EP2131826A2 - 17beta-hydroxysteroid-dehydrogenase-typ1-inhibitoren zur behandlung hormonabhängiger erkrankungen - Google Patents

17beta-hydroxysteroid-dehydrogenase-typ1-inhibitoren zur behandlung hormonabhängiger erkrankungen

Info

Publication number
EP2131826A2
EP2131826A2 EP08735534A EP08735534A EP2131826A2 EP 2131826 A2 EP2131826 A2 EP 2131826A2 EP 08735534 A EP08735534 A EP 08735534A EP 08735534 A EP08735534 A EP 08735534A EP 2131826 A2 EP2131826 A2 EP 2131826A2
Authority
EP
European Patent Office
Prior art keywords
hydroxy
hydroxyphenyl
naphthyl
phenyl
nhso
Prior art date
Legal status (The legal status 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 status listed.)
Withdrawn
Application number
EP08735534A
Other languages
German (de)
English (en)
French (fr)
Inventor
Rolf Hartmann
Martin Frotscher
Sandrine Oberwinkler
Erika Ziegler
Josef Messinger
Heinrich-Hubert Thole
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Universitaet des Saarlandes
Original Assignee
Universitaet des Saarlandes
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 Universitaet des Saarlandes filed Critical Universitaet des Saarlandes
Publication of EP2131826A2 publication Critical patent/EP2131826A2/de
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/01Hydrocarbons
    • A61K31/015Hydrocarbons carbocyclic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/498Pyrazines or piperazines ortho- and peri-condensed with carbocyclic ring systems, e.g. quinoxaline, phenazine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones

