WO2018216821A1 - Use of ascochlorin derivative for combination therapy - Google Patents

Use of ascochlorin derivative for combination therapy Download PDF

Info

Publication number
WO2018216821A1
WO2018216821A1 PCT/JP2018/020870 JP2018020870W WO2018216821A1 WO 2018216821 A1 WO2018216821 A1 WO 2018216821A1 JP 2018020870 W JP2018020870 W JP 2018020870W WO 2018216821 A1 WO2018216821 A1 WO 2018216821A1
Authority
WO
WIPO (PCT)
Prior art keywords
hydroxy
chloro
dien
methylpenta
methyl
Prior art date
Application number
PCT/JP2018/020870
Other languages
French (fr)
Inventor
Tatsuo Hoshino
Nobuo Shimma
Original Assignee
Nrl Pharma, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nrl Pharma, Inc. filed Critical Nrl Pharma, Inc.
Publication of WO2018216821A1 publication Critical patent/WO2018216821A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/15Oximes (>C=N—O—); Hydrazines (>N—N<); Hydrazones (>N—N=) ; Imines (C—N=C)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C47/00Compounds having —CHO groups
    • C07C47/52Compounds having —CHO groups bound to carbon atoms of six—membered aromatic rings
    • C07C47/575Compounds having —CHO groups bound to carbon atoms of six—membered aromatic rings containing ether groups, groups, groups, or groups
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C49/00Ketones; Ketenes; Dimeric ketenes; Ketonic chelates
    • C07C49/587Unsaturated compounds containing a keto groups being part of a ring
    • C07C49/703Unsaturated compounds containing a keto groups being part of a ring containing hydroxy groups
    • C07C49/747Unsaturated compounds containing a keto groups being part of a ring containing hydroxy groups containing six-membered aromatic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/155Amidines (), e.g. guanidine (H2N—C(=NH)—NH2), isourea (N=C(OH)—NH2), isothiourea (—N=C(SH)—NH2)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C221/00Preparation of compounds containing amino groups and doubly-bound oxygen atoms bound to the same carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C239/00Compounds containing nitrogen-to-halogen bonds; Hydroxylamino compounds or ethers or esters thereof
    • C07C239/08Hydroxylamino compounds or their ethers or esters
    • C07C239/14Hydroxylamino compounds or their ethers or esters having nitrogen atoms of hydroxylamino groups further bound to carbon atoms of hydrocarbon radicals substituted by doubly-bound oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C239/00Compounds containing nitrogen-to-halogen bonds; Hydroxylamino compounds or ethers or esters thereof
    • C07C239/08Hydroxylamino compounds or their ethers or esters
    • C07C239/20Hydroxylamino compounds or their ethers or esters having oxygen atoms of hydroxylamino groups etherified
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C239/00Compounds containing nitrogen-to-halogen bonds; Hydroxylamino compounds or ethers or esters thereof
    • C07C239/08Hydroxylamino compounds or their ethers or esters
    • C07C239/22Hydroxylamino compounds or their ethers or esters having oxygen atoms of hydroxylamino groups esterified
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/02Systems containing only non-condensed rings with a three-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/08Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms
    • C07D295/084Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms with the ring nitrogen atoms and the oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/088Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by singly bound oxygen or sulfur atoms with the ring nitrogen atoms and the oxygen or sulfur atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings to an acyclic saturated chain
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D305/00Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms
    • C07D305/02Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms not condensed with other rings
    • C07D305/04Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D305/08Heterocyclic compounds containing four-membered rings having one oxygen atom as the only ring hetero atoms not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring atoms

Definitions

  • the present invention is directed to a combination therapy using an ascochlorin derivative with metformin for use in the treatment or prevention of diseases or conditions that involve AMPK dysregulation, such as diabetes, obesity, and hyperlipidemia.
  • the invention is also directed to a pharmaceutical composition comprising an ascochlorin derivative for use in combination with administration of metformin, for the treatment or prevention of diseases or conditions that involve AMPK dysregulation such as diabetes, obesity, and hyperlipidemia.
  • AMPK adenosine monophosphate-activated protein kinase
  • AMPK is also known to have critical roles in regulating cell growth and autophagy through inhibition of m-TOR, as well as mitochondrial biogenesis.
  • AMP or ADP can directly bind to the ⁇ regulatory subunit of AMPK, leading to a conformational change that promotes AMPK phosphorylation at Thrl72 by LKB 1 (known as a tumor suppressor) or C AMKK2 (Ca ++ dependent kinase). Further, the conformational change also protects AMPK from dephosphorylation by protein phosphatases 2A and C so that AMPK remains activated.
  • the dysregulation of AMPK is considered as a pathogenic factor for the diseases such as cancer, type 2 diabetes, metabolic syndrome-associated diseases (dyslipidemia, adiposity, atherosclerotic cardiovascular disease, hypertension, and nonalcoholic fatty liver disease (NAFLD)), inflammation and Alzheimer disease, and thus AMPK is considered as a target for the prevention and therapy of these diseases [metabolic syndrome: Science in Medicine, 2013, Vol. 123 (7) page 2764, Molecular and Cellular Endocrinology, 2013, vol. 366 page 135; cancer and inflammation: Oncotarget, 2015, vol. 6 (10), page 7365; cardiovascular disease: Circ Res., 2009 vol. 104 (3), page 282; inflammation,
  • diseases such as cancer, type 2 diabetes, metabolic syndrome-associated diseases (dyslipidemia, adiposity, atherosclerotic cardiovascular disease, hypertension, and nonalcoholic fatty liver disease (NAFLD)
  • NAFLD nonalcoholic fatty liver disease
  • Alzheimer disease J Alzheimers Dis. 2009, vol. 16(4) page 731].
