc, -P(=O)2Raa, -P(=O)(Raa)2, -P(=O)2N(Rcc)2, -P(=O)(NRcc)2, Cl-10 alkyl, Cl—10 perhaloalkyl, C2-10 alkenyl, C2-I0 alkynyl, C3-10 carbocyclyl, 3-14 membered heterocyclyl, C6-14 aryl, and 5-14 membered heteroaryl, or two Rbb groups are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1,2,3,4, or 5 Rdd groups;
each instance of Rcc is, independently, selected from hydrogen, Cl-10alkyl, Cl-10 perhaloalkyl, C2-10 alkenyl, C2-10 alkynyl, C3-10 carbocyclyl, 3-14 membered heterocyclyl, C6-14 aryl, and 5-14 membered heteroaryl, or two Rcc groups are joined to 15 form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0,1,2,3,4, or 5 Rdd groups;
each instance of Rdd is, independently, selected from halogen, -CN, -NO2, N3, -SO2H, -SO3H, -OH, -ORcc, -ON(Rff)2, -N(Rff)2, -N(RfD3+X-, -N(ORee)Rff, 20 SH, -SRee, -SSRee, -C(=O)Ree, -CO2H, -CO2Ree, -OC(=O)Ree, -OCO2Ree, C(=O)N(Rff)2, -OC(t=O)N(Rff)2, -NRffC(=O)Rce, -NRffCO2Ree, -NRffC(=O)N(Rff)2, C(=NRÏÏ)ORee, -OC(=NRff)Ree, -OC(=NRff)ORee, -C(=NRff)N(Rff)2, OC(=NRff)N(Rff)2, -NRffC(=NRff)N(Rff)2,-NRffSO2Ree, -SO2N(RfD2, -SO2Ree, SO2ORcc, -OSO2Ree, -S(=O)Ree, -Si(Rcc)3, -OSi(Ree)3, -C(=S)N(RfD2, -C(=O)SRee, 25 C(=S)SRee. -SC(=S)SRce, -P(=O)2Rce, -P(=O)(Ree)2. -OP(=O)(Ree)2, -OP(=O)(ORee)2, C1-6 alkyl, Cl-6 perhaloalkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 carbocyclyl, 3-10 membered heterocyclyl, C6-I0 aryl, 5-10 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, I,
2,3,4, or 5 Rgg groups, or two geminal Rdd substituents can be joined to form -O or =S;
each instance of Ree is, independently, selected from Cl-6 alkyl, Cl-6 perhaloalkyl, C2-6 alkenyl, C2-6 alkynyl, C3-I0 carbocyclyl, C6-10 aryl, 3-10 membered heterocyclyl, and 3-10 membered heteroaryl, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0, 1,2,3,4, or 5 Rgg groups;
-82each instance of Rff is, independently, selected from hydrogen, CI-6 alkyl, C1-6 perhaloalkyl, C2-6 alkenyl, C2-6 alkynyl, C3-10 carbocyclyl, 3-10 membered heterocyclyl, C6-I0 aryl and 5-10 membered heteroaryl, or two Rff groups are joined to form a 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl, 5 alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0,1,2,3,4, or 5 Rgg groups; and each instance of Rgg is, independently, halogen, -CN, -NO2, -N3, -SO2H, SO3H, -OH, -OCI-6 alkyl, -ON(CI-6alkyl)2, -N(Cl-6alkyl)2, -N(CI-6 alkyl )3+X-,NH(CI-6 alkyl)2+X-, -NH2(Cl-6 alkyl) +X-.-NH3+X-, -N(OCI-6 alkyl)(C 1-6 alkyl), 10 N(OH)(C I -6 alkyl), -NH(OH), -SH, -SC 1-6 alkyl, -SS(C 1-6 alkyl), ~C(=O)(C 1-6 alkyl), CO2H, ~CO2(Cl-6 alkyl), -OC(=O)(Cl-6 alkyl),-OCO2(Cl-6 alkyl), -C(=O)NH2, C(=O)N(C 1-6 alkyl)2, -OC(=O)NH(CI-6 alkyl), -NHC(=O)( CI-6 alkyl), -N(Cl-6 alky!)C(=O)( Cl-6 alkyl), -NHCO2(Cl-6 alkyl), -NHC(=O)N(Cl-6 alkyl)2, NHC(=O)NH(C 1-6 alkyl), -NHC(=O)NH2, -C(=NH)O(Cl-6 alkyl),-OC(=NH)(CI-6 alkyl), -OC(=NH)OCl-6 alkyl, -C(=NH)N(Cl-6 alkyl)2, -C(=NH)NH(Cl-6 alkyl), C(-NH)NH2, -OC(=NH)N(Cl-6 alkyl)2, -OC(NH)NH(Cl-6 alkyl), -OC(NH)NH2, NHC(NH)N(Cl-6 alkyl)2, -NHC(=NH)NH2, -NHSO2(Cl-6 alkyl), -SO2N(Cl-6 alkyl)2, -SO2NH(CI-6 alkyl), -SO2NH2.-SO2C1-6 alkyl,-SO2OCl-6 alkyl,-OSO2C1-6 alkyl,SOC1-6 alkyl,-Si(C 1-6 alkyl)3,-OSi(Cl-6 alkyl)3 -C(=S)N(C 1-6 alkyl)2, C(=S)NH(CI20 6 alkyl), C(=S)NH2, -C(=O)S(Cl-6 alkyl), -C(=S)SCl-6 ajkyl, -SC(=S)SCl-6 alkyl,P(=O)2(Cl-6 alkyl), -P(=O)(Cl-6 alkyl)2, -OP(=O)(Cl-6 alkyl)2, -OP(=O)(OCI-6 alkyl)2, Cl-6 alkyl, Cl-6 perhaloalkyl, C2-6 alkenyl, C2-6 alkynyl, C3-I0 carbocyclyl, C6-10 aryl, 3-10 membered heterocyclyl, 5-10 membered heteroaryl; or two geminal Rgg substituents can be joined to form =0 or =S; wherein X- is a counterion.