Definitions

  • the invention relates to the use of non-steroidal 17beta-hydroxysteroid dehydrogenase Typl (17betaHSD1) inhibitors for the treatment and prophylaxis of hormone-dependent, in particular estrogen-dependent, diseases. Furthermore, suitable inhibitors and a process for their preparation are provided.
  • Steroid hormones are important chemical carriers of information for the long-term and global control of cellular functions. They control the growth as well as the differentiation and function of many organs. On the other hand, they can also have negative effects and the formation and proliferation of diseases in the organism such. For example, mammalian and prostatic carcinomas (Deroo, BJ et al., J. Clin Invest, 116: 561-570 (2006); Fernandez, SV et al., Int J. Cancer, 118: 1862-1868 (2006) )). The biosynthesis of steroids takes place in the testes or in the ovaries, where sex hormones are formed. In the adrenal glands, the display of gluco- and mineralocorticoids also takes place.
  • 17ß hydroxysteroid dehydrogenase type 1 (17ß-HSDl), which catalyzes the conversion of estrone to estradiol, is increasingly found in endomeric tissue and breast cancer cells, while at the same time there is a deficiency of 17ß-HSD type 2, which catalyzes the reverse reaction occurs (Bulun, S. E. et al., J. Steroid Biochem. Biol., 79: 19-25 (2001); Miyoshi, Y. et al., Int. J. Cancer, 94: 685-689 (2001)).
  • a major class of steroid hormones is formed by the estrogens, the female sex hormones whose biosynthesis v. a. runs in the ovaries and reaches its maximum immediately before ovulation. Estrogens also occur in adipose tissue, muscles and some tumors. The main tasks include a genital effect, d. h the development and maintenance of female sexual characteristics as well as an extragenital lipid-anabolic effect leading to the development of subcutaneous adipose tissue. In addition, they are involved in the development and proliferation of estrogen-dependent diseases such.
  • B. Endometriosis, Endometrial Carcinoma, Adenomyosis, and Breast Cancer (Bulun, SE et al., J.
  • estradiol (E 2 ), which is produced in premenopausal women mainly in the ovaries. It reaches the target tissues by endocrine route, where it exerts its effect through interaction with the estrogen receptor (ER) ⁇ . After menopause, the plasma E 2 level drops to 1/10 of the estradiol level in premenopausal women (Santner, SJ et al., J. Clin Endocrinol, Metab., 59: 29-33 (1984)). E 2 is now mainly in the peripheral tissue, eg. As breast tissue, endometrium, adipose tissue, skin from inactive precursors such.
  • peripheral tissue eg. As breast tissue, endometrium, adipose tissue, skin from inactive precursors such.
  • estrone sulfate egg S
  • DHEA dehydroepiandrosterone
  • DHEA-S DHEA-S
  • E 2 its concentration in the peripheral tissue, especially in estrogen-dependent diseases, is higher than in healthy tissue.
  • the growth of many breast cancer cell lines is stimulated by a locally increased estradiol concentration.
  • endometriosis is an estrogen-dependent disease affecting approximately 5-10% of all women of childbearing age (Kitawaki, J., Journal of Steroid Biochemistry & Molecular Biology, 83: 149-155 (2003)). 35 - 50% of women with abdominal pain u./o. Sterility has signs of endometriosis (Urdl, W., J. Reproductive Medicine Endocrinol., 3: 24-30 (2006)).
  • This disease is defined as histologically proven ectopic endometrial glandular and stromal tissue.
  • This chronic disease which is prone to recurrences, leads to pain of varying intensity and varying character as well as potentially to sterility if it is given a corresponding form.
  • Three macroscopic conditions are distinguished: peritoneal endometriosis, retroperitoneal deep infiltrating endometriosis including adenomyosis uteri and cystic ovarian endometriosis.
  • peritoneal endometriosis retroperitoneal deep infiltrating endometriosis including adenomyosis uteri and cystic ovarian endometriosis.
  • There are several explanatory theories for the pathogenesis of endometriosis eg. For example, the metaplasia theory, the transplantation theory and the theory of autotraumatization of the uterus by Leyendecker (Leyendecker, G. e
  • pluripotent coelomic epithelium under certain conditions should also have the ability in adults to differentiate and to form Endometrioseherde.
  • This theory is supported by the observation that in some women with missing uterus and gyna-atresia severe endometriosis can occur. Even in men treated for prostate cancer with high estrogen doses, endometriosis was detected in individual cases. According to Sampson (Halme, J.
  • Cytokines, enzymes, growth factors eg matrix metalloproteinases
  • Increased autonomic cycle-independent estrogen production and activity, as well as decreased estrogen inactivation, are a typical feature of endometriotic tissue.
  • This increased local estrogen production and activity is characterized by a significant over-expression of aromatase, normal expression of 17ß-HSD1, and decreased endogenous Inactivation of the potent E2 due to a deficiency of 17ß-HSD2 (Bulun, SE et al., J. Steroid Biochem. Mol. Biol., 79: 19-25 (2001); Kitawaki, J., Journal of Steroid Biochemistry & Molecular Biology, 83: 149-155 (2003); Karaer, O.
  • the polymorphous symptoms caused by endometriosis include any pelvic pain symptoms, low back pain, dyspareunia, dysuria and defecation complaints.
  • One of the most commonly used therapeutic measures in endometriosis is the surgical removal of endometrial implants (Urdl, W., J. Reproductive Medicine Endocrinol., 3: 24-30 (2006)).
  • the drug treatment remains in need of improvement despite new treatment concepts.
  • the purely symptomatic treatment of dysmenorrhea takes place by means of non-steroidal anti-inflammatory drugs (NSAID) such.
  • NSAID non-steroidal anti-inflammatory drugs
  • the causal drug therapy is based on an estrogen withdrawal with associated variable side effects and in general contraceptive character.
  • the gestagens with their antiestrogenic and antiproliferative effect on the endometrium play a major therapeutic role.
  • danazol is decreasing due to its androgenic side-effect profile with potential increases in weight, hirsutism and acne.
  • GnRH analogues include hot flashes, amenorrhoea, libido loss and osteoporosis, the latter mainly as part of long-term treatment.
  • Another therapeutic approach is the steroidal and nonsteroidal aromatase inhibitors.
  • Goserelin bind in the target organ, the pituitary gland, to specific membrane-bound receptors, which leads to an increased secretion of FSH and LH.
  • FSH and LH these two hormones, in turn, lead to a diminution of the GnRH receptors in a negative feedback in the pituitary cells.
  • the resulting desensitization of the pituitary cells with respect to GnRH leads to secretion inhibition of FSH and LH, so that the steroid hormone control loop is interrupted.
  • Side effects of these therapeutics include hot flashes, sweats and osteoporosis.
  • Another therapeutic option is the use of antiestrogens, antagonists of the estrogen receptor.
  • SERM selective estrogen receptor modulators
  • the enzymatically catalyzed estrogen biosynthesis can also be influenced by selective enzyme inhibitors.
  • the enzyme aromatase which converts C19 steroids into C18 steroids, was one of the first targets for lowering estradiol levels.
  • This enzyme complex which belongs to the cytochrome P-450 enzymes, catalyzes the aromatization of the androgenic A ring to form estrones. The methyl group in position 10 of the steroid is split off.
  • the first aromatase inhibitor used to treat breast cancer was aminogluthetimide. However, aminogluthetimide affects several enzymes of the cytochrome P-450 superfamily and thus inhibits a number of other biochemical transformations.
  • the compound intervenes so strongly in the adrenal steroid production that both a gluco- and a mineralocorticoid substitution may be necessary.
  • aromatase inhibitors on the market that can be subdivided into steroidal and nonsteroidal compounds.
  • steroidal inhibitors z. B. Exemestane, which has a positive effect on bone density, which is associated with the affinity for the androgen receptor (Goss, PE et al., Clin. Cancer Res., 10: 5717-5723 (2004)).
  • this type of compounds are irreversible inhibitors, which also have a greater number of side effects such. Eg hot flashes, nausea, tiredness.
  • non-steroidal compounds that are used therapeutically z. B. letrozole.
  • the advantage of these compounds is their lower side effects, they do not cause uterine hypertrophy, but they have no positive effect on bone density and lead to an increase in LDL (low-density lipoprotein), cholesterol and triglycerides (Goss, PE et al. Clin. Cancer Res., 10: 5717-5723 (2004); Nunez, NP et al., Clin. Cancer Res., 10: 5375-5380 (2004)).
  • Aromatase inhibitors are currently used primarily as second-line therapeutics. Meanwhile, in clinical studies, the equivalence or even superiority of aromatase inhibitors against SERM such.
  • the sulfatase pathway is the pathway to the formation of estrone / estradiol by the enzyme steroid sulfatase, an enzyme that catalyzes the conversion of estrone sulfate and DHEA-S to estrone and DHEA. In this way, 10 times more estrone is produced in the target tissue than in the aromatase pathway (Santner, SJ et al., J. Clin Endocrinol, Metab., 59: 29-33 (1984)). The estrone is then reduced to E2, the most potent estrogen, by the enzyme 17 ⁇ -HSD1.
  • the steroid sulphatase and 17 ⁇ -HSD1 are new targets in the fight against estrogen-dependent diseases, in particular for the development of therapeutic agents for breast cancers (Pasqualini, JR, Biochim, Biophys Acta., 1654: 123-143 (2004)).
  • Hydroxysteroid dehydrogenases can be divided into different classes.
  • the LL ⁇ -HSD modulate the activity of glucocorticoids, 3 ⁇ -HSD catalyzes the reaction of ⁇ 5-3 ⁇ -hydroxysteroids (DHEA or 5-androstene-3 ⁇ , 17 ⁇ -diol) to ⁇ 5-3 ⁇ -ketosteroids (androstenedione or testosterone).
  • 17 ⁇ -HSD converts the less active 17-ketosteroids to the corresponding highly active 17-hydroxy compounds (androstenedione to testosterone and egg to E 2 ) or vice versa (Payne, AH et al., Endocr. Rev., 25: 947-970 (2004 Peltoketo, H. et al., J.
  • HSD HSD play a crucial role in the activation as well as in the inactivation of steroid hormones.
  • they alter the potency of the sex hormones e.g. B.
  • E 1 is converted by means of 17-HSDL to the highly potent E 2
  • E 2 by means of 17-HSD2 is converted to the less potent egg, 17beta-HSD2 inactivated E 2 during 17-HSDL egg activated.
  • the amino acid identity between the different 17 ⁇ -HSD is very low at 20-30% (Luu-The, V., J. Steroid Biochem., Mol. Biol., 76: 143-151 (2001)) and is membrane bound or soluble enzymes.
  • the X-ray structure of 6 human subtypes is known (1,4,5,10,11,14) (Ghosh, D. et al., Structure, 3: 503-513 (1995); Kissinger, CR et al., J. Biol., 342: 943-952 (2004); Zhou, M. et al., Acta Crystallogr. D. Biol. Crystallogr., 58: 1048-1050 (2002).
  • 17ß-HSD is NAD (H) and NADP (H) -dependent enzymes, which play a crucial role in human hormonal regulation, differ in their tissue distribution, catalytic preference (oxidation or reduction), substrate specificity, and subcellular localization, and the same HSD It is likely that all 17ß-HSDs are expressed in the different estrogen-dependent tissues, but at different concentrations In diseased tissue, the ratio between the different subtypes is altered compared to healthy tissue, with some subtypes overexpressed while others may be missing This may result in an increase or decrease in the concentration of the corresponding steroid. Thus, 17ß-HSD plays an extremely important role in the regulation of the activity of the sex hormones. Furthermore, they are involved in the development of estrogen-sensitive diseases such.
  • ovarian, uterine and endometrial carcinomas and androgen-dependent diseases such as prostate cancer, benign prostatic hyperplasia, acne, hirsutism, etc.
  • 17 ⁇ -HSD are also involved in the development of other diseases, e.g. Pseudohermaphrodism (17 ⁇ -HSD3 (Geissler, WM et al., Nat. Genet., 7: 34-39 (1994)), bifunctional enzyme deficiency (17 ⁇ -HSD4 (van Grunsven, EG et al., Proc. Natl.
  • the best characterized member of the 17ß-HSD is the type 1 17ß-HSD.
  • the 17ß-HSD1 is an enzyme of the SDR family, which is also known as human placenta. Taestradioldehydrogenase (Gangloff, A. et al., Biochem J., 356: 269-276 (2001); Jornvall, H. et al., Biochemistry, 34: 6003-6013 (1995)).
  • the name assigned by the Enzyme Commission is EC1.1.1.62. Engel and coworkers (Langer, L.J.
  • the 17ß-HSD1 is encoded by a 3.2 kb gene, which consists of 6 exons and 5 introns and is converted into a 2.2 kb transcript (Luu-The, V., J. Steroid Biochem., Mol. Biol., 76 : 143-151 (2001); Labrie, F. et al., J. Mol. Endocrinol., 25: 1-16 (2000)). It is built up from 327 amino acids. The molecular weight of the monomer is 34.9 kDa. (Penning, TM, Endocr.
  • 17 ⁇ -HSD1 is expressed in placenta, liver, ovary, endometrium, prostate, peripheral tissue such as, e.g. B. adipose tissue and breast cancer cells (Penning, TM, Endo Rev., 18: 281-305 (1997)). It was isolated for the first time from human placenta (Jarabak, J. et al., J. Biol. Chem., 237: 345-357 (1962)). The main task of the 17ß-HSDl is the conversion of the less active estrone into the highly potent estradiol.
  • DHEA dehydroepiandrosterone
  • 5-androstene-3 ⁇ , 17 ⁇ -diol an estrogen-producing androgen
  • E 2 the enzyme catalyzes reduction and oxidation between egg and E 2 , while under physiological conditions it catalyzes only the reduction.
  • bisubstrate reactions proceed by a random catalytic mechanism, ie either steroid or cofactor first binds to the enzyme (Betz, G., J. Biol. Chem., 246: 2063-2068 (1971)). Also postulated is a catalytic mechanism in which the cofactor first binds to the enzyme (Neugebauer, A. et al., Bioorg. Med. Chem., Submitted (2005)).
  • the enzyme consists of a substrate binding site and a channel that opens into the cofactor binding site.
  • the substrate binding site is a hydrophobic one Tunnel, which has a high degree of complementarity to the steroid.
  • the 3-hydroxy and 17-hydroxy groups in the steroid form four hydrogen bonds to the amino acid residues His221, Glu282, Serl42 and Tyrl55.
  • the hydrophobic van der Waals interactions appear to form the major interactions with the steroid while the hydrogen bonds are responsible for the steroid specificity to the enzyme (Labrie, F. et al., Steroids, 62: 148-158 (1997)).
  • the cofactor binding site also includes the Rossmann fold, a region composed of ⁇ -helices and ⁇ -sheets ( ⁇ - ⁇ - ⁇ - ⁇ - ⁇ ) 2 , a commonly occurring motif Gly-Xaa-Xaa Xaa-Gly-Xaa-Gly and a nonsense region Tyr-Xaa-Xaa-Xaa-Lys within the active site.