  • Metformin is known as an antihyperglycemic agent to indirectly activate AMPK through inhibition of respiratory chain complex 1 [Clinical Science, 2012, vol. 122, page 253]. Metformin, however, is reported to activate AMPK in HepG2 cell only at surprisingly high concentration of mM level for in vitro cellular experiment.
  • AICAR 5-Aminoimidazole-4-carboxamide ribonucleotide
  • AICAR directly activates AMPK through binding to a regulatory ⁇ -subunit to promote phosphorylation of AMPK and to protect AMPK from dephosphorylation.
  • A-769662 is reported to be a direct AMPK- activator with a different binding mode, [Cell Metabolism, 2006, vol. 3, page 403].
  • AMPK-activators reported in literatures, but none of them is clinically used up to now, partly because of the insufficient in vivo efficacy and/or safety profile. Therefore, novel AMPK activators with good efficacy and safety profile would provide therapeutic benefits to the patients with diseases that involve AMPK dysregulations.
  • MAC 4-O-Methy-ascochlorin
  • Patent Document 1 JP H06-305959 A
  • Patent Document 2 WO 2004/074236
  • Non-Patent Document 1 Physiological Rev. 2009, vol. 89, page 1025,
  • Non-Patent Document 2 Nature Reviews/Molecular Cell Biology, 2012, vol.13 page 251, .
  • Non-Patent Document 3 Nature Cell Biology, 2011 , vol.13 (9), 1016,
  • Non-Patent Document 4 Molecular and Cellular Endocrinology, 2013 vol. 366, page 152
  • Non-Patent Document 5 J. Physiol., 2006, vol 574.1 page 33
  • Non-Patent Document 6 Science in Medicine, 2013 , 123 (7), 2764;
  • Non-Patent Document 7 Mol. Cell. Endocrinol., 2013, 366; 135;
  • Non-Patent Document 8 Oncotarget, 2015, 6 (10), 7365;
  • Non-Patent Document 9 Circ Res., 2009, 104 (3), page 282;
  • Non-Patent Document 10 Diabetologia, 2010, 53, 2406;
  • Non-Patent Document 11 Am. J. Respir. Cell Mol. Biol., 2010, 42, 676;
  • Non-Patent Document 12 J Alzheimers Dis. 2009, 16(4), 731;
  • Non-Patent Document 13 Clinical Science, 2012, 122, 253;
  • Non-Patent Document 14 Cell Metabolism, 2006, 3, 403.
  • Non-Patent Document 15 Biochemical and Biophysical Research Communications, 2011, vol.
  • the present inventors produced ascochlorin derivatives, which are confirmed to have AMPK activation profile, and significantly improved metabolic stability as compared with MAC (PCT application: PCT/JP2017/000902). Particularly, the inventors found that the ascochlorin derivatives also show good in vivo efficacy in several disease models especially for oncology and metabolic diseases. On the other hand, there are strong needs for measures to treat or prevent a certain number of diseases including diabetes, obesity, and hyperlipidemia.
  • the present inventors have conducted diligent studies and consequently completed the present invention by finding that a combination of an ascochlorin derivative with metformin has an excellent effect for treating or preventing diabetes, obesity, and hyperlipidemia.
  • the present invention is directed to a combination therapy using an ascochlorin derivative with metformin for treating or preventing a disease that involves AMPK dysregulation.
  • the invention is also directed to a pharmaceutical composition comprising an ascochlorin derivative for use in combination with administration of metformin for the treatment or prevention of diseases or conditions that involve AMPK dysregulation.
  • a pharmaceutical composition comprising a compound represented by formula (I):
  • R 2 is C 1-6 alkyl, in which the alkyl may be substituted with one to five fluorine atoms;
  • R 3 is a hydrogen atom or Ci-6 alkyl
  • R 4 is hydroxy, €].& alkylamino, C3-7 cycloalkylamino, or 3- to 7-membered
  • heterocycloalkylamino which contains -0-, -S-, -NR 6 -, -SO- or -SO2- as a ring atom, or
  • Z is a hydrogen atom, Ci-e alkyl, -CO(CH 2 ) n -R 5 or -(CH 2 ) complicat-R 5 ;
  • n is an integer selected from 1 to 4.
  • R 5 is -CO2R 6 , -CONR 7 R 8 , -OCONR 7 R 8 , -S0 2 NR 7 R 8 , -S0 2 R 9 , hydroxy, -NHSO2R 9 , or -
  • R 6 , R 7 and R 8 are each independently selected from a group consisting of a hydrogen atom, Ci- 6 alkyl, and C3-7 cycloalkyl; or R 7 and R 8 together with the nitrogen atom to which they are attached form a nitrogen containing heterocyclic ring which may further contain -0-, -S-, -NR 6 -, -SO- or -S0 2 - as a ring atom, in which the heterocyclic ring may be substituted with one or more substituents selected from hydroxy and Ci-e alkyl,
  • R 9 is Ci-6 alkyl, or C3-7 cycloalkyl
  • composition is for use in combination with administration of metformin.
  • composition according to any one of ( 1 - 1 ) to ( 1 -7), wherein the composition is for use is treating or preventing a disease selected from diabetes, obesity, and hyperlipidemia.
  • R 2 is Ci-6 alkyl, in which the alkyl may be substituted with one to five fluorine atoms;
  • R 3 is a hydrogen atom or Ci-6 alkyl
  • R 4 is hydroxy, Ci-6 alkylamino, C3-7 cycloalkylamino, or 3- to 7-membered
  • heterocycloalkylamino which contains -0-, -S-, -NR 6 -, -SO- or -SO2- as a ring atom, or
  • Z is a hydrogen atom, Ci- 6 alkyl, -CO(CH 2 ) n -R 5 or -(CH 2 ) n -R 5 ;
  • n is an integer selected from 1 to 4.