[00265] Alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl groups, as defined herein, are optionally substituted (e.g., “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 group). In general, the term “substituted, whether preceded by the term optionally or not, means that al least one hydrogen présent on a group (e.g., a carbon or nitrogen atom) is replaced with a permissible substituent, e.g., a substituent which upon substitution results in a stable compound, e.g., a compound which does not spontaneously undergo transformation such as by rearrangement, cyclizatîon, élimination, or other réaction. Unless otherwise indicated, a
-83substituted” group has a substituent at one or more substitutable positions of the group, and when more than one position in any given structure is substituted, the substituent is either the same or different at each position. The term substituted is contemplated to include substitution with ail permissible substituents of organic compounds, any of the substituents 5 described herein thaï results in the formation of a stable compound. The présent invention contemplâtes any and ail such combinations în order to arrive al a stable compound. For purposes of this invention, heteroatoms such as nitrogen may hâve hydrogen substituents and/or any suitable substituent as described herein which satisfy the valencîes of the heteroatoms and results in the formation of a stable moiety.
[00266] A “counterion or “anionic counterion” is a negatively charged group associated with a cationic quatemary amino group in order to maintain elcctronic neutrality. Exemplary counterions include halide ions (e.g., F“, Cf. Br“, Γ), NOj”, CIO/, OH, HiPOF, HSOZ, sulfonate ions (e.g., methansulfonate, trifluoromethanesulfonate, p-loluenesulfonate, bcnzcnesulfonate, 10-camphor sulfonate, naphthalene-2-sulfonate, naphthalene-l-sulfonic acid-5-sulfonate, ethan-l-sulfonic acid-2 -sulfonate, and the like), and carboxylate ions (e.g., acetate, ethanoate, propanoate, benzoate, glycerate, lactate, lartrate, glycolate, and the like).
[00267] Nitrogen atoms can be substituted or unsubstituted as valency permits, and include primary, secondary, tertiary, and quartemary nitrogen atoms. Exemplary nitrogen 20 atom substitutents include, but are not limited to, hydrogen, -OH, -ORM, -N(RCC)2, -CN, C(=O)R“ -C(=0)N(Rk)2, -CO2R“, -SOjR“, -C(=NRbb)R“, -C(=NRCC)OR“, C(=NRcc)N(Rce)2, -SO2N(Rcc)2, -SO2R“. -SO2ORk, -SOR“, -C(=S)N(Rceh, -C(=O)SR«, C(=S)SRcc, -P(=O):RM, -P(=O)(R“)î, -P(=OhN(RK)3, -P(=O)(NRW)2, C).lo alkyl, C|_lo perhaloalkyl, Cwo alkenyl, C2_io alkynyl, Cj_io carbocyclyl, 3-14 membered heterocyclyl, 25 Cô-h aryl, and 5-14 membered heteroaryl, or two Rcc groups attached to a nitrogen atom are joined to form u 3-14 membered heterocyclyl or 5-14 membered heteroaryl ring, wherein each alkyl, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aryl, and heteroaryl is independently substituted with 0,1,2,3,4, or 5 RdJ groups, and wherein R“, Rbb, R”and R***1 are as defined above.
[00268] In certain embodiments, the substituent présent on a nitrogen atom is a nitrogen protecting group (also referred to as an amino protecting group). Nitrogen protecting groups include, but are not limited to, -OH, -OR“, -N(RCC)2, -C(=O)R“ -C(=O)N(R*)2, CO2R“, -SO2R“, -C(=NR“)RM, -C(=NRcc)0Ra4, -C(=NRcc)N(Rcc)2, -SO2N(RCC)21 -SO2RCC, -SO2OR“, -SORm, -C(=S)N(Rcc)2, -C(=O)SRcc, -C(=S)SRcc, C|.1O alkyl (e.g., aralkyl,
heteroaralkyl), Cj-jq alkenyl, Cj.jo alkynyl, Cî-jo carbocyclyl, 3-I4 membered heterocyclyl, Cft_i4 aryl, and 5— 14 membered heteroaryl groups, wherein each alkyi, alkenyl, alkynyl, carbocyclyl, heterocyclyl, aralkyl, aryl, and heteroaryl is independently substituted with 0,1,
2,3,4, or 5 Rdd groups, and wherein RM, R*’6, Rreand R41 are as defined herein. Nitrogen protecting groups are well known in the art and include those described in detail in Protecting Groups in Organic Synthesis, T. W. Greene and P. G. M. Wuts, 3ri édition, John Wiley & Sons, 1999, incorporated herein by reference.