  • Important for the activity is a catalytic tetrad, consisting of Tyrl55- Lysl59-Serl42-Asnl l4, which stabilize the hydride in the steroid and the ribose in nicotinamide.
  • the 17ß-HSDl coding gene is linked to the mutations very susceptible and inheritable gene for breast and ovarian carcinoma, the BRCAl gene on chromosome 17qll-q21 (Labrie, F. et al., J. Mol. Endocrinol., 25: 1-16 (2000)). It has been shown that the activity of 17ß-HSDl is higher in endometriotic tissue and breast cancer cells than in healthy tissue, which entails high intracellular estradiol levels, which in turn cause proliferation and differentiation of the diseased tissue (Bulun, SE et al., J. Steroid Biochem Mol. Biol., 79: 19-25 (2001); Miyoshi, Y. et al., Int.J.
  • Estradiol or estrone moiety that interacts with and binds to the substrate binding site
  • a disadvantage of these steroidal compounds may be a low selectivity. With steroids, there is a risk that the compounds also attack other enzymes of steroid biosynthesis, resulting in side effects. In addition, because of their steroidal structure, they may have an affinity for steroid receptors and act as agonists or antagonists.
  • Thiophene-pyrimidinones have been investigated as further nonsteroidal inhibitors (US2005 / 038053; Messinger, J. et al., Mol. Cell. Endocrinol., 248: 192-198 (2006); WO2004 / 110459).
  • 6- (3-hydroxyphenyl) naphthalen-2-ol, 6- (2-hydroxyphenyl) naphthalen-2-ol and 6-phenylnaphthalen-2-ol are estrogenic as compounds Activity (Cassebaum, H., Chemische Berichte, 90: 2876-2888 (1957); WO2005 / 014551; WO2003 / 051805; Mewshaw, RE et al., J. Med. Chem., 48: 3953-3979 (2005); Shunk, C. et al., J. Am. Chem Soc, 71: 3946-3950 (1949); Tao, B. et al., Tetrahedron Letters, 43: 4955-4957 (2002); D. et al., J. Chem. Soc., 374-383 (1940).
  • R 1 is H, halogen, OH, CN, COOH, alkyl, alkoxy, alkylsulfanyl, aryl, heteroaryl, arylsulfanyl, -NHSO 2 R ', -R "-NHSO 2 R', -SO 2 NHR ', -R" - SO 2 NHR ', -NHCOR', -CONHR ', -
  • R 3 is H, halogen, OH, CN, COOH, alkyl, alkoxy, alkylsulfanyl, aryl, heteroaryl, arylsulfanyl, -NHSO 2 R ', -R "-NHSO 2 R', -SO 2 NHR ', -R" -SO 2 NHR ', -NHCOR', -CONHR ', -
  • R 4 is H or OH
  • R 5 is H, halogen, OH, CN, COOH, alkyl, alkoxy, alkylsulfanyl, aryl, heteroaryl, arylsulfanyl, -NHSO 2 R ', -R "-NHSO 2 R', -SO 2 NHR ', -R" -SO 2 NHR ', -NHCOR', -CONHR ', -
  • R 6 is H, halogen, OH, CN, COOH, alkyl, alkoxy, alkylsulfanyl, aryl, heteroaryl, arylsulfanyl, -NHSO 2 R ', -R "-NHSO 2 R', -SO 2 NHR ', -R" -SO 2 NHR ', -NHCOR', -CONHR ', - R "- N HCOR', -R" -CONHR ', -COOR', -OOCR ', -R "-COOR', -R" -OOCR ' , -CHNR ', -SO 2 R' or -SOR 'is,
  • R 7 is H, halogen, OH, CN, COOH, alkyl, alkoxy, alkylsulfanyl, aryl, heteroaryl, arylsulfanyl, -NHSO 2 R ', -R "-NHSO 2 R', -SO 2 NHR ', -R" -SO 2 NHR ', -NHCOR', -CONHR ', -
  • R "-N HCOR ', -R"-CONHR', -COOR ', -OOCR', -R "-COOR ', -R"-OOCR', -CHNR ', -SO 2 R' or -SOR ' , or if W -N is absent, R 8 is H, halogen, OH, CN, COOH, alkyl, alkoxy, alkylsulfanyl, aryl, heteroaryl, arylsulfanyl, -NHSO 2 R ', -R "-NHSO 2 R', -SO 2 NHR ', -R" -SO 2 NHR ', -NHCOR', -CONHR ', - R "- N HCOR', -R" -CONHR ', -COOR', -OOCR ', -R "-COOR', -R” -OOCR ', -CHNR ',
  • Rn is independently selected from H and lower alkyl or two Rn together with the linking N atom form a 5- to 7-membered saturated heterocycle, and pharmacologically acceptable salts thereof, for the treatment and prophylaxis of hormone-dependent diseases;
  • R 5 , R 6 and R 9 are H, R 4 is OH or H, then one of R 8 and Ri 0 is not OH and the other is H;
  • R 6 and R 9 are H, R 2 is COOH and R 4 is OH, then one of R 8 and R 10 is not OH and the other is H,
  • R 3 , R 4 , R 5 , R 6 and R 9 are H, then one of the radicals R 8 and Ri 0 is not
  • R, Ri, R 3, R 4, R 5, Re and R 9 are H, then one of the radicals R 8 and R 0 is not OH and the other is H;
  • a method for the treatment and prophylaxis of hormone-dependent disorders in a patient comprising administering to the patient a compound having the structure (I) as defined in (1) or (2).
  • the compound having the structure (I) as defined in (1) or (2), and the drug or the pharmaceutical composition as defined in (3) and the method as defined in (5) are according to the invention for the treatment and prophylaxis hormone-dependent, in particular estrogen-dependent diseases suitable.
  • they are useful in the treatment and prophylaxis of diseases in which modulation of estradiol levels is required, such as the treatment and prophylaxis of endometriosis, endometrial carcinoma, adenomyosis, breast cancer and ovarian carcinoma.
  • Alkyl radicals and "alkoxy radicals” in the meaning of the invention may be straight-chain, branched or cyclic and be saturated or (partially) unsaturated , Preferred alkyl radicals and alkoxy radicals are saturated or have one or more double and / or triple bonds.
  • straight-chain or branched alkyl radicals those having 1 to 10 C atoms, especially those having 1 to 6 C atoms, very particularly those having 1 to 3 C atoms, are preferred.
  • “Lower alkyl radicals” and “lower alkoxy radicals” for the purposes of the invention are straight-chain, branched or cyclic saturated lower alkyl radicals and lower alkoxy radicals or those having a double or triple bond.
  • straight-chain ones those having 1 to 6 C atoms, in particular having 1 to 3 C atoms, are particularly preferred.
  • those with 3 to 8 C atoms are particularly preferred.
  • Aryls for the purposes of the present invention include mono-, bi- and tricyclic aryl radicals having 3 to 18 ring atoms, which may optionally be fused with one or more saturated rings. Particularly preferred are anthracenyl, dihydro-naphthyl, fluorenyl, hydrindanyl, indanyl, indenyl, naphthyl, naphthenyl, phenanthrenyl, phenyl and tetralinyl.
  • heteroaryl radicals are mono- or bicyclic heteroaryl radicals having 3 to 12 ring atoms, which preferably have 1 to 5 heteroatoms selected from nitrogen, oxygen and sulfur and which may be fused with one or more saturated rings ,
  • the preferred nitrogen-containing monocyclic and bicyclic heteroaryls include benzimidazolyl, benzene zothiazolyl, benzoxazolyl, quinazolinyl, quinolyl, quinoxalinyl, cinnolinyl, dihydroindolyl, dihydroisoindolyl, dihydropyranyl, dithiazolyl, furyl, homopiperidinyl, imidazolidinyl, imidazolinyl, imidazolyl, indazolyl, indolyl, isoquinolyl, isoindolyl, isothiazolidinyl, isothiazolyl, isoxazolidinyl, isox
  • mono- or bicyclic heteroaryl radicals having from 5 to 10 ring atoms, which preferably have from 1 to 3 nitrogen atoms, very particular preference to oxazolyl, imidazolyl, pyridyl and pyrimidyl.
  • “5- to 7-membered saturated heterocycles” in the context of this invention include the saturated equivalents of the heteroaryl compounds mentioned above, with morpholine, piperazine and piperidine being particularly preferred.
  • “Alkylenes”, “lower alkylenes”, “arylenes” and “heteroarylenes” in the context of this invention are the bivalent equivalents of the alkyl, lower alkyl, aryl and heteroaryl radicals defined above.
  • “Halogen” includes fluorine, chlorine, bromine and iodine.
  • Halogenated or “optionally halogenated” radicals for the purposes of the present invention comprise all radicals in which one to all the H atoms have been replaced by the abovementioned halogen atoms, including combinations of these halogen atoms.
  • “Pharmaceutically acceptable salts” for the purposes of the present invention thereby include salts of the compounds with organic acids (such as lactic acid, acetic acid, amino acid, oxalic acid, etc.), inorganic acids (such as HCl, HBr, phosphoric acid, etc.) and, if the compounds have acid substituents , also with organic or inorganic bases. Preferred are salts with HCl.
  • “Pharmacologically suitable carriers” in the sense of the present invention are selected by the person skilled in the art as a function of the desired administration form.
  • the preferred embodiments of the compound of the formula (I) are shown in (1) and (2).
  • R is H, halogen, lower alkyl, lower alkoxy or lower alkylsulfanyl.
  • R 3 and R 5 are H, halogen, OH, CN, COOH, alkyl, alkoxy, alkylsulfanyl, aryl, heteroaryl, arylsulfanyl, -NHSO 2 R ', -R "-NHSO 2 R ', -SO 2 NHR', -R "-SO 2 NHR ', -NHCOR', -CONHR ', -R"-NHCOR', -R "-CONHR ', -COOR', -OOCR ', -R "-COOR ', -R"-OOCR', -CHNR ', -SO 2 R' or -SOR ', wherein R' and R "have the meaning given above and as well as the aryl, arylene, heteroaryl and heteroarylene having 1 to 3 radicals R '' may be substituted and R "'has the meaning given above.
  • R 3 and R 5 the other of the two radicals is preferably, as well as the residues Ri, R 2, R 6 and R 7, selected from H, halogen, OH, CN, COOH, lower alkyl, lower alkoxy, lower alkylsulfanyl, aryl, heteroaryl, arylsulfanyl, -NHSO 2 R ', -SO 2 NHR', -NHCOR ', - CONHR', -COOR ', -OOCR ', CHNR', -SO 2 R 'and -SOR', wherein R 'is lower alkyl, phenyl or pyridinyl.
  • R 8 is H, halogen, hydroxy, CN, COOH, lower alkyl, lower alkoxy, lower alkylsulfanyl, aryl, heteroaryl, arylsulfanyl, -NHSO 2 R ', -SO 2 NHR', -NHCOR ', -CONHR ', -COOR', -OOCR ', -CHNR', -SO 2 R 'or -SOR', wherein R 'is lower alkyl, phenyl or pyridinyl.
  • R 3 is selected from H, halogen, OH, CN, COOH, alkyl, alkoxy, alkylsulfanyl, aryl, heteroaryl, arylsulfanyl, -NHSO 2 R ', -R "-NHSO 2 R' , -SO 2 NHR ', -R "-SO 2 NHR', -NHCOR ', -CONHR', -R" - N HCOR ', -R "-CONHR', -COOR ', -OOCR', -R” Is -COOR ', -R "-OOCR', -CHNR ', -SO 2 R' and -SOR ', preferably aryl, heteroaryl, arylsulfanyl, -NHSO 2 R', -R" -NHSO 2 R ', -SO 2 NHR ', -R "-SO 2 N
  • R 1, R 2 , R 5 , R 6 and R 7 are independently selected from H, halogen, hydroxyl, CN, COOH, lower alkyl, lower alkoxy, lower alkylsulfanyl, phenyl, pyridyl, phenylsulfato nyl, -NHSO 2 R ', -SO 2 NHR', -NHCOR ', -CONHR', SO 2 R 'and -SOR', where R 'is lower alkyl, phenyl or pyridinyl.
  • radicals mentioned independently of one another are H, F, Cl, CN, lower alkyl or lower alkoxy.
  • R 8 is H, halogen, hydroxyl, CN, COOH, lower alkyl, lower alkoxy, lower alkylsulfanyl, phenyl, pyridyl, phenylsulfanyl, -NHSO 2 R ', -SO 2 NHR', -NHCOR ', -CONHR', -SO 2 R 'or -SOR', where R 'is lower alkyl, phenyl or pyridinyl.
  • R 8 is H, F, Cl, OH, CN, COOH, lower alkyl or lower alkoxy.
  • Ri 0 is H, OH, CN, COOH or CH 2 OH.
  • Particularly preferred compounds are those in which R "has a 1,3-arylene or 1,3-heteroarylene linkage (ie, the constituents -NHSO 2 R ', -SO 2 NHR', etc. of the respective moiety are relative in the meta position to the linkage of R "to the central bicycle, or in which R 3 is aryl or heteroaryl, and the substituent R '" is in the meta position relative to the linkage of R 3 to the central bicycle.
  • R "has a 1,3-arylene or 1,3-heteroarylene linkage ie, the constituents -NHSO 2 R ', -SO 2 NHR', etc. of the respective moiety are relative in the meta position to the linkage of R "to the central bicycle, or in which R 3 is aryl or heteroaryl, and the substituent R '" is in the meta position relative to the linkage of R 3 to the central bicycle.
  • phenylnaphthalenes are those compounds which carry two hydroxyl groups (preferably at the positions R 4 and R 10) and another substituent, in particular a radical R 3 as defined above.
  • Preferred radicals of R 3 are explicitly furan-3-yl, pyridine din-3-yl, pyridin-4-yl, 4-methoxypyridin-3-yl, phenyl, 3-hydroxyphenyl, 3-amino-phenyl, and the substituted phenyl radicals shown below:
  • Very particularly preferred compounds of structure (I) are the following: 3- (2-naphthyl) phenol (3), 3- (6-hydroxy-2-naphthyl) pyridine (4), 3 ( 6-hydroxy-2-naphthyl) benzoic acid (7), 4- (6-hydroxy-2-naphthyl) benzoic acid (8), / V- [3- (6-hydroxy-2-naphthyl) phenyl] acetamide (9) , 6- [3- (hydroxymethyl) phenyl] -2-naphthol (10), 6- [4- (hydroxymethyl) phenyl] -2-naphthol (11), 2- (3-hydroxyphenyl) quinoline 6-ol (12), 3- (quinolin-3-yl) phenol (13), 3- (4-hydroxyphenyl) quinolin-7-ol (15), 3- (3-hydroxyphenyl) quinoline-7 ol (16), 5- (6-hydroxy-naphthalen-2-yl) pyri
  • the process for producing the compound defined in (2) having the structure (I) according to embodiment (3) of the invention is carried out by means of a Suzuki reaction comprising reacting the compounds (II) and (III) in the presence of Pd (PPh 3 ) 4 or a comparable Pd catalyst.
  • the coupling can be carried out according to methods A, B or C outlined below.
  • method A to an oxygen-free mixture of halogen derivative (1 eq) in toluene / ethanol 2/1 or DME and 2% sodium carbonate solution (2 eq) tetrakis (triphenylphosphine) palladium (0) (0.1 eq) and boric acid (1 eq ) are added under nitrogen atmosphere.
  • the reaction mixture is refluxed for up to 24 h at 80 0 C.
  • the halogen derivative (0.2 mmol), boric acid (0.4 mmol), K 2 CO 3 (0.6 mmol) and Pd (OAc) 2 in 5 ml of DME / water / ethanol 7/3 / 2 and subjected to microwave irradiation for 300 s at 150 0 C.
  • bromine derivative (1 eq), boric acid (1.3 eq) and Pd (PPh 3 ) 4 (0.05 eq) are suspended in 1.5 ml of DMF in a suitable reaction vessel with magnetic stirrer, NaHCOs (3 eq). in 1.5 ml of water is added and the mixture is exposed for 15 min at 140 0 C and 100 W microwave radiation.
  • the compounds of the present invention are brought the invention into a suitable application form.
  • the compounds can be administered to the patient in any form of administration familiar to the person skilled in the art, although oral administration is the preferred form of administration.
  • the amount administered drug, ie the dose used depends on the nature and severity of the disease to be treated, the application and therapy, the age and the constitutional nature of the patient and is individually by the attending physician in the context of his general expertise adjusted the given situation. The invention will be explained in more detail with reference to the following examples, which, however, do not limit the invention.
  • Microwave Reactions The microwave used for the synthesis was an Emrys Optimizer Workstation.
  • NMR Nuclear Magnetic Resonance
  • the filtrate is extracted with ethyl acetate and the organic phase dried over MgSO 4 , filtered and rotary evaporated in vacuo.
  • the product was purified by column chromatography with a mixture of hexane / ethyl acetate 8/2 as eluant in a yield of 20% (700 mg).
  • 2-Methoxy-6-methylaniline 100 mg (0.73 mmol, 1 eq) are dissolved in a mixture of 10 ml of dichloromethane and 4 ml of methanol. After addition of 284.7 mg (0.73 mmol, 1 eq) of BTMBr 3 and 292 mg (292 mmol) of calcium carbonate is stirred for one hour at room temperature. Then the calcium carbonate is filtered off and 10 ml of water are added. Hydrophilic and lipophilic phase are separated and the water phase is extracted four times with ether. The combined organic extracts are dried over magnesium sulfate, filtered and the solvent is removed in vacuo. Purification of the product was not necessary (quantitative yield, 157 mg).