  • R 5 is -CO2R 6 , -CONR 7 R 8 , -OCONR 7 R 8 , -S0 2 NR 7 R 8 , -SO2R 9 , hydroxy, -NHSO2R 9 , or -
  • R 6 , R 7 and R 8 are each independently selected from a group consisting of a hydrogen atom, Ci- 6 alkyl, and C3_7 cycloalkyl; or R 7 and R 8 together with the nitrogen atom to which they are attached form a nitrogen containing heterocyclic ring which may further contain -0-, -S-, -NR 6 -, -SO- or -S0 2 - as a ring atom, in which the heterocyclic ring may be substituted with one or more substituents selected from hydroxy and Ci-6 alkyl,
  • R 9 is Ci-6 alkyl, or C3-7 cycloalkyl; a pharmaceutically acceptable salt or a solvate thereof.
  • composition according to any one of (2-1) to (2-7), wherein the composition is for use is treating or preventing a disease selected from diabetes, obesity, and hyperlipidemia.
  • a method for treating or preventing a disease that involves AMPK dysregulation comprising:
  • R 2 is G-6 alkyl, in which the alkyl may be substituted with one to five fluorine atoms;
  • R 3 is a hydrogen atom or G-6 alkyl
  • R 4 is hydroxy, G-6 alkylamino, C3-7 cycloalkylamino, or 3- to 7-membered
  • heterocycloalkylamino which contains -0-, -S-, -NR 6 -, -SO- or -SO2- as a ring atom, or
  • Z is a hydrogen atom, G. 6 alkyl, -CO(CH 2 ) n -R 5 or -(CH 2 ) felicit-R 5 ;
  • n is an integer selected from 1 to 4.
  • R 5 is -C0 2 R 6 , -CONR 7 R 8 , -OCONR 7 R 8 , -S0 2 NR 7 R 8 , -S0 2 R 9 , hydroxy, -NHSO2R 9 , or -
  • R 6 , R 7 and R 8 are each independently selected from a group consisting of a hydrogen atom, G- 6 alkyl, and C3-7 cycloalkyl; or R 7 and R 8 together with the nitrogen atom to which they are attached form a nitrogen containing heterocyclic ring which may further contain -0-, -S-, -NR 6 -, -SO- or -SO2- as a ring atom, in which the heterocyclic ring may be substituted with one or more substituents selected from hydroxy and G-6 alkyl,
  • R 9 is G-6 alkyl, or C3-7 cycloalkyl; a pharmaceutically acceptable salt or a solvate thereof, in combination with administering a therapeutically effective amount of metformin.
  • the pharmaceutical composition is useful in treating or preventing a disease selected from diabetes, obesity, hyperlipidemia, cardiovascular diseases, and metabolic syndrome-associated diseases, particularly diabetes, obesity, and hyperlipidemia, more particularly type-2 diabetes.
  • the compound of the present invention is useful as a preventive or therapeutic agent for the disease in which AMPK is dysregulated; such as metabolic syndrome, type 2 diabetes, dyslipidemia, adiposity, atherosclerotic cardiovascular disease, hypertension, and nonalcoholic fatty liver disease
  • the pharmaceutical composition of the present invention is used for preventing or treating a disease in which
  • AMPK is dysregulated; such as metabolic syndrome, type 2 diabetes, dyslipidemia, adiposity, atherosclerotic cardiovascular disease, hypertension, and nonalcoholic fatty liver disease (NAFLD), inflammation and Alzheimer disease.
  • metabolic syndrome such as metabolic syndrome, type 2 diabetes, dyslipidemia, adiposity, atherosclerotic cardiovascular disease, hypertension, and nonalcoholic fatty liver disease (NAFLD), inflammation and Alzheimer disease.
  • NAFLD nonalcoholic fatty liver disease
  • Figure 1 shows the results of western blot analysis to indicate AMPK activation and downstream signal, ACC in HepG2 cell.
  • Figure 2 shows pharmacokinetic profile of Compound 19 in SD rats.
  • Figure 3 shows the results of assay indicating the effect of Compound 5 on the level of triglyceride in blood of HFD-STZ model mice.
  • Figure 4 shows the results of assay indicating the effect of the compounds of formula (I) on the level of total cholesterol in blood of mice.
  • Figure 5 shows the results of assay indicating the effect of Compound 5 on the blood glucose level of mice (HFD-STZ model) in a fasting state.
  • Figure 6 shows the results of oral glucose tolerant tests conducted on Day 0 (baseline) in Test Example 7..
  • Figure 7 shows the results of oral glucose tolerant tests conducted on Day 28 (endpoint) in Test Example 7.
  • Figure 8 shows fasting blood glucose levels measured in Test Example 7.
  • Figure 9 shows serum triglyceride levels measured in Test Example 7.
  • Figure 10 shows hemoglobin Ai c levels measured in Test Example 7.
  • Ci-6 alkyl refers to a linear or branched saturated hydrocarbon group having 1 to 6 carbon atoms. Examples thereof include methyl, ethyl, n-propyl, iso- propyl, butyl, iso-butyl, tert-butyl, n-pentyl, pentan-3-yl and the like.
  • Ci-6 alkylamino means a group: -NH-(Ci-6 alkyl), in which Ci-6 alkyl is as defined above.