[00269] For example, nitrogen protecting groups such as amide groups (e.g., — CfsOJR4) include, but are not limited to, formamide, acetamide, chloroacetamide, trichloroacetamîde, trifluoroacetamide, phenylacetamide, 3-phenylpropanamîde, picolinamide, 3-pyridylcarboxamide, N-benzoylphenylalanyl dérivative, benzamide,pphenylbenzamide, o-nitophenylacetamide, o-nitrophenoxyacetamide, acetoacetamide, (Ndithiobenzyloxyacylamino)acetamide, 3-<p-hydroxyphenyl)propananùde, 3-{onitrophenyl)propanamide, 2-methyl-2-(t>-nitrophenoxy)propanamide, 2-methyl-2-(i>15 phenylazophenoxyjpropanamide, 4-chlorobutanamide, 3-methyl-3-nitrobutanamide, onitrocinnamide, N-acetylméthionine dérivative, o-nitrobenzamide and o~ (be nzoy loxyme thyl Jbenz ami d e.
[00270] Nitrogen protecting groups such as carbamate groups (e.g., -C(=O)ORM) include, but are not limited to, methyl carbamate, ethyl carbamantet9-fluorenylmethyl carbamate (Fmoc), 9-(2-sulfo)fIuorcnylmethyl carbamate, 9-(2,7-dibromo)fluoroenylmeihyl carbamate, 2,7—di—r—buty 1—[9—( 10,10-dioxo-10,10,10,10-tetrahydrothioxanthyl)]methyl carbamate (DBD-Tmoc), 4-methoxyphenacyl carbamate (Phenoc), 2,2,2-trichloroethyl carbamate (Troc), 2-trimethylsllylethyl carbamate (Teoc), 2-phenylethyl carbamate (hZ), I(J-adamantyl)-l-methyleihyl carbamate (Adpoc), l,l-dimethyl-2-haloethylcarbamate,
1, l-dimethyl-2,2-dibromoethyl carbamate (DB-/-BOC), 1, l-dimethyl-2,2,2-trichloroethyl carbamate (TCBOC), 1—methyl—1—<4—biphenylyljethyl carbamate (Bpoc), 1-(3,5-di-1butylphenyl)-l-methylethyl carbamate (f-Bumeoc), 2-(2’-and 4‘-pyridyl)ethyl carbamate (Pyoc), 2-(N, N-dicyclohexylcarboxamido)eihyl carbamate, r—butyl carbamate (BOC), 1adamantyl carbamate (Adoc), vinyl carbamate (Voc), allyl carbamate (Alloc), 130 isopropylallyl carbamate (Ipaoc), cinnamyl carbamate (Coc), 4-nitrocinnamyl carbamate (Noc), 8-qulnolyi carbamate, A-hydroxypiperidinyl carbamate, alkyldithio carbamate, benzyl carbamate (Cbz), p-methoxybenzyl carbamate (Moz), p-nitobenzyl carbamate, p~ bromobenzy! carbamate, p-chlorobenzyl carbamate, 2,4-dîchlorobenzyl carbamate, 4mcthylsulfinylbenzyl carbamate (Msz), 9-anthrylmethyl carbamate, diphenylmethyl
carbamate, 2-methylthioethyl carbamate, 2-methylsulfonylethyl carbamate, 2-(ptoluenesulfonyl)ethyl carbamate, [2-( 1,3-dithianyl)]methyl carbamate (Dmoc), 4methylthiophenyl carbamate (Mtpc), 2,4-dimethylthiophenyl carbamate (Bmpc), 2phosphonioethyl carbamate (Peoc), 2-triphenylphosphonioisopropyl carbamate (Ppoc), 1,1— dimethy|-2-cyanoethyl carbamate, m-chloro-p-acyloxybenzyl carbamate, p~ (dihydroxyboryl)benzyl carbamate, 5-benzisoxazolyl methyl carbamate, 2-(trifluoromethyl)-
6-chromonylmethyl carbamate (Tcroc), m-nitrophenyl carbamate, 3,5-dimethoxybenzyl carbamate, o-nitro benzyl carbamate, 3,4-dimethoxy-6-nitrobenzyl carbamate, phenyl(onitrophenyl)methyl carbamate, r-amyl carbamate, S-benzyl thiocarbamate, p-cyanobenzyl carbamate, cyclobutyl carbamate, cyclohexyl carbamate, cyclopentyl carbamate, cyclopropylmethyl carbamate, p-decyloxybenzyl carbamate, 2,2-dimethoxyacylvinyl carbamate, o-f/V,/V-dimethylcarboxamido)benzyl carbamate, 1,1—dimethyl—3—(TV, jV— dimethylcarboxamidojpropyl carbamate, 1,1-dimethylpropynyl carbamate, di(2pyridyl) methyl carbamate, 2-furanyl methyl carbamate, 2-iodoelhyl carbamate, isoborynl carbamate, isobutyl carbamate, isonicotinyl carbamate, p-(p '-methoxyphenylazojbenzyl carbamate, I-methylcyclobutyl carbamate, 1-methylcyclohexyl carbamate, I-methyl-1cyclopropylmethyl carbamate, l-methyl-l-(3,5-dimethoxyphenyl)ethyl carbamate, 1methyl-l-(p-phenylazophenyl)ethyl carbamate, 1 -methyl- l-phenylethyl carbamate, 1methyl— 1—(4—pyridyljethyl carbamate, phenyl carbamate, p-(phenylazo)benzyl carbamate,
2,4,6-tri-r-butylphenyl carbamate, 4-(trimethylammonium)benzyl carb amate, and 2,4,6trimethylbenzyl carbamate.