  • the reaction is carried out in a water / ice bath. Dissolve 500 mg (2.31 mmol, 1 eq) of 4-bromo-2-methoxy-6-methylaniline in a mixture of 7 ml of acetic acid and 3 ml of water, add 0.8 ml of concentrated hydrochloric acid (37%), and Add 207 mg (3.00 mmol, 1.5 eq) of sodium nitrite dissolved in 1 ml of water. The mixture is stirred for 30 minutes and then added to 8 ml of ice-cooled 50% by weight of hypophosphorous acid. The reaction is stirred at 0 ° C. for 8 hours and allowed to stand at RT overnight. For working up, it is extracted with ethyl acetate, dried over magnesium sulfate, filtered and the solvent evaporated in vacuo. The crude product was not further purified (yield 90%, 418 mg).
  • Tin chloride dihydrate (13.7 g, 72 mmol, 2 eq) is added and 28 ml conc. HCl was added dropwise over a period of 90 min at 100 0 C. The reaction mixture is stirred overnight, the resulting precipitate is filtered off and washed twice each with 11 ml of 1% HCl and water (yield 70%, 6.6 g).
  • the compound was prepared according to the procedure in J. Med. Chem., 2005, 48, 3953-3979.
  • 6-bromo-2-methoxy-1-naphthaldehvd 9.7 ml of TiCl 4 (2.1 eq) and 4.2 ml of dichloromethyl methyl ether (1.1 eq) are dissolved in 20 ml of dichloromethane at 0 ° C.
  • a solution of 10 g of 2-bromo-6-methoxynaphtalin (1 eq) is added dropwise, so that the temperature does not exceed 5 0 C.
  • the reaction mixture is stirred at RT overnight and then 300 ml of 1% HCl are added. Organic and aqueous phase are separated and the aqueous phase extracted with dichloromethane.
  • 6-Bromo-2-naphthol 500 mg, 2.242 mmol, 1 eq
  • NBS 558.7 mg, 3.319 mmol, 1.4 eq
  • acetone 4.4 ml HCl 1N 22 ⁇ l for 15 min at RT touched.
  • Ethyl acetate is added and this organic phase is washed with 1N HCl three times. After drying over magnesium sulfate, filtering and evaporating in vacuo, the desired product is obtained in quantitative yield (677 mg).
  • 6-Bromo-2-methoxynaphthalene (5.9 g, 24.81 mmol, 1 eq) and NBS (4.41, 24.81 mmol, 1 eq) is boiled in THF 50 ml for 2 h under reflux. After washing with 1N HCl, the oragnic phase is dried over magnesium sulfate, filtered and concentrated in vacuo. The compound is obtained in quantitative yield (7.8 g).
  • Lithium hydroxide (477 mg, 11.37 mmol, 3.5 eq) is added to a solution of methyl 3-methoxy-7- (3-methoxyphenyl) -2-naphthylic acid (1.05 g, cooled to 0 ° C.).
  • aqueous phase is extracted several times with ethyl acetate and the combined organic phases are washed with a saturated NaCl solution, dried over MgSO 4 , filtered and evaporated to dryness.
  • a column chromatographic purification (eluent dichloromethane / hexane 7/3) gives the desired product in a yield of 15% (760 mg).
  • Method A To an oxygen-free mixture of halogen derivative (1 eq) in toluene / ethanol 2/1 or DME and 2% sodium carbonate solution (2 eq) is added tetrakis (triphenylphosphine) palladium (0) (0.1 eq) and boric acid (1 eq) under nitrogen atmosphere. The reaction mixture is up to 24 hours refluxed at 80 0 C. To work up the reaction, the hydrophilic and the lipophilic phase are separated from one another and the hydrophilic phase is extracted with dichloromethane or ethyl acetate. The combined organic phases are then washed with a 2% hydrochloric acid solution to remove any remaining boric acid, and alkalized with 2% sodium carbonate solution. After further washing with water and subsequent drying by means of magnesium sulfate, the solvent is removed in vacuo. Purification of the desired product was usually carried out by column chromatography.
  • Method B In a dry reaction vessel, the halogen derivative (0.2 mmol), boric acid (0.4 mmol), K 2 CO 3 (0.6 mmol) and Pd (OAc) 2 in 5 ml of DME / water / ethanol 7 / 3/2 suspended and a microwave irradiation for 300 s at 150 0 C subjected. The reaction mixture is then filtered and concentrated in vacuo. Purification is carried out by means of preparative HPLC (Waters fraction Lynx Autopurification System, Varian Inertsil C18 column 50 ⁇ 21 mm, particle size 3 ⁇ m, gradient with isocratic end period, solvent: acetonitrile, water, formic acid (0.01%) 0-100%).
  • preparative HPLC Waters fraction Lynx Autopurification System, Varian Inertsil C18 column 50 ⁇ 21 mm, particle size 3 ⁇ m, gradient with isocratic end period, solvent: acetonitrile, water, formic acid (0.01%) 0-
  • the compound is prepared by reaction of 3-bromoquinoline (500 mg, 2.40 mmol, 1 eq) with 4-methoxyphenylboronic acid (365 mg, 2.40 mmol, 1 eq), corresponding to
  • the compound is prepared by reaction of 3-bromoquinoline (200 mg, 0.96 mmol, 1.06 eq) and 3-methoxyphenylboronic acid (138 mg, 0.91 mmol, 1 eq), according to Method A, in 19 h.
  • a column chromatographic purification with a mixture of hexane / ethyl acetate 3/1 gives the desired product in a yield of 70% (147 mg).
  • the compound is reacted in a reaction of 3-bromo-7-methoxyquinolines (255 mg, 1.07 mmol, 1.06 eq) and 3-methoxyphenylboronic acid (154 mg, 1.01 mmol, 1 eq), according to method A, shown in 4.5 hours.
  • a column chromatographic purification with a mixture hexane / ethyl acetate 9/1 provides the desired product in a yield of 76% (215 mg).
  • the compound is prepared by reaction of 6-methoxynaphthaleneboronic acid (258 mg, 1.28 mmol, 1.2 eq) with 3-bromo-5-methoxypyridine (200 mg, 1.06 mmol, 1 eq) according to method A, in 24 h.
  • a column chromatographic purification with a mixture of hexane / ethyl acetate 2/1 gives the desired product in a yield of 84% (237 mg).
  • the compound is prepared by reaction of 2-bromo-6-methoxynaphthalene (500 mg, 2.11 mmol, 1 eq) with 4-methoxyphenylboronic acid (313 mg, 2.12 mmol, 1 eq) according to method A, in 5 h shown.
  • the desired product was precipitated from hexane in 56% (311 mg) yield.
  • the compound is prepared by reaction of 2-bromo-6-methoxynaphthalene (500 mg, 2.11 mmol, 1 eq) with 3-methoxyphenylboronic acid (321 mg, 2.11 mmol, 1 eq). speaking the method A, presented within 22 h. A column chromatography with a mixture of hexane / ethyl acetate 9/1 provides the desired product in a yield of 81% (451 mg).
  • the compound is prepared by reaction of 2-bromo-6-methoxynaphthalene (300 mg, 1.26 mmol, 1 eq) with 2-methoxyphenylboronic acid (192 mg, 1.26 mmol, 1 eq), according to method A, within 18 h.
  • a column chromatographic purification was not carried out, the crude product was used for further syntheses.
  • the compound is prepared by reaction of 3-bromonitrobenzene (1 g, 4.95 mmol, 1 eq) with 6-methoxynaphthaleneboronic acid (1 g, 4.95 mmol, 1 eq) according to method A, within 20 h.
  • a column chromatographic purification with hexane as eluent gives the desired product in a yield of 40%, 557 mg.
  • the compound is prepared by reaction of 6-bromo-2-naphthol (500 mg, 2.24 mmol, 1 eq) with phenylboronic acid (1 eq), according to method A, within 20 h.
  • a column chromatographic purification with hexane / ethyl acetate 7/3 as eluent gives the desired product in a yield of 87%, 429 mg.
  • the compound is prepared by reacting 2-bromonaphthalene (303 mg, 1.46 mmol, 1 eq) with 3-hydroxyphenylboronic acid (242 mg, 1.76 mmol, 1.2 eq) according to Method A, within 18 h , A column chromatographic purification with dichloromethane as eluent gives the desired product in a yield of 47%, 152 mg.
  • the compound is prepared by reaction of 5-oxo-5,6,7,8-tetrahydronaphthalene-2-yl trifluoromethanesulfonate (1.78 g, 6.04 mmol, 1 eq) with 3-hydroxyphenylboronic acid (1 g, 7.25 mmol, 1 , 2 eq), according to method A, within 4 h.
  • a column chromatographic purification with dichloromethane as eluent gives the desired product in a yield of 29%, 900 mg.
  • the compound is prepared by reaction of 5-oxo-5,6,7,8-tetrahydronaphthalene-2-yl trifluoromethanesulfonate (1.61 g, 5.48 mmol, 1 eq) with 4-hydroxyphenylboronic acid (1 g, 6.58 mmol, 1 , 2 eq), according to method A, within 4 h.
  • a column chromatographic purification with dichloromethane / hexane 7/3 as eluent gives the desired product in a yield of 72%, 1.2 g.
  • the compound is prepared by reaction of 2-hydroxy-7-triflate naphthalene (292 mg, 1 mmol, 1 eq) with 3-hydroxyphenylboronic acid (166 mg, 1.2 mmol, 1.2 eq), according to method A, within 5 h.
  • a column chromatographic purification with hexane / ethyl acetate 8/2 as the eluent gives the desired product in a yield of 26%, 61 mg.
  • the compound is prepared by reacting (E) -3- (3-bromo-5-methoxy-phenyl) -acrylic acid (751 mg, 2.92 mmol, 1 eq) with 6-methoxynaphthaleneboronic acid (1 eq), according to Method A. , shown in 26h. Acidify the aqueous phase with conc. HCl provided the desired compound as a precipitate in 64% yield, 624 mg.
  • the compound is prepared by reaction of 4-bromo-2-methoxyaniline (400 mg, 1.98 mmol, 1 eq) with 6-methoxynaphthaleneboronic acid (600 mg, 2.97 mmol, 1.5 eq) according to method A, in 2.5 h. Acidification of the organic phase with a solution of HCl 2 M in diethyl ether afforded the desired compound as a precipitate in a yield of 50%, 312 mg.
  • the compound is prepared by reaction of 1-bromo-3-methoxy-5-methylbenzene (200 mg, 1.00 mmol, 1 eq) with 6-methoxynaphthaleneboronic acid (201 mg, 1.00 mmol, 1 eq) according to Method A. , shown in 22h.
  • a column chromatographic purification with hexane / ethyl acetate 9/1 as the eluent gave the desired compound in a yield of 60%, 167 mg.
  • connection is won in two steps.
  • 1,3-dibromo-5-methoxybenzene (290 mg, 1.09 mmol, 1 eq) reacts with 6-methoxynaphthaleneboronic acid (331 mg, 1.64 mmol, 1.5 eq) according to method A for 18 h.
  • 6-methoxynaphthaleneboronic acid (331 mg, 1.64 mmol, 1.5 eq) according to method A for 18 h.
  • a column chromatographic purification with hexane as eluent gave the intermediate (yield 32%), which is subjected to another Suzukirecision.
  • the intermediate (182 mg, 0.53 mmol, 1 eq) is subjected to a reaction with 4-methoxybenzene boronic acid (121 mg, 0.79 mmol, 1.5 eq) for a further 18 h, according to method A.
  • a column chromatographic purification with hexane as eluent gives the desired compound in a
  • the compound is prepared by reaction of l, 3-dibromo-5-methoxybenzene (250 mg, 0.93 mmol, 1 eq) with 6-methoxy-2-naphthaleneboronic acid (472 mg, 2.34 mmol, 2.5 eq), according to the method A, shown.
  • the compound is prepared by reaction of methyl 7-bromo-3-methoxy-2-naphthylate (2.2 g, 7.45 mmol, 1 eq) with 3-methoxyphenylboronic acid (1.37 g, 8.95 mmol, 1.5 eq ), according to the method A, shown.
  • a column chromatographic purification with a gradient of hexane / dichloromethane 1/1 to 3/7 and dichloromethane / methanol 95/5 as eluent afforded the desired compound in a yield of 66%, 1.58 g.
  • the compound is prepared by reaction of 3-bromo-7-methoxy-1-naphthonitrile (1.4 g, 1.76 mmol, 1 eq) with 3-methoxyphenylboronic acid (320 mg, 2.11 mmol, 1.2 eq), according to the method A, shown in 24 h. Purification by column chromatography with hexane / ethyl acetate 9/1 as eluent gave the desired compound in a yield of 53%, 300 mg.
  • the compound is prepared by reaction of 8-cyano-6-methoxynaphthalene-2-yl trifluoromethanesulfonate (466 mg, 1.4 mmol, 1 eq) with 3-methoxyphenylboronic acid (260 mg, 1.7 mmol, 1.2 eq). , according to the method A, shown overnight. Column chromatographic purification with hexane / ethyl acetate 7/3 as eluent afforded the desired compound in a yield of 25%, 113 mg.
  • the compound is prepared by reaction of 6-bromo-2-methoxy-1-naphthaldehyde (2 g, 7.55 mmol, 1 eq) with 3-methoxyphenylboronic acid (1.3 g, 8.30 mmol, 1.1 eq), according to the method A, shown.
  • a column chromatographic purification with hexane / ethyl acetate 1/1 as eluent afforded the desired compound in a yield of 86%, 1.9 g.
  • the compound is prepared by reaction of 1,6-dibromo-2-methoxynaphthalene (5.75 g, 18.20 mmol, 1 eq) with 3-methoxyphenylboronic acid (2.77 g, 18.20 mmol, 1 eq), corresponding to Method A, shown.
  • a column chromatographic purification with hexane as the eluent gave the desired compound in a yield of 81%, 5.1 g.
  • the compound is prepared by reaction of 1-bromo-6- (3-hydroxy-phenyl) -2-naphthol (50 mg, 0.16 mmol, 1 eq) with phenylboronic acid (19.4 mg, 0.16 mmol, 1 eq ), according to the method A, shown.
  • the compound is prepared by reaction of l, 6-dibromonaphthalene-2-ol (200 mg, 0.66 mmol, 1 eq) with 3-hydroxybenzeneboronic acid (182.8 mg, 1.32 mmol, 2 eq), according to method A, shown in 21 h.
  • a column chromatographic purification with dichloromethane / methanol 99/1 provides the desired compound in a yield of 5%, 11 mg.
  • the compound is prepared by reaction of 1-bromo-6- (3-hydroxyphenyl) naphthalene-2-ol (80 mg, 0.25 mmol, 1 eq) with 3-furanboric acid (28.4 mg, 0.25 mmol, 1 eq), according to method A, in 21 h.
  • a column chromatographic purification with dichloromethane / methanol 98/2 gives the desired compound in a yield of 46%, 35 mg.
  • the compound is prepared by reaction of 1-bromo-6- (3-hydroxyphenyl) naphthalene-2-ol (100 mg, 0.32 mmol, 1 eq) with 3-pyridinboric acid (39 mg, 0.32 mmol, leq) according to the method A, shown in 21 h.
  • Column chromatographic purification with hexane / ethyl acetate 7/3 yields the desired compound in a yield of 59%, 59 mg.
  • the compound is prepared by reaction of 1-bromo-6- (3-hydroxyphenyl) naphthalene-2-ol (100 mg, 0.32 mmol, 1 eq) with 4-pyridineboronic acid (39 mg, 0.32 mmol, 1 eq). , according to method A, shown in 1 h.
  • a column chromatographic purification with hexane / ethyl acetate 8/2 gives the desired compound in a yield of 44%, 44 mg.
  • the compound is prepared by reaction of 1-bromo-6- (3-hydroxyphenyl) naphthalene-2-ol (150 mg, 0.48 mmol, 1 eq) with 3-pyrimidineboronic acid (59.0 mg, 0.48 mmol, 1 eq), according to method A, in 15 h.
  • a column chromatographic purification with dichloromethane / methanol 99/1 gives the desired compound in a yield of 9%, 14 mg.
  • the compound is prepared by reaction of l-bromo-6- (3-hydroxyphenyl) naphthalene-2-ol (100 mg, 0.32 mmol, 1 eq) with 6-methoxypyridin-3-ylboronic acid (48.6 mg, 0, 32 mmol, 1 eq), according to method A, in 3 h.
  • a column chromatographic purification with hexane / ethyl acetate 8/2 gives the desired compound in a yield of 84%, 92 mg.
  • the compound is prepared by reaction of 1-bromo-6- (3-hydroxyphenyl) naphthalene-2-ol (100 mg, 0.32 mmol, 1 eq) with 3-carboxybenzeneboroboric acid (52.7 mg, 0.32 mmol, 1 eq), according to method A, shown overnight.
  • a column chromatography with hexane / ethyl acetate 3/2 gives the desired compound in a yield of 15%, 17 mg.