  • C3.7 cycloalkyl refers to a saturated carbocyclic group having 3 to 7 carbon atoms. Examples thereof include cyclopropyl (cPr), cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and the like.
  • C3-7 cycloalkylamino means a group: -NH-(C3-7 cycloalkyl), in which "C3-7 cycloalkyl” is as defined above. Examples thereof include cyclopropylamino, cyclobutylamino, cyclopentylamino, cyclohexylamino, cycloheptylamino and the like.
  • heterocycloalkylamino refers to an amino group substituted with a saturated heterocyclic group having 3 to 7 ring atoms, which includes one or more hetero atoms selected from -0-, -S-, -NR. 6 -, -SO- and -SO2-. Examples thereof include pyrrolidinylamino, piperidinylamino, piperazinylamino, N-methylpiperazinylamino, morpholinylamino, thiomorpholinylamino,
  • a nitrogen-containing 3- to 7-membered heterocyclic ring refers to a heterocyclic ring having 3 to 7 ring atoms, which includes at least one nitrogen atom and one or more hetero atoms selected from -0-, -S-, -NR. 6 -, -SO- and -SO2-.
  • heterocyclic ring having 3 to 7 ring atoms, which includes at least one nitrogen atom and one or more hetero atoms selected from -0-, -S-, -NR. 6 -, -SO- and -SO2-. Examples thereof include pyrrolidine, piperidine, piperazine, N-methylpiperazine, morphorine, thiomorpholine, imidazolidine, imidazoline,
  • R 2 is Ci- ⁇ alkyl which may be substituted with 1 to 3 fluorine atoms, for example, CH 3 , CH2CH3, CHF 2 , CH 2 F, CF 3 , and CH2CF3, preferably C3 ⁇ 4 or CH 2 F.
  • R 3 is selected from hydrogen, methyl and ethyl.
  • R 4 is selected from a group consisting of hydroxy, methylamino, ethylamino, cyclopropylamino, oxetan-3-ylamino, (3-morpholinopropyl)amino, pirrolidin-3-ylamino or (3-(2-methlypipelidin-l -yl)propyl)amino.
  • R 4 is selected from hydroxyl, ethylamino, cyclopropylamino, and oxetan-3-ylamino.
  • the compound of formula (I) is selected from the compounds of Nos. (5), (8), (9), (10), (11), (12), (14), (15), (16), (19), (21), (24), (25), (26), (27), (28) indicated above, and particularly preferably a compound selected from the compounds of Nos. (5), (10), (12), (15), (16), (19), (21 ), and (25) indicated above.
  • the compound of formula (I) has activity to increase phosphorylation of AMPK and its downstream effector, acetyl-CoA carboxylase (ACC) [Test Example 1]. Further, the compound has significantly improved metabolic stability against degradation by liver microsome (human and mouse) [Test Example 3], improved PK profile that allows once a day oral treatment [Test Example 4], and improved solubility [Test Example 2] as compared with 4-O-methyl-ascoclorin (MAC) and its prodrug, a Schiff base of MAC with glycine amide (compound #13 in WO2004/074236).
  • ACC acetyl-CoA carboxylase
  • the compound of formula (I) exhibited blood glucose lowering activity as well as anti- hyperlipidemic activity (lowering blood triglyceride) [Test Example 5].
  • the compounds of formula (I) may be administered in the form of a pharmaceutically acceptable salt.
  • pharmaceutically acceptable salt refers to a salt, which is non-toxic and is prepared with a pharmaceutically acceptable base or acid.
  • the pharmaceutically acceptable salt thereof is generally prepared by adding the compound with a suitable organic or inorganic acid.
  • Examples of the acid-addition salt include acetate, benzenesulfonate, benzoate, citrate, fumarate, glutamate, hydrobromide, hydrochloride, lactate, maleate, mandelate, mesylate, oxalate, palmitate, phosphate/diphosphate, salicylate, stearate, sulfate, succinate, tartrate, and tosylate.
  • the pharmaceutically acceptable salt thereof is generally prepared by adding the compound with a suitable organic or inorganic base.
  • suitable organic or inorganic base examples include salts derived from inorganic bases including aluminum, ammonium, calcium, potassium, sodium and the like. Particularly preferred are the ammonium, calcium, potassium, and sodium salts.
  • Examples of the base-addition salt also include salts derived from primary, secondary, and tertiary amines such as arginine, betaine, 2-diethylaminoethanol, 2- dimethylaminoethanol, ethanolamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, lysine, methylglucamine, morpholine, piperazine, piperidine, triethylamine, tripropylamine, and the like.
  • salts derived from primary, secondary, and tertiary amines such as arginine, betaine, 2-diethylaminoethanol, 2- dimethylaminoethanol, ethanolamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, lysine, methylglucamine, morpholine, piperazine, piperidine, triethylamine, trip
  • the compound of formula (I) may be administered as a prodrug.
  • the prodrug can be prepared in line with technical knowledge commonly available to a person skilled in the art.
  • a prodrug can be prepared by converting the carboxy group to a pharmaceutically acceptable ester group, such as methyl, ethyl, or pivaloyloxymethyl ester, or by converting the hydroxy group to a pharmaceutically acceptable ester group, such as methyl, ethyl, or pivaloyloxymethyl ester, or by converting the hydroxy group to a
  • ester group such as acetate or maleate.
  • Use of a prodrug may be beneficial for modifying the solubility or hydrolysis characteristics for use as sustained-release or prodrug formulations.
  • the compound of formula (I) may be used in an amorphous form or a crystalline form.
  • the crystalline form may include solvent to form a solvate such as a hydrate. It should be understood that the present invention covers any use of such a solvate or any mixture such as a solution, suspension or solid mixture containing the compound of formula (I) or metformin.