[00271] Nitrogen protecting groups such as sulfonamide groups (e.g., -S(=O)iR“) include, but are not limited to, p-toluenesulfonamide (Ts), benzenesul fonamide, 2,3,6,trimet hy l-4-mc thox ybenzenesu I fonamide (Mtr), 2,4,6-trime t hoxybenzenesu I fonamide (Mtb), 2,6-dimethyl-4-methoxybenzenesul fonamide (Pme). 2,3,5,6-tetramethyl-4methoxybenzenesulfonamide (Mte), 4-methoxybenzenesul fonamide (Mbs), 2,4,6trimethylbenzenesulfonamide (Mts), 2,6-dimethoxy-4~methylbenzenesulfonamide (iMds), 2,2,5,7,8-pentamethylchroman-6-sulfbnamidc (Pmc), methanesul fonamide (Ms), βtrimethylsilylethanesul fonamide (SES), 9-anthracenesul fonamide, 4-(4’,8’30 dimethoxynaphthylmethyl)benzencsul fonamide (DNMBS), benzylsulfonamide, trifluoromethylsulfonamide, and phenacylsulfonamide.
[00272] Other nitrogen protecting groups include, but are not limited to, phenothiazinyl-(lO)-acyl dérivative, N’-p-toluenesulfonylaminoacyl dérivative, N‘~ phcnylaminothioacyl dérivative, M-benzoylphenylalanyl dérivative, /V-acetylmcthioninc
-86dérivative, 4,5-diphenyl-3-oxazolin-2-one, /7-phthalimide, N-dithiasuccînimide (Dts), N2,3-diphenylmaleimide, A/-2,5-dimethylpyrrole, N-1,1,4,4tetramethyldisilylazacyclopentaneadduct(STABASE),5-substituted 1,3-dimethyl-l,3,5triazacyclohexan-2-one,5-substituted l,3-dibenzyl-l,3,5-triazacyclohexan-2-one, I5 substituted 3,5-dinitro-4-pyridonc, A'-methylamine, JV-allylamine, /7-(2(trimethylsilyl)ethoxyjmethylamine (SEM), /7-3-acetoxypropylamine, W-(|-isopropyl-4nitro-2-oxo-3-pyroolin-3-yl)amine, quatemary ammonium salts, A-benzyl amine, jV—di(-4— methoxyphenyl)methylamine, jV-5-dibenzosuberylamine, AMriphenylmethylamîne (Tr), N((4-methoxyphenyl)diphenylmethyl]amine (MMTr), N-9-phenylfluorenylamine (PhF), N10 2,7-dichloro-9-fluorenylmethyleneamine, M-ferrocenylmethylamino (Fcm), /7-2picolyl amino Ν'-oxide, JV-l.l-dimethylthiomethyleneamine, jV-benzylideneamine, N-pmethoxybenzylideneamine, jV-diphenylmethyleneaminc, JV—[(2— pyridyl)mesityl]methyleneamine, /V-(A/’,/V’-dimethylaminomethylene)amîne, N,Nisopropylîdenediamine, M-p-nitrobenzylideneamine, AA-salicylideneamînc, /7-515 chlorosalicylideneamine, /V-(5-chloro-2-hydroxyphenyl)phenylmethyleneamine, Ncyclohcxylideneamine, jV-(5,5-dimelhyl-3-oxo-l-cyclohexenyl)amine, /V-borane dérivative, /V-diphenylborinic acid dérivative, JV-[phenyl(pentaacylchromium- or tungsten)acyl]amine, M-copper chelate, jV-zinc chelale, /V-nitroamine, /V-nitrosoamine, amine /V-oxide, diphenylphosphinamide (Dpp), dimethylthiophosphinamide (Mpt), diphenylthiophosphinamide (Ppt), dialkyl phosphoramidates, dibenzyl phosphoramidate, diphenyl phosphoramidate, benzenesulfenamide, o-nitrobenzenesulfenamide (Nps), 2,4dinitrobenzenesulfenamide, pentachlorobenzenesulfenamide, 2-nitro-4methoxybenzenesulfenamide, triphenylmethylsulfenamide, and 3-nitropyridinesulfenamide (Npys).
[00273] In certain embodiments, the substituent présent on an oxygen atom is an oxygen protecting group (also referred to as a hydroxyl protecting group). Oxygen protecting groups include, but are not limited to, -RM, -NfR**):, -C(=O)SR“, -C(=O)R“, -CO2RM, -C(=NRbb)R“, -C(=NRbb)OR“, -C(=NRbb)N(Rbb)2, -S(=O)RU, -SO2R“, Si(R“b.-P(RM)2, -P(Rcc)j. -P(=O)2RM, -P(=O)(R“)2, -P(=O)(OR“)2, -P(=O)2N(Rbb)2, and 30 P(=O)(NRbb)2, wherein RM, Rbb, and Rcc are as defined herein. Oxygen protecting groups are well known in the art and include those described in detail in Protecting Groups in Organic Synthesis, T. W. Greene and P. G. M. Wuts, 3M édition, John Wiley & Sons. 1999, incorporated herein by référencé.