  • the compound is prepared by reaction of l-bromo-6- (3-hydroxyphenyl) naphthalene-2-ol (100 mg, 0.32 mmol, 1 eq) with N- (3- (4,4,5,5-tetramethyl -l, 3,2-dioxaborolan-2-yl) phenyl) methanesulfonamide (94.3 mg, 0.32 mmol, 1 eq), according to method A, in 2 h.
  • a column chromatographic purification with hexane / ethyl acetate 3/2 gives the desired compound in a yield of 64%, 83 mg.
  • the compound is prepared by reaction of 1-bromo-6- (3-hydroxyphenyl) naphthalene-2-ol (150 mg, 0.48 mmol, 1 eq) with 4-morpholinophenylboronic acid (98.6 mg, 0.48 mmol, 1 eq), according to method A, in 21 h.
  • a column chromatography with dichloromethane / methanol 99/1 provides the desired compound in a yield of 7%, 13 mg.
  • the compound is prepared by reaction of l-bromo-6- (3-hydroxyphenyl) naphthalene-2-ol (150 mg, 0.48 mmol, 1 eq) with N- (3- (4,4,5,5-tetramethyl -l, 3,2-dioxaborolan-2-yl) phenyl) acetamide (124.3 mg, 0.48 mmol, 1 eq), according to method A, in 2 h.
  • a column chromatographic purification with hexane / ethyl acetate 8/2 gives the desired compound in a yield of 77%, 136 mg.
  • the compound is prepared by reaction of 1-bromo-6- (3-hydroxyphenyl) naphthalene-2-ol (100 mg, 0.32 mmol, 1 eq) with 4-oxo-4- (4- (4,4,5 , 5-tetramethyl-l, 3,2-dioxaborolan-2-yl) phenylamino) butanoic acid (101.3 mg, 0.32 mmol, 1 eq), according to method A, in 10 min.
  • a column chromatography with dichloromethane / methanol 90/10 provides the desired compound in a yield of 12%, 16 mg.
  • the compound is prepared by reaction of 3-bromo-N- (thiazol-2-yl) benzenesulfonamide (88.8 mg, 0.28 mmol, 1 eq) with 2-methoxy-6- (3-methoxyphenyl) naphthalene-1-one. ylboronic acid (100 mg, 0.36 mmol, 1.3 eq), according to Method C. A column chromatographic purification with dichloromethane / methanol 95/5 gives the desired compound in a yield of 14%, 19 mg.
  • the compound is prepared by reaction of 3-bromo-N- (5-methyl-1,3,4-thiadiazol-2-yl) benzenesulfonamide (92.4 mg, 0.28 mmol, 1 eq) with 2-methoxy-6 - (3-methoxyphenyl) naphthalene-1-ylboronic acid (100 mg, 0.36 mmol, 1.3 eq), according to Method C.
  • the crude product was not characterized but subjected directly to ether cleavage.
  • the compound was prepared by reaction of 3-methoxy-5- (6-methoxynaphthalene-2-yl) benzoic acid (500 mg, 1.62 mmol, 1 eq) with methylamine solution 33% (1 eq) according to the method D. Purification by column chromatography with hexane / ethyl acetate 1/1 as eluent yielded the desired product in 28% yield, 148 mg.
  • the compound was prepared by reaction of 3-methoxy-5- (6-methoxynaphthalene-2-yl) benzoic acid (500 mg, 1.62 mmol, 1 eq) with aniline (1 eq), according to method D. Purification by column chromatography with hexane / dichloromethane 1/1 as the eluant gave the desired product in 11% yield, 68 mg.
  • the compound was prepared by reacting (E) -3- [3-methoxy-5- (6-methoxynaphthalene-2-yl) -phenyl] -acrylic acid (300 mg, 0.89 mmol, 1 eq) with methylamine 33% (1 eq), according to method D.
  • a column chromatographic purification with hexane / ethyl acetate 2/1 as the eluent gave the desired product in a yield of 53%, 200 mg.
  • the compound was prepared by reacting (E) -3- [3-methoxy-5- (6-methoxy-naphthalen-2-yl) -phenyl] -acrylic acid (300 mg, 0.89 mmol, 1 eq) with aniline (1 eq), according to method D.
  • a column chromatographic purification with hexane / ethyl acetate 2/1 as the eluent gave the desired product in a yield of 62%, 227 mg.
  • the compound was prepared by reacting (E) -3- (2-methoxy-6- (3-methoxyphenyl) naphthalene-1-yl) acrylic acid (200 mg, 0.60 mmol, 1 eq) with methylamine 33% (1 eq), according to method D. A column-chromatographic purification was not necessary. The desired compound was obtained in quantitative yield.
  • the compound was prepared by reacting (E) -3- (2-methoxy-6- (3-methoxyphenyl) naphthalene-1-yl) acrylic acid (200 mg, 0.60 mmol, 1 eq) with aniline (1 eq). , according to the method D. A characterization of the compound was not made. The crude product was used directly for ether cleavage.
  • the compound was prepared by reaction of 2-methoxy-6- (3-methoxyphenyl) -1-naphthoic acid (300 mg, 0.97 mmol, 1 eq) with methylamine 33% (1 eq), according to method D. A column chromatographic Purification was not necessary. The desired compound was obtained in quantitative yield (311 mg).
  • the compound was prepared by reaction of 2-methoxy-6- (3-methoxyphenyl) -l-naphthoic acid (300 mg, 0.97 mmol, 1 eq) with aniline (1 eq), according to Method D.
  • a column-chromatographic purification with Hexane / ethyl acetate 2/1 as the eluant gave the desired compound in 85% yield, 316 mg.
  • the compound was prepared by reaction of 2-methoxy-6- (3-methoxyphenyl) -1-naphthoic acid (300 mg, 0.97 mmol, 1 eq) with m-anisole (1 eq), according to Method D.
  • the compound was prepared by reaction of 2-methoxy-6- (3-methoxyphenyl) -1-naphthoic acid (300 mg, 0.97 mmol, 1 eq) with Boc-piperazine (1 eq), according to method D. A characterization the connection was not made. The crude product was used directly for ether cleavage.
  • 2-Methoxy-6- (3-methoxyphenyl) -1-naphthoic acid (200 mg, 0.65 mmol, 1 eq) is boiled under reflux with 5 ml of thionyl chloride under a nitrogen atmosphere for 30 minutes, according to method E.
  • the residue is taken to be dry Dissolved THF and added to 1 ml of cooled to 0 0 C morpholine.
  • the reaction mixture is boiled overnight under reflux. After evaporation of the solvent, a column chromatographic purification with dichloromethane / methanol 95/5 is made as eluent. A characterization of the compound was not made.
  • the crude product was used directly for ether cleavage.
  • the compound was prepared by reaction of 2-methoxy-6- (3-methoxyphenyl) -1-naphthoic acid (450 mg, 1.46 mmol, 1 eq) with 2-aminothiazole (90 mg, 0.87 mmol, 0.6 eq), according to Method E. No characterization of the compound was performed. The crude product was used directly for the electron cleavage C 22 H 18 O 3 N 2 S, MW 390
  • the compound was prepared by reaction of 2-methoxy-6- (3-methoxyphenyl) -1-naphthoic acid (450 mg, 1.46 mmol, 1 eq) with 2-amino-3,4-dimethylisoxazole (0.6 eq). , according to the method E.
  • a column chromatographic purification with hexane / ethyl acetate 7/3 as the eluent gave the desired product in a yield of 55%, 323 mg.
  • the compound was prepared by reaction of 2-methoxy-6- (3-methoxyphenyl) -1-naphthoic acid (450 mg, 1.46 mmol, 1 eq) with 2-amino-5-methyl-1,3,4-thiadiazole (0.6 eq), according to Method E. No characterization of the compound was performed. The crude product was used directly for ether cleavage. C 22 H 19 O 3 N 3 S, MW 405
  • the compound was prepared by reaction of 2-methoxy-6- (3-methoxyphenyl) -1-naphthoic acid (450 mg, 1.46 mmol, 1 eq) with 2-aminopyridine (0.6 eq), according to Method E. Column chromatographic purification with hexane / ethyl acetate 7/3 as eluent gave the desired product in 48% yield, 269 mg.
  • the compound was prepared by reaction of 2-methoxy-6- (3-methoxyphenyl) -1-naphthoic acid (450 mg, 1.46 mmol, 1 eq) with 2-aminopyrimidine (0.6 eq), according to Method E. A characterization of the compound was not made. The crude product was used directly for ether cleavage. C 23 H 19 O 3 N 3 , MW 385
  • the reaction is carried out in the microwave at 110 ° C. for 15 minutes. After cooling, the reaction mixture is dissolved in dichloromethane and purified by column chromatography with dichloromethane and 2% methanol as eluent. The compound was obtained in quantitative yield.
  • Method F The corresponding methoxy compound (1 eq) is dissolved in 15 ml of toluene and aluminum chloride (5 eq) added. The mixture is boiled for 2 hours under reflux and nitrogen atmosphere and added after cooling with 2% Na 2 CO 3 . Hydrophilic and lipophilic phases are separated and the water phase is extracted with ethyl acetate. The combined organic phases are dried over magnesium sulfate. After filtering off and removal of the solvent in vacuo, the purification is carried out by column chromatography.
  • the compound is prepared by reaction of 5-methoxy-2- (3-methoxyphenyl) -1H-indole (89 mg, 0.35 mmol, 1 eq) with boron tribromide (2.1 mL, 2.1 mmol, 6 eq). , according to the method G. Purification by means of preparative thin layer chromatography with hexane / ethyl acetate 4/6 gives the desired product in a yield of 76%, 60 mg.
  • the compound is prepared by reaction of 6-methoxy-2- (3-methoxyphenyl) quinoline (77 mg, 0.29 mmol, 1 eq) with boron tribromide (15 eq), according to Method G.
  • a column chromatographic purification with hexane / ethyl acetate 7/3 as eluent gives the desired product in a yield of 10%, 7 mg.
  • the compound is prepared by reaction of 3- (3-methoxyphenyl) quinolines (101 mg, 0.43 mmol, 1 eq) with aluminum trichloride (341 mg, 2.57 mmol, 6 eq), according to Method F. Purification by means of preparative thin layer chromatography with dichloromethane / methanol 95/5 (1 mm thick TLC plates) yields the desired product in 85% yield, 81 mg.
  • the compound is prepared by reaction of 3- (4-methoxyphenyl) quinoline with aluminum trichloride (564 mg, 4.23 mmol, 5 eq), according to Method F.
  • a column chromatographic purification with dichloromethane / methanol 99/1 as eluent gives the desired Product in a yield of 82%, 156 mg.
  • the compound is prepared by reaction of 7-methoxy-3- (4-methoxyphenyl) quinoline (96 mg, 0.37 mmol, 1 eq) with aluminum trichloride (392 mg, 2.95 mmol, 6 eq) according to Method F A column chromatographic purification with dichloromethane / methanol 96/4 as eluent gives the desired product in a yield of 63%, 55 mg.
  • the compound is prepared by reaction of 7-methoxy-3- (3-methoxyphenyl) quinoline (108 mg, 0.40 mmol, 1 eq) with aluminum trichloride (433 mg, 3.26 mmol, 8 eq) according to Method F A column chromatographic purification with dichloromethane / methanol 95/5 as eluent gives the desired product in a yield of 76%, 73 mg.
  • the compound is prepared by reaction of 3-methoxy-5- (6-methoxynaphthalen-2-yl) pyridine (200 mg, 0.75 mmol, 1 eq) with aluminum trichloride
  • the compound is prepared by reaction of 2-methoxy-6- (2-methoxyphenyl) naphthalene (150 mg, 0.57 mmol, 1 eq) with boron tribromide (8.5 mL, 8.5 mmol, 15 eq), corresponding to Method G.
  • a column chromatographic purification with hexane / ethyl acetate as eluent yields the desired product in a yield of 7%, 9 mg.
  • the compound is prepared by reaction of 2-methoxy-6- (3-methoxyphenyl) naphthalene (51 mg, 0.19 mmol, 1 eq) with boron tribromide (0.6 mL, 0.60 mmol, 3 eq), corresponding to Method G.
  • a column chromatographic purification with dichloromethane / methanol 98/2 as eluent gives the desired product in a yield of 52%, 23 mg.
  • the compound is prepared by reacting 2-methoxy-6- (4-methoxyphenyl) naphthalene (150 mg, 0.57 mmol, 1 eq) with boron tribromide (3.4 mL, 3.4 mmol, 3 eq), corresponding to Method G.
  • Column chromatographic purification with hexane / ethyl acetate 9/1 as eluent yields the desired product in 98% yield, 132 mg.
  • the compound is prepared by reaction of 6- (4-methoxyphenyl) -n-naphthol (57 mg, 0.23 mol, 1 eq) with boron tribromide (2.3 mL, 2.28 mmol, 10 eq) according to the method G.
  • a column chromatographic purification with 6- (4-methoxyphenyl) -n-naphthol 57 mg, 0.23 mol, 1 eq
  • boron tribromide 2.3 mL, 2.28 mmol, 10 eq
  • the compound is prepared by reaction of 6-methoxy-6- (3-nitro-phenyl) -naphthalene (200 mg, 0.72 mmol, 1 eq) with boron tribromide (5.6 eq), according to Method G.
  • a column chromatographic Purification with hexane / ethyl acetate 9/1 as eluent yields the desired product in a yield of 47%, 90 mg.
  • the compound is prepared by reaction of 3- (6-methoxynaphthalen-2-yl) -phenylamines (87.9 mg, 0.35 mmol, 1 eq) with boron tribromide (5 eq), according to Method G.
  • a column chromatographic Purification with hexane / ethyl acetate 9/1 as eluent gives the desired product in a yield of 14%, 12 mg.
  • the compound is prepared by reaction of 2-methoxy-6- (3-methoxy-5-methylphenyl) naphthalene (26 mg, 0.10 mmol, 1 eq) with boron tribromide solution (1 mL, 1 mmol, 10 eq) according to the method G. After working up, the analytically clean compound was obtained (quantitative yield, 25 mg).
  • the compound is prepared by reaction of 2- (4 ', 5-dimethoxy-1, 1'-biphenyl-3-yl) -6-methoxynaphthalene (40 mg, 0.11 mmol, 1 eq) with boron tribromide solution (1.35 ml, 1.35 mmol, 12 eq), according to Method G.
  • the desired product was obtained as a precipitate after addition of water in a yield of 92%, 34 mg.
  • the compound is prepared by reaction of 2-methoxy-6- [3-methoxy-5- (6-methoxy-2-naphthyl) phenyl] naphthalene (100 mg, 0.24 mmol, 1 eq) with boron tribromide solution (3 , 6 ml, 3.6 mmol, 15 eq), according to Method G.
  • a column chromatographic purification with hexane / ethyl acetate 9/1 gave the desired product in a yield of 99%, 90 mg.
  • the compound is prepared by reaction of 3-methoxy-5- (6-methoxynaphthalene-2-yl) -N-methyl-benzamide (110 mg, 0.34 mmol, 1 eq) with boron tribromide solution (5 eq). , according to method G. A column-chromatographic purification was not necessary. The desired product was obtained after working up in quantitative yield (99.6 mg).
  • the compound is prepared by reaction of 3-methoxy-5- (6-methoxynaphthalene-2-yl) -N-phenylbenzamide (300 mg, 0.78 mmol, 1 eq) with boron tribromide solution (5 eq). , according to method G.
  • a column chromatographic purification with hexane / dichloromethane 1/99 gave the desired product in a yield of 11%, 32 mg.
  • the compound is prepared by reacting (E) -3- [3-methoxy-5- (6-methoxynaphthalen-2-yl) -phenyl] -N-methyl-acrylamide (200 mg, 0.58 mmol, 1 eq) with boron tribromide solution (5 eq), according to Method G. Purification by preparative thin layer chromatography with dichloromethane / methanol 90/10 gave the desired product in a yield of 13%, 25 mg.
  • the compound is prepared by reacting (E) -3- [3-methoxy-5- (6-methoxynaphthalene-2-yl) -phenyl] -N-phenyl-acrylamide (126 mg, 0.31 mmol, 1 eq) with boron tribromide solution (5 eq), according to Method G. Purification by preparative thin layer chromatography with dichloromethane / methanol 98/2 gave the desired product in a yield of 14%, 17 mg.
  • the compound is prepared by reaction of 3- [3-methoxy-5- (6-methoxynaphthalene-2-yl) -phenyl] -N-methyl-propionamide (47.8 mg, 0.16 mmol, 1 eq). with aluminum trichloride (213.3 mg, 1.60 mmol, 10 eq), according to method F. Purification by preparative thin-layer chromatography with dichloromethane / methanol 98/2 gave the desired product in a yield of 35%, 16 mg.