  • a method for treating or preventing a disease that involves AMP dysregulation which comprises administering to a patient in need thereof a therapeutically effective amount of a compound represented by formula (I) or a pharmaceutically acceptable salt thereof in combination with administration of metformin.
  • the disease that involves AMPK dysregulation is exemplified as described above.
  • An appropriate route of the administration of the compound represented by formula (I) may be determined by a person skilled in the art.
  • Examples thereof include oral, endorectal, parenteral (intravenous, intramuscular, or subcutaneous), intracisternal, vaginal, intraperitoneal, intravesical, topical (drip infusion, powder, ointment, gel, or cream) application, and inhalation (intraoral or nasal spray).
  • the dosage form of the compound represented by formula (I) for the administration includes tablets, capsules, granules, powders, pills, aqueous and non-aqueous solutions or suspensions for oral use, and parenteral solutions loaded into containers.
  • An appropriate route of the administration of metformin may be determined by a person skilled in the art. Examples thereof include oral, endorectal, parenteral (intravenous, intramuscular, or subcutaneous), intracisternal, vaginal, intraperitoneal, intravesical, topical (drip infusion, powder, ointment, gel, or cream) application, and inhalation (intraoral or nasal spray).
  • the dosage form of metformin for the administration includes tablets, capsules, granules, powders, pills, aqueous and nonaqueous solutions or suspensions for oral use, and parenteral solutions loaded into containers.
  • a pharmaceutical composition comprising a compound represented by formula (I) or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient or additive, for use in combination with
  • a pharmaceutical composition comprising metformin and a pharmaceutically acceptable excipient or additive, for use in combination with administration of a compound represented by formula (I) or a pharmaceutically acceptable salt thereof.
  • the composition may be administered in an appropriate route, for example, oral, endorectal, parenteral (intravenous, intramuscular, or subcutaneous), intracisternal, vaginal, intraperitoneal, intravesical, topical (drip infusion, powder, ointment, gel, or cream) application, and inhalation (intraoral or nasal spray).
  • the dosage form for the administration includes tablets, capsules, granules, powders, pills, aqueous and non-aqueous solutions or suspensions for oral use, and parenteral solutions loaded into containers.
  • the pharmaceutical composition may be administered as a pharmaceutical formulation.
  • the pharmaceutical formulation can be produced by well-known methods, using additives such as excipients, coating agents, lubricants, binders, disintegrators, stabilizers, corrigents, diluents, solvent and surfactants or emulsifiers.
  • excipients include starches such as potato starch and cornstarch, lactose, crystalline cellulose, and calcium hydrogen phosphate.
  • coating agents include ethylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, shellac, talc, carnauba wax, and paraffin.
  • Examples of lubricants include magnesium stearate, and stearic acid.
  • binders examples include polyvinylpyrrolidone, macrogol, and the same substances exemplified as the excipients.
  • disintegrators include the same substances exemplified as the excipients and croscarmellose sodium, sodium carboxymethylstarch, and chemically modified starch or celluloses.
  • stabilizers include paraoxybenzoic acid esters such as methylparaben and propylparaben; alcohols such as chlorobutanol, benzylalcohol, and phenylethylalcohol; benzalkonium chloride; phenols such as phenol and cresol; thimerosal; dehydroacetic acid; and ascorbic acid.
  • paraoxybenzoic acid esters such as methylparaben and propylparaben
  • alcohols such as chlorobutanol, benzylalcohol, and phenylethylalcohol
  • benzalkonium chloride phenols such as phenol and cresol
  • thimerosal thimerosal
  • dehydroacetic acid and ascorbic acid.
  • Examples of corrigents include sweeteners which are conventionally used.
  • diluents include the substances exemplified as excipients.
  • solvents examples include ethanol, phenol, chlorocresol, purified water, and distilled water.
  • surfactants or emulsifiers examples include polysorbate 80, polyoxyl 40 stearate, lauromacrogol, soybean oil, Maisin 35-1 (Gattefosse, France) and Cremophor (BASF, Germany).
  • the dose of the compounds of formula (I) or pharmaceutically acceptable salts thereof differs depending on the symptoms, age, body weight, relative health condition, presence of other medication, method of administration, and the like.
  • a generally effective dose for an oral agent as the active ingredient for a patient is a daily dose of preferably 0.-1 to 1000 mg per kg body weight, more preferably 1 to 30 mg per kg body weight.
  • the daily dose for an adult patient with a normal body weight is preferably in the range of 10 to 1000 mg.
  • the daily dose is preferably 0.1 to 1000 mg per kg body weight, more preferably 10 to 800 mg per kg body weight.
  • such a dose is administered at one time or in several portions per day, or intermittent schedule depending on the symptoms.
  • Metformin may be administered, by a route and in an amount that are commonly known therefor, simultaneously or sequentially with a compound of formula (I). When the compound is administered simultaneously with metformin, these ingredients may be contained in a single unit dosage form or two or more separate dosage forms. In the combination therapy, the compound of formula (I) and metformin are administered on different overlapping schedules. It is also contemplated that in the combination therapy, the compounds of formula (I) and metformin may be administered in lower doses than that administered each alone.
  • the dosage of the compounds of formula (I) and metformin used for the subject invention is determined depending on age of a subject, body weight of a subject, symptom, degree of seriousness, therapeutic effect, administration route, administration regimen, and period of the treatment,
  • the compound represented by formula (I) is administered in an amount from 0.1 to 100 mg kg/day, particularly 0.5 to 30 mg/kg/day, more particularly 1 to 16 mg/kg/day.