-87[00274] Exemplary oxygen protectlng groups include, but are not limited to, methyl, methoxylmethyl (MOM), methy Ithiomethyl (MTM), t-butylthiomethyl, (phenyldimethylsilyl)methoxymethyl (SMOM), benzyloxymethyl (BOM), pmethoxybenzyloxymethyl (PMBM), (4-methoxyphenoxy)methyl (p-AOM), guaiacolmethyl (GUM), t-butoxymethyl, 4-pentenyloxymethyl (POM), siloxymethyl, 2methoxyethoxymethyl (MEM), 2,2,2-trichloroethoxymethyl, bis(2-chloroethoxy)methyl, 2(trimethylsilyl)ethoxymethyl (SEMOR), tetrahydropyranyl (THP), 3bromotetrahydropyranyl, tetrahydrothiopyranyl, l-methoxycyclohexyl, 4methoxytetrahydropyranyl (MTHP), 4-methoxytetrahydrothiopyranyl, 410 methoxytetrahydrothiopyranyl S,S-dioxide, l-[(2-chloro-4-methyl)phenyl]-4~ methoxypiperidin-4-yl (CTMP), l,4-dioxan-2-yl, tetrahydrofuranyl, tetrahydrothiofuranyl, 2,3,3a,4,5,6,7,7a-Octahydro-7,8,8-trimetliyl-4,7-methanobenzofuran-2-yl, l-ethoxyethyl, l-(2-chIorœthoxy)ethyl, l-methyl-l-methoxyethyl, l-methyl-l-benzyloxyethyl, Imethyl-l-benzyloxy-2-nuoroethyl, 2,2,2-trichloroethyl, 2-trimethylsilylethyl, 2I5 (phenylselenyl)ethyl, t- butyl, ail yl, p-chlorophenyl, p-methoxyphenyl, 2,4-dinitrophenyl, benzyl (Bn), />-methoxybenzyl, 3,4-dimethoxybenzyl, o-nitrobenzyl, p-nitrobenzyl, phalobenzyl, 2,6-dichlorobenzyl,p-cyanobenzyl,p-phenyIbenzyl, 2- picolyl, 4-picolyl, 3methyl-2-picolyl N-oxido, diphenylmethyl.p.p’-dinitrobenzhydryl, 5-dibenzosuberyl, triphenylmethyl, α-naphthyldîphenylmethyl, p-methoxyphenyldiphenylmethyl, di(p20 methoxyphenyl)phenylmcthyl, tri(p-methoxyphenyl)methyl, 4-(4'bromophenacyloxyphenyl)diphenylmethyl, 4,4',4~tris(415dichlorophthalimidophenyljmethyl, 4,4',4''-tris(levulinoyloxyphenyl)methyl,4,4',4tris(benzoyloxyphenyl)methyl, 3-(ÎmidazoI-l-yI)bis(4',4''-dimethoxyphenyl)methyl, I, Ibis(4-methoxyphenyl)-r-pyrenylmethyl, 9-anthryl, 9-(9-phenyl)xanthenyl, 9-(9-phenyl25 IO~oxo)anthryl, 1,3-benzodisulfuran-2-yl, benzisolhiazolyl S,S-dioxido, trimcthylsilyl (TMS), triethylsilyl (TES), triisopropylsilyl (TIPS), dimethylisopropylsily I (IPDMS), diethylisopropylsilyl (DEIPS), dîmethylthexylsilyl, /-butyldimethylsilyl (TBDMS), tbutyldiphenylsilyl (TBDPS), tribenzylsilyl, tri-p- xylylsilyl, triphenylsilyl, diphenylmethylsilyl (DPMS), t-butylmethoxyphenylsilyl (TBMPS), formate, benzoylformate, acetate, chloroacetate, dichloroacetate, trichloroacetate, trifluoroacetate, methoxyacetate, triphenylmelhoxyacetate, phenoxyacetate, p-chlorophenoxyacetate, 3phenylpropionate, 4-oxopentanoate (levulinate), 4,4-(ethylenedithio)pentanoate (levulinoyldithioacetal), pivaloate, adamanloate, crotonate, 4-methoxycrotonate, benzoate, pphenylbenzoate, 2,4,6-trimethylbenzoate (mesitoate), alkyl methyl caibonate, 916917
-88fiuorenylmethyl carbonate (Fmoc), alkyl ethyl carbonate, alkyl 2,2,2-trichloroethyl carbonate (Troc), 2-(trimethylsilyl)eihyl carbonate (TMSEC), 2—(phenylsulfonyl) ethyl carbonate (Psec), 2-(triphenylphosphonio) ethyl carbonate (Peoc), alkyl isobutyl carbonate, alkyl vinyl carbonate alkyl allyl carbonate, alkyl p-nitrophenyl carbonate, alkyl benzyl carbonate, alkyl p-melhoxybenzyl carbonate, alkyl 3,4—di methoxy benzyl carbonate, alkyl o-nitrobenzyl carbonate, alkyl p-nitrobenzyl carbonate, alkyl S-benzyl thiocarbonate, 4-ethoxy-lnapththyl carbonate, methyl dithiocarbonate, 2-iodobenzoate, 4-azidobutyrate, 4-nitro-4methylpentanoate, o-(dibromomethyl)benzoate, 2-formylbenzenesulfonate, 2(methylthiomethoxy)ethyl, 4-(methylthiomethoxy)butyrate, 210 (methylthiomethoxymethyl)benzoate, 2,6-dichloro-4-methylphenoxyacetate, 2,6-dichIoro-
4—{1 ,l,3,3-tetrameihylbutyl)phenoxyacetate, 2,4—bis( 1, l-dimethylpropyl)phenoxyacetate, chlorodiphenylacetate, isobutyrate, monosuccinoate, (£)-2-melhyl-2-butenoate, o(methoxyacyl)benzoate, α-naphthoate, nitrate, alkyl Ν,Ν,Ν',Ν'tetramethylphosphorodiamidate, alkyl M-phenylcarbamate, borate, dïmethylphosphinothioyl, alkyl 2,4-dinitrophenyIsulfenate, sulfate, methanesulfonate (mesylate), benzylsulfonate, and tosylate (Ts).