  • the compound is prepared by reacting 3- [3-methoxy-5- (6-methoxynaphthalene-2-yl) -phenyl] -N-phenyl-propionamide (55 mg, 0.13 mmol, 1 eq) with A - liumium trichloride (291.3 mg, 2.19 mmol, 12 eq), according to method F. Purification by preparative thin layer chromatography with hexane / ethyl acetate 1/1 gave the desired product in a yield of 23%, 17 mg.
  • the compound is prepared by reaction of / V- [2-methoxy-4- (6-methoxy-2-naphthyl) phenyl] acetamide (54 mg, 0.17 mol, 1 eq) with boron tribromide solution (2.52 ml, 2 , 52 mmol, 15 eq), according to Method G. Purification by preparative thin layer chromatography with dichloromethane / methanol 95/5 gave the desired product in a yield of 45%, 22 mg.
  • the compound is prepared by reaction of / V- [2-methoxy-4- (6-methoxy-2-naphthyl) phenyl] benzamide (49 mg, 0.13 mol, 1 eq) with boron tribromide solution (2.25 ml, 2 , 25 mmol, 15 eq), according to Method G. Purification by means of prepa- rative thin-layer chromatography with dichloromethane / methanol 95/5 as eluent yielded the desired product in quantitative yield.
  • the compound is prepared by reaction of 3-methoxy-7- (3-methoxyphenyl) - / V-methyl-2-naphthamide (93 mg, 0.29 mol, 1 eq) with boron tribromide solution (5.5 mL, 5.5 mmol, 19 eq), according to method G. Purification was not necessary. The desired product was obtained in quantitative yield.
  • the compound is prepared by reaction of 3-methoxy-7- (3-methoxyphenyl) - / V-phenyl-2-naphthamide (86 mg, 0.22 mol, 1 eq) with boron tribromide solution (4.45 ml, 4 , 45 mmol, 20 eq), according to Method G. Purification by preparative thin layer chromatography with hexane / ethyl acetate 6/4 as eluent gave the desired product in a yield of 50%, 39 mg.
  • the compound is prepared by reacting (E) -3- (2-hydroxy-6- (3-hydroxyphenyl) naphthalen-1-yl) -N-methylacrylamide (127 mg, 0.37 mmol, 1 eq) with boron tribromide solution ( 2.9 ml, 2.31 mmol, 8 eq), according to Method G.
  • a column chromatographic purification with dichloromethane / methanol 95/5 as eluent gave the desired product in a yield of 60%, 71 mg.
  • the compound is prepared by reacting (E) -3- (2-hydroxy-6- (3-hydroxyphenyl) naphthalen-1-yl) -N-phenylacrylamide (50 mg, 0.12 mmol, 1 eq) with boron tribromide solution ( 0.98 mmol, 8 eq), according to Method G.
  • a column chromatographic purification with dichloromethane / methanol 95/5 as eluent gave the desired product in a yield of 13%, 6 mg.
  • the compound is prepared by reaction of 2-methoxy-6- (3-methoxyphenyl) -N-methyl-1-naphthamide (250 mg, 0.78 mmol, 1 eq) with boron tribromide solution (3.89 mmol, 5 eq), according to Method G.
  • a column chromatographic purification with dichloromethane / methanol 98/2 as eluent gave the desired product in a yield of 95%, 217 mg.
  • the compound is prepared by reaction of 2-methoxy-6- (3-methoxyphenyl) -N-phenyl-1-naphthamide (217 mg, 0.57 mmol, 1 eq) with boron tribromide solution (3.39 mmol, 6 eq), according to Method G.
  • a column chromatographic purification with dichloromethane / methanol 95/5 as eluent gave the desired product in a yield of 23%, 47 mg.
  • the compound is prepared by reaction of 2-methoxy-N, 6-bis (3-methoxyphenyl) -l-naphthamide (150 mg, 0.36 mmol, 1 eq) with boron tribromide solution (2.9 mL, 2.9 mmol, 8 eq), according to Method G.
  • a column chromatographic purification with dichloromethane / methanol 93/7 as eluent gave the desired product in a yield of 75%, 100 mg.
  • the compound is prepared by reaction of 2-methoxy-6- (3-methoxyphenyl) naphthalene-1-yl) (morpholino) methanone (195 mg, 0.52 mmol, 1 eq) with boron tribromide solution (2.6 ml, 2, 58 mmol, 5 eq), according to Method G.
  • a column chromatographic purification with dichloromethane / methanol 95/5 as eluent gave the desired product in a yield of 93%, 169 mg.
  • the compound is prepared by reaction of tert-butyl 4- (2-methoxy-6- (3-methoxyphenyl) -l-naphthoyl) piperazine-1-carboxylate (200 mg, 0.42 mmol, 1 eq) with boron tribromide solution (2 , 5 ml, 2.52 mmol, 6 eq), according to Method G. A column-chromatographic purification was not necessary. Extraction of the aqueous phase with ethyl acetate provides the desired product in 60% yield, 88 mg.
  • the compound is prepared by reaction of 2-methoxy-6- (3-methoxyphenyl) -N- (thiazol-2-yl) -l-naphthamide (117 mg, 0.30 mmol, 1 eq) with boron tribromide solution (7 eq). , according to the method G.
  • a column chromatographic purification with dichloromethane / methanol 95/5 as eluent yielded the desired product.
  • the compound is prepared by reaction of the corresponding methoxy compound (117 mg, 0.30 mmol, 1 eq) with boron tribromide solution (7 eq), according to Method G.
  • a column chromatography with dichloromethane / methanol 95/5 as eluent provided the desired product in a yield of 20%, 23 mg.
  • the compound is prepared by reaction of the corresponding methoxy compound (117 mg, 0.30 mmol, 1 eq) with boron tribromide solution (7 eq), according to Method G.
  • a column chromatography with dichloromethane / methanol 95/5 as eluent provided the desired product in a yield of 36%, 38 mg.
  • the compound is prepared by reaction of the corresponding methoxy compound (117 mg, 0.30 mmol, 1 eq) with boron tribromide solution (7 eq), according to Method G.
  • a column chromatography with dichloromethane / methanol 95/5 as eluent provided the desired product in a yield of 56%, 60 mg.
  • the compound is prepared by reaction of 1-bromo-2-methoxy-6- (3-methoxyphenyl) naphthalene (500 mg, 1.46 mmol, 1 eq) with boron tribromide solution (7.3 mL, 7.3 mmol, 5 eq ), according to method G. Purification of the crude product was not necessary. The desired product was obtained in quantitative yield (460 mg).
  • the compound is prepared by reaction of 3- (2-methoxy-6- (3-methoxyphenyl) naphthalen-1-yl) benzenamine (1 g, 2.82 mmol, 1 eq) with boron tribromide solution (14.1 ml, 14 , 1 mmol, 5 eq), according to the method G. A column chromatographic purification was not necessary, the desired product was obtained after work-up in quantitative yield (922 mg).
  • the compound is prepared by reaction of 3- (2-methoxy-6- (3-methoxyphenyl) naphthalen-1-yl) -N- (thiazol-2-yl) benzenesulfonamide (75.3 mg, 0.15 mmol, 1 eq ) with boron tribromide solution (14.1 ml, 14.1 mmol, 5 eq), according to the method G. Purification by preparative thin layer chromatography with dichloromethane / methanol 92.5 / 7.5 as eluent afforded the desired compound in a yield of 27%, 19 mg.
  • the compound is prepared by reaction of 3- (2-methoxy-6- (3-methoxyphenyl) naphthalen-1-yl) -N- (5-methyl-1,3,4-thiadiazol-2-yl) -benzenesulfonamide (0, 28 mmol, 1 eq) with boron tribromide solution (2.8 ml, 2.8 mmol, 10 eq), according to Method G.
  • a column chromatographic purification with dichloromethane / methanol 95/5 as eluent afforded the desired compound in a yield of 47%, 64 mg.
  • the compound was prepared by refluxing 3- (2-methoxy-6- (3-methoxyphenyl) naphthalen-1-yl) -N- (methylsulfonyl) benzamide (80 mg, 0.17 mmol, 1 eq) with pyridinium hydrochloride ( 302 mg, 15 eq). After cooling, 2 ml of 1N HCl was added and the water phase extracted with ethyl acetate. The organic phase was dried and concentrated in vacuo. After purification by column chromatography with 10% methanol in dichloromethane, the desired compound was obtained in 42% yield, 31 mg.
  • reaction mixtures for the preparation of compound 75, 77, 81 were according to this The procedure was as follows: To scavenge excess acid chloride, a spatula tip was polymer bound tris (2-aminoethyl) amine and a spatula tip of polymer bound isocyanate was used to scavenge the excess amine. After filtration and evaporation, these mixtures were purified by preparative HPLC (Waters fraction Lynx autopurification system, Varian Inertil C18 column 50 ⁇ 21 mm, particle size 3 ⁇ m, gradient with isocratic end period, solvent: acetonitrile, water, formic acid (0.01%). 0 - 100%).
  • preparative HPLC Waters fraction Lynx autopurification system, Varian Inertil C18 column 50 ⁇ 21 mm, particle size 3 ⁇ m, gradient with isocratic end period, solvent: acetonitrile, water, formic acid (0.01%). 0 - 100%).
  • the compound was prepared by reaction of 2-methoxy-6- (3-methoxyphenyl) -1- (phenylsulfanyl) naphthalenes (376 mg, 1.01 mmol, 1 eq.) With boron tribromide (8 ml of a 1 M solution in cyclohexane, 8 mmol, 8 eq) according to method G. Yield: 325 mg (94%).
  • 17 ⁇ -HSD1 Assay Assay Recombinant protein (0.1 ⁇ g / ml) obtained from Sf9 insect cells is treated with 20 mM KH 2 PO 4 pH 7.4, 30 nM 3 H-estrone and 1 mM NADPH for 30 min at RT incubated in the presence of the potential inhibitor at a concentration of 1 ⁇ M or 100 nM.
  • the stock solutions of compounds are prepared in DMSO (dimethylsulfoxide) such that the final concentration of DMSO in the sample is 1%. After the prescribed incubation time, the reaction is stopped by addition of trichloroacetic acid (10% final concentration).
  • the samples are centrifuged in microtiter plates at 400 rpm for 10 min and the supernatants are added to a reverse phase HPLC equipped with a Waters Symmetry C18 column and Waters Sentry Guard column.
  • the isocratic HPLC run is carried out at RT at a flow rate of 1 ml / min and acetonitrile / water 48:52 as eluent. Radioactivity was measured by Packard Flow Scintillation Analyzer. The total radioactivity of estrone and estradiol was calculated using the formula: cpm estradiol in sample with inhibitor
  • 17ß-HSD2 Assay Assay Recombinant protein (0.1 ⁇ g / ml) obtained from Sf9 insect cells is treated with 20 mM KH 2 PO 4 pH 7.4, 30 nM 3 H-estradiol and 1 mM NAD + for 30 min at RT incubated in the presence of the potential inhibitor at a concentration of 1 ⁇ M or 100 nM.
  • the stock solutions of compounds are prepared in DMSO so that the final concentration of DMSO in the samples is 1%. After the prescribed incubation time, the reaction is stopped by addition of trichloroacetic acid (10% final concentration).
  • the samples are centrifuged in microtiter plates at 400 rpm for 10 min and the supernatants are placed on a reverse phase HPLC equipped with a Waters Symmetry C18 column and Waters Sentry Guard column.
  • the isocratic HPLC run is performed at RT at a flow rate of 1 ml / min and acetonitrile / water 48:52 as eluent. Radioactivity was measured by Packard Flow Scintillation Analyzer. The percent inhibition was calculated by the formulas given above. The results are summarized in table.
  • Estrogen Receptor Assay The percent binding of the compounds to the estrogen receptor ⁇ and ⁇ were determined according to the method described by Zimmermann et al. (Zimmermann, J. et al., J. Steroid Biochem., Mol. Biol., 94: 57-66 (2005)). Slight changes were made, binding incubation was carried out for 2 h at RT with shaking and, after addition of hydroxylapaptite (HAP), incubated on ice for 15 min and vortexed every 5 min. The results are summarized in Table 2.
  • IC TM Values The determination of the IC 50 values was carried out with enzyme isolated from placenta (Luu-The, V. et al., J. Steroid Biochem., Mol. Biol., 55: 581- 587 (1995); Sam, KM et al., Drug Des. Discov., 15: 157-180 (1997)).
  • the microsomal fraction obtained by working-up contains the enzyme 17ß-HSD2, while in the cytosolic fraction the 17ß-HSDl required for the test can be found.
  • the absorptive and secretory permeabilities (Papp (ab) and Papp (ba)) were determined. Erythromycin was used as a substrate and verapamil as an inhibitor of P-gp. Each experiment was performed three times. The integrity of the monolayer was determined by means of TEER (transepithelial electrical resistance) and for each assay the permeability using Lucifer Yellow. All samples from the Caco-2 transport experiments were analyzed by LC / MS / MS after dilution with buffer (1: 1 with 2% acetic acid).
  • the apparent The permeability coefficient (P app ) was calculated by the following formula (1), wherein dQ / dt reflects the recovery rate of the mass in the acceptor compartment, A the surface of the transwell membrane, and Co the initial concentration in the donor compartment. The results are summarized in Table 4.
  • Acetonitrile which was diluted with a mixture of acetonitrile and 10 mM ammonium acetate / 0.1% formic acid (1: 1, V: V) to a final concentration of 10 ug / ml.
  • Fill scan mass spectra were recorded in positive mode. Characteristic fragments were recorded using the following parameters: 350 ° C ion source temperature, 3.8 kV capillary tension, 0.8 mbar argon.
  • Metabolic Stability The stock solutions (10mM in ACN) are diluted to give working concentrations in 20% ACN which are 10 fold higher than the incubation concentrations of the compounds.
  • the assay was performed on rat liver microsomes.
  • the incubation solution (180 ⁇ l) consists of 90 ⁇ l of a microsomal suspension of 0.33 mg / ml protein in phosphate buffer 100 mM pH 7.4 with 90 ⁇ l NADP-regenerating system (NADP: 1 mM, glucose-6-phosphate 5 mM, Glucose-6-phosphate dehydrogenase: 5 U / ml, MgCl 2 5 mM).
  • the reaction is started by adding 20 ⁇ l of the compound to be tested in 20% ACN to the 37 ° C preincubated microsome / buffer mixture. 200 ⁇ l of sample solution are removed after 0, 15, 30 and 60 minutes and subjected to ACN precipitation. Isolate the compounds by adding 200 ⁇ l ACN containing the internal standard (1 ⁇ M) to 200 ⁇ l sample solution and calibration standard. After shaking for 10 sec and centrifugation at 4000 g, an aliquot of the supernatant is added to the LC-MS / MS. Two controls are run, a positive control with 7-ethoxycoumarin as a reference to control microsomal enzyme activity, and a negative control using microsomes heated for 25 minutes without regenerating system to ensure that Substance loss actually goes back to metabolicization.
  • P450 inhibition assav The assay is performed in 96-well plates.
  • the compounds to be tested are dissolved in acetonitrile in a conc. Of 50 mM, whereby the solubility of the compounds can be increased by adding 3% (v / v) 1 N HCl if necessary.
  • the 50 mM stock solution in the first well is diluted 50-fold, followed by a 1: 3 dilution of well 1 to well 8.
  • CYP1A2 a 1 mM stock solution is used.
  • a cofactor mix consisting of an NADP + regenerating system is used. and a cofactor-acetonitrile mix. The final concentration of acetonitrile is 1%.
  • the reaction is started by adding the preheated enzyme / substrate mixture. Incubation of 200 ul / well at 37 0 C for 15 min. The reaction is stopped by adding 75 ⁇ l / well of stop solution consisting of 60% acetonitrile and 40% 0.1 M Tris, pH 9.
  • Table 8 IC 50 values [ ⁇ M] of the test compounds and control inhibitors for the CYP enzymes
  • Concentration ti / 2z half-life (determined from the slope of the falling part of the concentration-time curve AUCo-tz area under the concentration-time curve up to the time t z