  • metformin is administered in an amount from 50 to 1000 mg/kg/day, particularly 100 to 500 mg/kg/day, more particularly 250 to 350 mg/kg/day.
  • the compound represented by formula (I) is administered in an amount from 0.1 to 100 mg/kg/day, and metformin is administered in an amount from 50 to 1000 mg/kg/day.
  • the compound represented by formula (I) is administered in an amount from 0.5 to 30 mg/kg/day, and metformin is administered in an amount from 100 to 500 mg/kg/day.
  • the compound represented by formula (I) is administered in an amount from 1 to 16 mg/kg/day, and metformin is administered in an amount from 250 to 350 mg/kg/day.
  • a starting material, ascochlorin can be obtained by fermentation of Ascochyts viciae Libert as described in JP S45-009832 B published in 1970.
  • R 2 , R 3 , R 4 , R 5 , R 7 , Y, and Z are as defined hereinbefore.
  • Scheme l shows a method for producing compound (IV), (Via) or (Vlb).
  • the compound (II) in which R 2 is methyl (4-O-methyl-ascochlorin) can be prepared by the method described in JP H6- 305959 A.
  • the compound (II) in which R 2 is fluorinated alkyl group can be obtained by O-alkylation with a fluoroalkylating agent in the presence of base in an appropriate solvent. Examples of the base include potassium carbonate, cesium carbonate and the like.
  • fluoroalkylating agents include bromofluoromethane, bis(trifluoromethyl)-bis(trifluoromethyloxy)-sulfane, 5-tert-butyl-2'- (trifluoromethoxy)biphenylyl-2-diazonium hexafluoroantimonate, 2,2,2-tris(fluoranyl)ethyl methanesulfonate, 2-diazo- 1 , 1,1 -tris(fluoranyl)ethane, tris(fluoranyl)methane, 1 -chloranyl-4-[chloranyl- bis(fluoranyl)methyl]sulfonyl-benzene and the like.
  • the compound (IV) can be prepared by reacting a compound (II) with NH2OY (III) in the presence of base such as pyridine, sodium carbonate, sodium hydrogencarbonate and the like.
  • the compound (Vlb) can be prepared by reacting compound (IV) with NH2OZ (V) in the presence of base such as pyridine, sodium carbonate, sodium hydrogencarbonate and the like.
  • the compound (Via) can be directly prepared from the compound (II) by reacting two or more equivalents of hydroxyamine (III) in the same reaction condition as described above.
  • Scheme 2 shows a method for producing compound (X).
  • the compound (VIII) in which R 2 is methyl or fluorinated alkyl can be prepared from ascochlorin via O-acetyl derivative (VII) wherein R 2 is methyl or fluorinated alkyl by the method described in JP 2005- 225851 A.
  • the intermediate (IX) can be prepared by reacting the intermediate (VIII) with NH 2 O-Z (V) in the presence of base such as pyridine, sodium carbonate, sodium hydrogencarbonate and the like.
  • the compound (X) can be prepared by acidic hydrolysis of the acetal group of compound (IX) in an appropriate solvent by the method known to a person skilled in the art.
  • Scheme 3 shows another method for producing compound (X).
  • the compound (X) can be prepared from the corresponding oxime derivative (VI) by treatment with hydrochloric acid in THF/H2O at 100 ° C.
  • Scheme 4 shows a method for producing compounds (XII) and (XIII).
  • the intermediate (XI) can be prepared by reacting the intermediate (VIII) with an alkylating agent, such as alkyl lithium, alkyl Grignard agent or alkyl cupper lithium reagent, in an appropriate solvent.
  • the compound (XII) can be prepare by acidic hydrolysis of the acetal group of the intermediate (XI) in an appropriate solvent.
  • the compound (XIII) can be prepared by reacting the intermediate (IV) with an alkylating agent stated above in an appropriate solvent.
  • the compound (XIII) can be also prepared by reacting the compound (XII) with NH2-OY (III) in the presence of base such as pyridine, sodium carbonate, sodium hydrogen carbonate and the like.
  • the compound (XII) can be also prepared from the compound (XIII) under similar reaction condition as described in Scheme 3.
  • Scheme 5a shows a method for producing compound (XV) and (XVI) wherein R 11 is Ci-e alkyl,
  • the intermediate (XIV) can be prepared by reductive amination of the intermediate (VIII) with a primary amine, R"-NH2, in one step or stepwise via intermediate imine (XVII), using the reducing agent such as sodium cyanoborohydride, sodium triacetoxyborohydride and the like.
  • the compound (XV) can be prepared by acidic hydrolysis of the acetal group of compound
  • the compound (XIV) in an appropriate solvent by the method known to the skilled art.
  • the compound (XVI) can be prepared by reductive amination of the intermediate (IV) with a primary amine, R U -NH2 by the same method as described above either directly or step wise fashion via intermediate (XVIII).
  • the compound (XVI) wherein R 11 is Ci-e alkyl, C3-7 cycloalkyl or 3- to 7-membered heterocycloalkylamino which contains -0-, -S-, -NR 6 -, -SO- or -SO2- as a ring atom such as oxetane can be prepared from the intermediate (IV) in 2 steps: (i) by reductive amination with ammonium acetate/sodium cyanoborohydride in ethanol, (ii) followed by second reductive amination of the resulting intermediate (XIX) with a ketone corresponding to R" such as oxetan-3-one and sodium triacetoxyborohydride in dichloromethane.
  • Scheme 6 shows a method for producing compound (XXI), wherein R 11 is C ⁇ .e alkyl, C3-7 cycloalkyl, or 3- to 7-membered heterocycloalkyl which contains -0-, -S-, -NR 6 -, or -SO2- as a ring atom.