[00275] In certain embodiments, the substituent présent on a su 1 fur atom is an sulfur protecting group (also referred to as a thiol protecting group). Sulfur protecting groups include, but are not Iimited to, -RM, -Ν^Ύ -C(=O)SRM, -C(=O)RM, -C02R“, 20 C(=0)N(Rbb)2, -C(=NRbb)R“, -C(=NRbb)OR“, -C(=NRbb)N(Rbb)2t -S(=O)R“, -SO2RM, Si(RM)3.-P(Rcc)’. -P(R“)j. -P(=O)2R“. -P(=O)(RM)21 -P(=O)(ORCC)2, -P(=O)2N(Rbb)2, and P(=O)(NRbb)2, wherein RM, Rbb, and Rcc are as defined herein. Sulfur protecting groups are well known in the art and include those described in detail in Protecting Groups in Organic Synthesis, T. W. Greene and P. G. M. Wuts, 31 édition, John Wiley & Sons, 1999, incorporated herein by reference.
Other définitions [00276] '‘Pharmaceutically acceptable sait” refers to a sait 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 30 organic acid addition salis 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 acetîc acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartane
acid, citric acid, benzoic acid. 3-(4-hydroxybenzoyl) benzoic acid, ctnnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, l,2-ethanc-disulfonic acid, 2hy droxyethanesul fonic acid, benzenesulfonic acid, 4-chlorobcnzenesulfonic acid, 2~ naphthalenesulfonic acid, 4-toluenesulfonic acid, camphorsulfonic acid, 45 mcthylbicyclo[2.2.2]-oct-2-ene-l-carboxylic acid, glucoheptonic acid, 3-phcnylpropionic acid, trimethylacetic acid, tertiary bu ty lace tic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphlhoic acid, salicylic acid, stearic acid, muconic acid, and the like; or (2) salts formed when an acidic proton présent in the parent compound either is replaced by a métal ton, e.g., an alkali métal ion, an alkaline earth ion, or an aluminum ion; or coordinate s with an organic base such as ethanolamine, diethanolamine, tricthanolamine, Nmethylglucamine and the like. Salts further include, by way ofexample only, sodium, potassium, calcium, magnésium, 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 countcrion of an acidic functional group. Such calions are exemplified by sodium, potassium, calcium, magnésium, ammonium, tetraalkylammonium cations, and the like (sec, e.g., Berge, et al., J. Pharm. Sci. 66( l): l-79 (Jan.’77).
[00277] 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 molécule of the structural Formula indicated herein. [00278] “Prodrugs refers to compounds, Including dérivatives of the compounds of the invention, which hâve cleavable groups and become by solvolysis or under physiological conditions a compound of tbe invention that are pharmaceutically active in vivo. [00279] Solvaté” 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 physîcal association includes hydrogen bonding. Conventional solvents include water, éthanol, 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 solvatés include pharmaceutically acceptable solvatés, such as hydrates, and further include both stoichtometric solvatés and non-stoichiometrie solvatés. In certain instances the solvaté will be capable of isolation, for example when one or more solvent molécules are incorporated In the crystal lattîce of the
-90crystalline solid. “Solvaté en com passes both solution-phase and isolable solvatés. Représentative solvatés include hydrates, ethanolates and methanolates.
[00280] A “subject” to which administration is contemplated includes, but is not limited to, humans (i.e., a male or female of any âge group, e.g., a pédiatrie 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, rhésus 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 nonhuman animal. The terms human”, “patient and “subject” are used interchangeably herein.
I0 [00281] As used herein the term “enantiomerically pure” or “pure enantiomer dénotés 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 ail enantiomers or stereoisomers of the compound.
[00282] 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 al 20 least about 99% by weight R-compound and at most about I % 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.
[00283] 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 25 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.
[00284] 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 (47)

  1. WHATIS CLAIMED IS:
    1. The compound S-^hydroxyphenylM-methyl^-^-p-IORl-S-methylpyrrolidin-1yllclhoxy)phenyl)-2H-chromen-7-ol, which has the chemical structure:
    or a sait thereof.
  2. 2. The compound (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:
    or a sait thereof.