Landscapes

  • Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Endocrinology (AREA)
  • Diabetes (AREA)
  • Reproductive Health (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Pyridine Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Indole Compounds (AREA)
  • Heterocyclic Compounds Containing Sulfur Atoms (AREA)
  • Quinoline Compounds (AREA)
  • Thiazole And Isothizaole Compounds (AREA)
  • Nitrogen- Or Sulfur-Containing Heterocyclic Ring Compounds With Rings Of Six Or More Members (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Steroid Compounds (AREA)
EP08735534A 2007-03-27 2008-03-27 17beta-hydroxysteroid-dehydrogenase-typ1-inhibitoren zur behandlung hormonabhängiger erkrankungen Withdrawn EP2131826A2 (de)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DE102007015169A DE102007015169A1 (de) 2007-03-27 2007-03-27 17Beta-Hydroxysteroid-Dehydrogenase-Typ1-Inhibitoren zur Behandlung hormonabhängiger Erkrankungen
PCT/EP2008/053672 WO2008116920A2 (de) 2007-03-27 2008-03-27 17beta-hydroxysteroid-dehydrogenase-typ1-inhibitoren zur behandlung hormonabhängiger erkrankungen

Publications (1)

Publication Number Publication Date
EP2131826A2 true EP2131826A2 (de) 2009-12-16

Family

ID=39514644

Family Applications (1)