  • the intermediate (XX) can be prepared by Scheme 2 or 3.
  • the compound (XXI) can be prepared by esterification of XX with a carboxylic acid of formula R 12 C02H by an active ester method or with condensation agent that are known to those skilled in the art.
  • NMR analysis was performed using Brucker, A VANCE 400MHz or 300MHz, and NMR data were expressed as chemical shifts in ppm (parts per million) ( ⁇ ), and the deuterium lock signal from the sample solvent was referenced.
  • Mass spectrum data was obtained using a LC-MS, Shimazu LCMS-2020 or -2010 equipped with a photodiode array, SPD-M20A, ESI-SQD 2020 or 2010 (Shimadzu)
  • Root temperature means temperatures ranging from about 20°C to 25°C. All non-aqueous reactions were carried out under a nitrogen atmosphere. Concentration or distillation of solvents under reduced pressure means that a rotary evaporator was used.
  • the resulting solution was stirred for 3 h at 0°C.
  • the reaction was then quenched by addition of sat. aqueous ammonium chloride (30 mL).
  • the resulting mixture as extracted with diethyl ether (30 mL x3) and the organic layers were combined and washed with saturated aqueous sodium chloride (50 mL).
  • the organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under vacuum.
  • LC-MS (ESI, m/z): 507 [M-H]-
  • Flash-Prep-HPLC condition (IntelFlash-1): Column, C18 silica gel; mobile phase,
  • Flash-Prep-HPLC condition (IntelFlash-1): Column, silica gel; mobile phase, CH3CN
  • the mixture was stirred for 4 h at 25°C, and then extracted with of dichloromethane (100 mLx5).
  • the organic layer was combined and the pH value of the solution was adjusted to 7-8 with aqueous sodium bicarbonate.
  • the resulting organic layer was separated, dried and concentrated under vacuum.
  • Flash-Prep-HPLC condition (IntelFlash-1): Column, C 18 silica gel; mobile phase,
  • the titled compound was prepared from 4-O-methyl-ascochlorine by the similar manner as described in Example 4 except that O-methyl hydroxyamine hydrochloride was used instead of hydroxyamine hydrochloride.
  • the titled compound was prepared from 4-O-fluoromethyl-ascochlorine by the similar manner as described in Example 8 except that O-methyl hydroxyamine hydrochloride was used instead of hydroxyamine hydrochloride.
  • Prep-HPLC condition (2#-AnalyseHPLC-SHIMADZU(HPLC-10)): Column, XBridge Prep CI 8 OBD Column, 5 urn, 19* 150mm; mobile phase, Water with 10mmolNH 4 HCO3 and CH 3 CN (hold 80.0% CH 3 CN in 12 min); Detector, U V 254nm.
  • Prep-HPLC condition Column: X Bridge CI 8, 19*250 mm, 5 urn; Mobile Phase
  • A Water/10mm NH4HCO3, Mobile Phase B: CH3CN; Flow rate: 30 mL/min; Gradient: 88%B to 88%B in 8 min; 254nm; Detector.
  • the titled compound was prepared by the method similar to that described in Example 12 except that 0-(2-(dimethylamino)ethyl)hydroxyamine hydrochloride was used instead of hydroxyamine hydrochloride.
  • the titled compound was prepared by the method similar to that described in Example 12 except that 0-(2-(morpholin-4-yl)ethyl)hydroxyamine hydrochloride was used instead of hydroxyamine hydrochloride.
  • the titled compound was prepared by the method similar to that described in Example 12 except that 0-(2-(N-methylpipearzin-4-yl)ethyl)hydroxyamine hydrochloride was used instead of hydroxyamine hydrochloride.
  • the titled compound was prepared by the method similar to that described in Example 12 except that ethyl 2-(aminooxy)acetate was used instead of hydroxyamine hydrochloride.
  • Prep-HPLC condition (2#-AnalyseHPLC-SHIMADZU(HPLC-10)): Column, XBridge BEH CI 8 OBD Prep Column, 5 ⁇ , 19 mm 250 mm; mobile phase, Water with lOmmol NH4HCO3 and CH3CN (hold 90.0% CH3CN in 9 min); Detector, uv 254&220nm.
  • Prep-HPLC (2#-AnalyseHPLC-SHIMADZU(HPLC-10)): Column, XBridge Shield RP18 OBD Column, 5um,19*150mm; mobile phase, WATER WITH 0.05%TFA and CH 3 CN (hold 72.0% CH 3 CN in 11 min); Detector, UV 254nm.
  • the titled compound was prepared by the similar method as described in Example 6 except that 0-2-(morpholin-4-yl)ethyl hydroxyamine hydrochloride was used instead of hydroxyamine hydrochloride.
  • Chiral-Prep-HPLC condition Column: XBridge Shield RP18 OBD Column, 5um, 19* 150mm; Mobile Phase A: Water with lOmmol NH4HCO3, Mobile Phase B: ACN; Flow rate: 20 mL/min;
  • the titled compound was prepared by the similar method described in Example 21 except that cyclopropyl amine was used instead of ethylamine.
  • Prep-HPLC condition (2#-AnalyseHPLC-SHIMADZU(HPLC-10)): Column, XBridge Shield RP18 OBD Column, 5um, 19* 150mm; mobile phase, Water with 10mmolNH 4 HCO 3 and CH 3 CN (hold 82.0% ACN in 10 min); Detector, UV 254nm.