  3. 3. The compound S-^-hydroxyphenylH-methyl^-^^-iPRj-S-methylpyrrolidÎn-l yljethoxy | phenyl)-2H-chromen-7-ol, which has the chemical structure:
    OP-1038 in the form of a solvaté, hydrate, or N-oxide.
  4. 5 4. The compound (2S)-3-(4-hydroxyphenyl)-4-methyl-2-(4-(2-[(3R)-3methylpyrrolidin-l-yl]ethoxy]phenyl)-2H-chromen-7-ol, which has the chemical structure:
    OP-1074
    I0 în the form of a solvaté, hydrate, or N-oxide,
    5. The compound of claim I wherein the sait is pharmaceutically acceptable.
  5. 6. The compound of claim 2 wherein the sait is pharmaceutically acceptable.
    I5
  6. 7. The compound of claim 1 which is not in the form of a sait.
  7. 8. The compound of claim 2 which is not in the form of a sait
  8. 9. A compound of the structure:
    wherein R1 and R2 are independently either:
    -93(ii) R9 which is independently selected from H, halogen (Cl, Br, I or F), natural or nonnaturally occurring amino acid (bound through either the OC(O)- or C(O)O- (an ester) or the amino (through either -C(O)-N- or -N-C(O)- (an amide linkage)), R10, -OR10, or-SR10 where R10 is -C(=O)Rcl, -C(=O)ORcl, -C(=O)SRcl, -C(=O)N(Rcl):; 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(=O)2Rci, -S(=O)iORcl, -S(=O)RCI, -S(=O)ORC1, -P(=O)3RC1, -P(=O)IORCI, -P(=O)(ORclh, -P(=O)(RCI)2, or-P(Rcl)(ORcl); 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 -NH- on administration);
    and RCI 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 RCI groups are joined to form an substituted or unsubstituted heterocyclic ring.
    or its pharmaceutically acceptable sait.
  9. 10. A compound of the structure:
    wherein R1 and R2 are independently either:
    -94R9 which is independently selected from H, halogen (Cl, Br, I or F), natural or nonnaturally occurring amino acid (bound through either the OC(O)- or C(O)O- (an ester) or the amino (through either -C(O)-N- or -N-C(O)- (an amide lînkage)), Rl°, -OR10, or-S R10
    5 where R10 is -C(=O)Rcl, -C(=O)ORcl, -C(=O)SRcl, -C(=O)N(Rcl)?; 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;
    10 -S(=O)3RCI, -S(=O)2ORci, -S(=O)Rci, *S(=O)ORCI, -P(=O):RCI,
    -P(=O)2ORC', -P(=O)(ORcl)i, -P(»O)(Rclh, or-P(Rcl)(ORc‘); 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 15 group (to produce -NH- on administration);
    and Ra 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 20 substituted or unsubstituted heteroaryl, or two RCI groups are joined to form an substituted or unsubstituted heterocyclic ring.
    or its pharmaceutically acceptable sait
  10. 11. The compound of claim 9, wherein R| and Rj are -OR9.
    25
  11. 12. The compound of claim 10, wherein R| and R, are -OR9.
  12. 13. The compound of claim 9, wherein R| is hydrogen.
  13. 14. The compound of claim 10, wherein Ri is hydrogen.
  14. 15. The compound of claim 11, wherein -OR9 is C(O)alkyI.
    J 6. The compound of claim 12, wherein -OR9 is C(O)alkyl
    30 17. The compound of claim 11, wherein -OR9 is C(O)aryl.
  15. 18. The compound of claim 12, wherein -OR9 is C(O)aryl.
  16. 19. The compound of claim 11, wherein -OR9 is OPOjHj.
  17. 20. The compound of claim 12, wherein -OR9 is OPOjHj.
  18. 21. The compound of claim 9, selected from
  19. 22. The compound of clai m 10, se lected from
  20. 23. A pharmaceutical composition comprising a pharmaceutically effective amount of the compound of claim l in a pharmaceutically acceptable carrier.
  21. 24. A pharmaceutical composition comprising a pharmaceutically effective amount of the
    IO compound of claim 2 in a pharmaceutically acceptable carrier.
  22. 25. A pharmaceutical composition comprising a pharmaceutically effective amount of the compound of claim 3,5,7,9, 11,13, 15,17, or I9 in a pharmaceutically acceptable carrier.
  23. 26. A pharmaceutical composition comprising a pharmaceutically effective amount of the
    15 compound of claim 4,6,8,10, I2, 14,16, 18 or 20 in apharmaceutically acceptable carrier.
  24. 27. The pharmaceutical composition of Claim 21 wherein the carrier is suitable for oral delivery.
  25. 28. The pharmaceutical composition of claim 22 wherein the carrier is suitable for oral
    20 delivery.
  26. 29. Use of the compound of claim l, optionally in a pharmaceutically acceptable carrier in the manufacture of a pharmaceutical composition for treating a disorder mediated by the estrogen receptor in a patient, said pharmaceutical composition being for administration to the patient.
    25
  27. 30. Use of the compound of claim 2, optionally in a pharmaceutically acceptable carrier in the manufacture of a pharmaceutical composition for treating a disorder mediated
    -98by the estrogen receptor tn a patient, said pharmaceutical composition being for administration to the patient
  28. 31. Use ofthe compoundofclaim 3,5,7,9,11,13,15,17, or 19, optionally in a pharmaceutically acceptable carrier in the manufacture of a pharmaceutical composition for treating a disorder mediated by the estrogen receptor in a patient, said pharmaceutical composition for administration to the patient.