Application Number Title Priority Date Filing Date
EP08735534A Withdrawn EP2131826A2 (de) 2007-03-27 2008-03-27 17beta-hydroxysteroid-dehydrogenase-typ1-inhibitoren zur behandlung hormonabhängiger erkrankungen

Country Status (5)

Country Link
US (1) US8546392B2 (uk)
EP (1) EP2131826A2 (uk)
JP (1) JP2010523484A (uk)
DE (1) DE102007015169A1 (uk)
WO (1) WO2008116920A2 (uk)

Families Citing this family (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090142832A1 (en) 2007-11-29 2009-06-04 James Dalton Indoles, Derivatives, and Analogs Thereof and Uses Therefor
KR101648377B1 (ko) * 2008-10-15 2016-08-16 깃세이 야쿠힌 고교 가부시키가이샤 축합환 유도체 및 그 의약용도
EP2440053A4 (en) 2009-06-09 2012-10-31 California Capital Equity Llc BENZYL SUBSTITUTED TRIAZINE DERIVATIVES AND THERAPEUTIC APPLICATIONS THEREOF
ES2557453T3 (es) 2009-06-09 2016-01-26 Nantbioscience, Inc. Derivados de isoquinolina, quinolina y quinazolina como inhibidores de la señalización de Hedgehog
CN102573480B (zh) 2009-06-09 2015-06-10 加利福尼亚资本权益有限责任公司 三嗪衍生物及其治疗应用
EP2624695B1 (en) * 2010-10-08 2015-09-23 Nivalis Therapeutics, Inc. Novel substituted quinoline compounds as s-nitrosoglutathione reductase inhibitors
AU2015224464B2 (en) * 2010-10-08 2016-12-15 Laurel Therapeutics Ltd Novel substituted quinoline compounds as S-nitrosoglutathione reductase inhibitors
EP2651223A4 (en) * 2010-12-16 2015-07-22 Nivalis Therapeutics Inc NOVEL BICYCLIC AROMATIC COMPOUNDS SUBSTITUTED AS INHIBITORS OF S-NITROSOGLUTATHION REDUCTASE
KR101961973B1 (ko) 2011-09-29 2019-03-25 미쓰이가가쿠 아그로 가부시키가이샤 3,4-디히드로이소퀴놀린 유도체의 제조방법 및 그 제조중간체
KR101401002B1 (ko) * 2012-04-05 2014-05-30 부산대학교 산학협력단 Hs1792 또는 이의 약학적으로 허용되는 염을 포함하는 골량 저하 관련 질환의 예방 또는 치료용 약학 조성물 및 건강기능식품
PT2841428T (pt) 2012-04-24 2018-11-29 Vertex Pharma Inibidores de adn-pk
AU2014216178B2 (en) 2013-02-15 2018-06-28 KALA BIO, Inc. Therapeutic compounds and uses thereof
US9688688B2 (en) 2013-02-20 2017-06-27 Kala Pharmaceuticals, Inc. Crystalline forms of 4-((4-((4-fluoro-2-methyl-1H-indol-5-yl)oxy)-6-methoxyquinazolin-7-yl)oxy)-1-(2-oxa-7-azaspiro[3.5]nonan-7-yl)butan-1-one and uses thereof
ES2831625T3 (es) 2013-02-20 2021-06-09 Kala Pharmaceuticals Inc Compuestos terapéuticos y sus usos
EP3985003B1 (en) 2013-03-12 2023-08-09 Vertex Pharmaceuticals Incorporated Dna-pk inhibitors
CN103193601A (zh) * 2013-04-11 2013-07-10 山东大学 2-苯基萘衍生物及其在制备抗肿瘤药物中的应用
SI3057964T1 (sl) 2013-10-14 2020-03-31 Eisai R&D Management Co., Ltd. Selektivno substituirane spojine kinolina
JP6483666B2 (ja) 2013-10-14 2019-03-13 エーザイ・アール・アンド・ディー・マネジメント株式会社 選択的に置換されたキノリン化合物
CN105814036B (zh) 2013-10-17 2018-10-26 沃泰克斯药物股份有限公司 作为dna-pk抑制剂的共晶
US9890173B2 (en) 2013-11-01 2018-02-13 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
JP6426194B2 (ja) 2013-11-01 2018-11-21 カラ ファーマシューティカルズ インコーポレイテッド 治療用化合物の結晶形態及びその使用
US9884839B2 (en) 2014-01-03 2018-02-06 Elexopharm Gmbh Inhibitors of 17Beta-hydroxysteroid dehydrogenases type 1 and type 2
US10399946B2 (en) 2015-09-10 2019-09-03 Laurel Therapeutics Ltd. Solid forms of an S-Nitrosoglutathione reductase inhibitor
CN106187881B (zh) * 2016-07-07 2019-04-23 河南大学 一类具有3-(2-羟基苯基)喹啉结构单元的化合物、其制备方法及应用
BR112019004463A2 (pt) 2016-09-08 2019-05-28 Kala Pharmaceuticals Inc formas cristalinas de compostos terapêuticos, seus processos de obtenção e seus métodos de uso
EP3509422A4 (en) 2016-09-08 2020-05-20 Kala Pharmaceuticals, Inc. CRYSTALLINE FORMS OF THERAPEUTIC COMPOUNDS AND USES THEREOF
US10392399B2 (en) 2016-09-08 2019-08-27 Kala Pharmaceuticals, Inc. Crystalline forms of therapeutic compounds and uses thereof
KR20190062485A (ko) 2016-09-27 2019-06-05 버텍스 파마슈티칼스 인코포레이티드 Dna-손상제 및 dna-pk 저해제의 조합을 사용한 암 치료 방법
CN108727262A (zh) * 2018-04-12 2018-11-02 苏州康润医药有限公司 一种3-溴-7-甲氧基喹啉的合成方法

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1181485A (en) * 1914-12-04 1916-05-02 Hoechst Ag Oxyphenylquinolin-dicarboxylic acid and process of making same.
US5658931A (en) 1994-09-20 1997-08-19 Eli Lilly And Company Method for inhibiting mammalian breast carcinoma with tamoxifen, and analogs thereof, and certain naphthyl compounds
JP4166433B2 (ja) * 1997-09-11 2008-10-15 第一製薬株式会社 ホルモン依存性疾患治療剤
TWI306450B (en) 2001-12-13 2009-02-21 Wyeth Corp Substituted phenyl naphthalenes as estrogenic agents
DE10228103A1 (de) 2002-06-24 2004-01-15 Bayer Cropscience Ag Fungizide Wirkstoffkombinationen
BR0203724B1 (pt) * 2002-09-12 2011-08-09 bomba de fluidos e placa de transferência de fluidos e sensor indutivo para bomba de fluidos.
US7465739B2 (en) 2003-06-10 2008-12-16 Solvay Pharmaceuticals B.V. Compounds and their use in therapy
US7754709B2 (en) 2003-06-10 2010-07-13 Solvay Pharmaceuticals Bv Tetracyclic thiophenepyrimidinone compounds as inhibitors of 17β hydroxysteroid dehydrogenase compounds
US7563748B2 (en) 2003-06-23 2009-07-21 Cognis Ip Management Gmbh Alcohol alkoxylate carriers for pesticide active ingredients
TWI390006B (zh) 2003-08-07 2013-03-21 Nippon Steel Chemical Co Organic EL materials with aluminum clamps
TWI331154B (en) * 2003-11-12 2010-10-01 Solvay Pharm Gmbh Novel 17-hydroxysteroid dehydrogenase type i inhibitors
US20050228038A1 (en) 2004-04-08 2005-10-13 Vander Jagt David L 11-Beta hydroxysteroid dehydrogenase type 1 inhibitors as anti-obesity/anti-diabetes compounds and 17-beta hydrosteroid dehydrogenase type I inhibitors as useful agents for the treatment of cancers, especially breast cancer
RU2346996C2 (ru) 2004-06-29 2009-02-20 ЮРОПИЭН НИКЕЛЬ ПиЭлСи Усовершенствованное выщелачивание основных металлов
DE102004032673A1 (de) 2004-07-02 2006-01-26 Schering Ag Neue 2-substituierte D-Homo-estra-1,3,5(10)-triene als Inhibitoren der 17ß-Hydroxysteroiddehydrogenase Typ 1
EP1838296B1 (en) 2004-10-20 2012-08-08 Resverlogix Corp. Flavanoids and isoflavanoids for the prevention and treatment of cardiovascular diseases
DE602006020138D1 (de) 2005-06-29 2011-03-31 Compumedics Ltd Sensoranordnung mit leitfähiger brücke
UA94921C2 (en) * 2005-12-08 2011-06-25 Новартис Аг 1-orthofluorophenyl substituted 1, 2, 5-thiazolidinedione derivatives as ptp-as inhibitors
AR059574A1 (es) 2006-02-14 2008-04-16 Wyeth Corp Formulaciones farmaceuticas acuosas de ligandos selectivos er-beta

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2008116920A2 *

Also Published As

Publication number Publication date
US20100204234A1 (en) 2010-08-12
US8546392B2 (en) 2013-10-01
WO2008116920A3 (de) 2009-04-02
JP2010523484A (ja) 2010-07-15
DE102007015169A1 (de) 2008-10-02
WO2008116920A2 (de) 2008-10-02

Similar Documents

Publication Publication Date Title
EP2131826A2 (de) 17beta-hydroxysteroid-dehydrogenase-typ1-inhibitoren zur behandlung hormonabhängiger erkrankungen
DE102007040243A1 (de) 17Beta-Hydroxysteriod-Dehydrogenase Typ1 Inhibitoren zur Behandlung hormonabhängiger Erkrankungen
MX2008006885A (es) Agentes de enlace de receptores nucleares.
RU2572244C2 (ru) Ингибиторы ароматазы
US20090186900A1 (en) Compound
CN102020563A (zh) 新的姜黄素类似物及其用途
CN103570625A (zh) N-(3-杂芳基芳基)-4-芳基芳基甲酰胺和类似物作为Hedgehog通路抑制剂及其应用
TW200848049A (en) Pyrimidine derivatives
El-Kabbani et al. Inhibitors of human 20α-hydroxysteroid dehydrogenase (AKR1C1)
EP3919489A1 (en) Aromatic ring or heteroaromatic ring compounds, preparation method therefor and medical use thereof
EP2609089A1 (en) Selective 17beta-hydroxysteroid dehydrogenase type 1 inhibitors
EP3091005B1 (en) 1,2-naphthoquinone derivative and method for preparing same
Krzystyniak Current strategies for anticancer chemoprevention and chemoprotcction
WO2011095807A1 (en) Combinations of mek and hh inhibitors
CN110467616B (zh) 含杂芳基取代哒嗪酮结构的三唑并吡嗪类化合物的制备及应用
WO2015101670A2 (en) Inhibitors of 17beta-hydroxysteroid dehydrogenases type 1 and type 2
JP2018516971A (ja) ベータナフトイソフラボン、これを含有する組成物及びその使用
CN114456143B (zh) 一种uPA的抑制剂及其制备方法和应用
JP6038037B2 (ja) キナーゼ活性を増加させるための化合物とその応用
CN101402587A (zh) 取代乙酰肼类衍生物及其制备方法和用途
CN107709325B (zh) 用作Lp-PLA2抑制剂的双环类化合物、其制备方法及医药用途
KR101634758B1 (ko) 에스트로겐 수용체 리간드로서의 2-하이드록시-7-메틸 옥타하이드로페난트렌 유도체 또는 이의 약학적으로 허용가능한 염을 유효성분으로 함유하는 약학 조성물
Oster 3.3 Bicyclic substituted Hydroxyphenylmethanone Type Inhibitors of 17β-Hydroxysteroid Dehydrogenase Type 1 (17β-HSD1): The Role of the Bicyclic Moiety

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20090929

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

RIN1 Information on inventor provided before grant (corrected)

Inventor name: ZIEGLER, ERIKA

Inventor name: HARTMANN, ROLF

Inventor name: FROTSCHER, MARTIN

Inventor name: OBERWINKLER, SANDRINE

Inventor name: THOLE, HEINRICH-HUBERT

Inventor name: MESSINGER, JOSEF

17Q First examination report despatched

Effective date: 20120309

DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20140624