  29. 32. Use of the compound of claim 4,6,8,10, 12,14,16,18 or 20, optionally in a pharmaceutically acceptable carrier in the manufacture of a pharmaceutical composition for treating a disorder mediated by the estrogen receptor in a patient, said pharmaceutical composition being for administration to the patient.
  30. 33. The use of claim 27, wherein the disorder is breast cancer.
  31. 34. The use of claim 28, wherein the disorder is breast cancer.
  32. 35. The use of claim 29, wherein the disorder is breast cancer.
  33. 36. The use of claim 30, wherein the disorder is breast cancer.
  34. 37. The use of claim 27, wherein the disorder is selected from the group consisting of ovarian, endométrial, or vaginal cancer, endometriosis or lung cancer.
  35. 38. The use of claim 28, wherein the disorder is selected from the group consisting of ovarian, endométrial, or vaginal cancer, endometriosis or lung cancer.
  36. 39. The compound of claim I for use in medical therapy.
  37. 40. The compound of claim 2 for use in medical iherapy.
  38. 41. The compound of claim 3,5,7,9, II, 13, 15,17, or 19, for use in medical therapy.
  39. 42. The compound of claim 4,6,8, 10,12, 14, 16, 18 or 20, for use in medical therapy.
  40. 43. The use of claim 29, wherein said administration further comprises administration of lhe compound in combination or alternation with another anti-cancer agent for the treatment of cancer.
  41. 44. The use of claim 30, wherein said administration further comprises administration of the compound in combination or alternation with another anti-cancer agent for the treatment of cancer.
  42. 45. The use of claim 31, wherein said administration further comprises administration of the compound tn combination or alternation with another anti-cancer agent for the treatment of cancer.
  43. 46. The use of claim 32, wherein said administration further comprises administration of the compound in combination or alternation with another anti-cancer agent for the treatment of cancer.
  44. 47. The use of claim 29, wherein said administration further comprises administration of the compound in combination or alternation with estrogen or a partial estrogen receptor angatonist for the treatment of a postmenopausal disorder.
  45. 48. The use of claim 30, wherein said administration further comprises administration of
    5 the compound in combination or alternation with estrogen or a partial estrogen receptor angatonist for the treatment of a postmenopausal disorder.
  46. 49. The use ofclaim3l, wherein said administration further comprises administration of the compound in combination or alternation with estrogen or a partial estrogen receptor angatonist for the treatment of a postmenopausal disorder another anti-cancer
    10 agent.
  47. 50. The use of claim 32, wherein said administration further comprises administration of the compound in combination or alternation with estrogen or a partial estrogen receptor angatonist for the treatment of a postmenopausal disorder.
OA1201400252 2011-12-16 2012-12-17 Novel benzopyran compounds, compositions and uses thereof. OA16917A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
US61/576,890 2011-12-16

Publications (1)

Publication Number Publication Date
OA16917A true OA16917A (en) 2016-01-25

Family

ID=

Similar Documents

Publication Publication Date Title
US9018244B2 (en) Benzopyran compounds, compositions and uses thereof
WO2014203132A1 (en) Substituted benzopyran compounds, compositions and uses thereof
EP3411034B1 (en) Selective estrogen receptor degraders and uses thereof
CN107485612B (en) Imidazopyridine compounds and uses thereof
CA2848212C (en) Neuroactive steroids, compositions, and uses thereof
AU2014369834B2 (en) Cortistatin analogues and syntheses and uses thereof
BR112018006121B1 (en) TETRAHYDRO-1H-PYRIDO[3,4-b]INDOL COMPOUNDS, COMPOSITIONS COMPRISING SAID COMPOUNDS AND USES THEREOF
TW201511755A (en) Combinations of benzopyran compounds, compositions and uses thereof
AU2017217536A1 (en) Inhibitor of indoleamine-2,3-dioxygenase (IDO)
JP7348665B2 (en) Substituted benzothiophene analogs as selective estrogen receptor degraders
JP2014055156A (en) Compound and method for inhibiting interaction of bcl protein with binding partner
CA3143545A1 (en) Compounds for treating cns disorders
JP2023501306A (en) Benzothiophene derivative regulators, their preparation and use
CN112047944A (en) Selective kinase inhibition compound and application thereof
EP4083032A1 (en) Pd-l1 antagonist compound
CA2995161A1 (en) 2,3,4,5-tetrahydropyridin-6-amine derivatives
OA16917A (en) Novel benzopyran compounds, compositions and uses thereof.
EP3092220A1 (en) Vitamin c prodrugs and uses thereof
RU2823231C1 (en) Pd-l1 antagonist compound
TWI846678B (en) Caffeic acid derivatives and uses thereof
EP3947382A1 (en) Compounds for uses in pharmacological induction of hbf for treatment of sickle cell disease and beta-thalassemia
CN116836216A (en) Fulvestrant derivative, preparation method and medical application thereof
TW202116760A (en) Four-membered ring derivative regulator, preparation method and application thereof
WO2020033377A1 (en) Histone demethylase 5 inhibitors and uses thereof