  • Prep-HPLC condition (2#-AnalyseHPLC-SHIMADZU(HPLC-10)): Column, Gemini-NX 5u C18 11 OA, AXIA Packed, 150 x 21.2 mm; mobile phase, Column: Mobile Phase A: Water with lOmmol CF3COOH, Mobile Phase B: CH3CN; Flow rate: 30 mL/min; Gradient: 60% B to 95% B in 10 min; 254 nm; Detector, uv 254nm.
  • Step 3 A solution of 4-chloro-2-[(2E,4E)-5-[(l R,2R,6R)-3-(cyclopropylamino)-l ,2,6- trimethylcyclohexyl]-3-methylpenta-2,4-dien-l -yl]-6-(diethoxymethyl)-3-(fluorometh ⁇
  • methylphenoi (460 mg, 0.83 mmol) in THF/water/acetic acid (10/20/10 mL) was placed in a 100-mL round-bottom flask. The mixture was stirred for 3 h at room temperature and concentrated under vacuum. The residue was dissolved in water (10 mL). The mixture was extracted with ethyl acetate (20 mL) and the organic layer was combined, then concentrated under vacuum.
  • Prep-HPLC condition (2#-AnaIyseHPLC-SHIMADZU(HPLC- 10)): Column, Gemini-NX C 18 AXAI Packed, 21.2 x 150mm 5um 1 l nm; mobile phase, Water with l Ommol NH4HCO3 and CH3CN (hold 95.0% CH 3 CN in 8 min); Detector, uv 254nm.
  • Flash-Prep-HPLC condition (IntelFlash-1): Column, CI 8 silica gel; mobile phase,
  • Flash-Prep-HPLC condition (IntelFlash-1): Column, C18 silica gel; mobile phase,
  • Prep-HPLC condition (Prep HPLCJvlC 1 ): Column: XBridge Shield RP18 OBD Column, 5um, 19* 150mm; Mobile Phase A: Waters(0.1 %FA), Mobile Phase B: ACN; Flow rate: 20 mL/min; Gradient: 56% B to 80% B in 8 min; Detector: 254 nm,.
  • p-AMPK and p-ACC levels were measured by Western blot analysis using the antibodies for p- AMPK and p-ACC. Following reagents were purchased from the vendors, and the instruments listed below were used.
  • Reagents AMP a (D5A2) rabbit mAb, phospho-acetyl-CoA carboxylase (Ser79) (D7D11) rabbit mAb, anti-rabbit IgG, HRP-linked antibody, and beta-actin (13E5) rabbit mAb were purchased from Cell Signaling Technology. ECL Western Blotting Detection Reagents were purchased from Thermo.
  • HepG2 cells were cultured in MEM medium supplemented with 10% FBS at 37°C and under 5% CO2 condition. HepG2 cells (1 X 10 6 cells/well) were seeded into 6-well cell culture plates for 24hr. The cells were starved in serum-free MEM medium (3ml/well) for 24hr.
  • the compounds were dissolved in DMSO to make the stock solution of 5, 25, 50mM respectively.
  • the compounds were diluted to final concentration in serum-free MEM medium to be 5, 25, and 50 ⁇ .
  • Metformin was dissolved in PBS to make the stock concentration as 1M. Metformin was diluted to final concentration in serum-free MEM medium to be 5, and 50 mM
  • Electrophoresis was run at 120 V constant voltage until the blue marker reaches the end of the gel.
  • the proteins were transferred to a PVDF membrane using Bio-Rad's Trans blot for 40 min at 300 mA.
  • test compounds and positive control compound were prepared in DMSO at the concentration of 30 mM.
  • Diclofenac was used as positive control in the assay.
  • the LC system comprised a Shimadzu liquid chromatograph separation system equipped with degasser DGU-20A3, solvent delivery unit LC-20AD, system controller CBM-20A, column oven CTO- 1 OAS VP and CTC Analytics HTC PAL System. Mass spectrometric analysis was performed using an API 4000 instrument from AB Inc (Canada) with an ESI interface. The data acquisition and control system were created using Analyst 1.6 software from ABI Inc. The results of the measurements are indicated in Table 2.
  • PK profiles of Compound 19 in the blood were analyzed by LC/MS/MS after single dose administration of the test compounds in SD rats.
  • LC/MS/MS instrument [LC: Shimazu, MS: AB API4000 (ion source: ESI)], LC column: XSELECTTM CSHTM C I 8 2.5 ⁇ (2.1 ⁇ 50 mm), mobile phase: 5% acetonitrile (0.1 % formic acid) and 95% acetonitrile (0.1 % formic acid).
  • C57BL/6J mice were fed with high fat diet (Research Diets, D 12492) for 64 days, and streptozotocin (10 mg/kg) was given orally on day 22 and day 29. Compound treatment was done from day 36 to day 63.
  • mice were randomly assigned to respective groups using a computer-generated randomization procedure.
  • Vehicle group HFD/STZ + vehicle
  • Treatment dose Compound 5 (po. qd x 28: 0.2mg/kg, 1 mg/kg and 5mg/kg)
  • Pioglitazone a positive control (po. qd x 28: 30mg/kg)
  • Normal diet group normal diet + vehicle
  • Compound 5 and pioglitazone were administered orally once a day for 4 weeks with the dose indicated above.
  • Blood glucose level was measured with the blood from tail vein every 2nd week by using Accu-Chek Aviva System.
  • Figures 3 and 4 show the effect of the compounds of the present invention on the levels of triglyceride and total cholesterol in blood of mice.
  • Figure 5 shows the effect of the compounds of the present invention on the blood glucose level of mice in fasting state.
  • mice were randomly assigned to 8 groups, based on body weight and fasting blood glucose level thereof on day 0.
  • the study design is shown in Table 4. Table 4. Study design
  • Cmpd 19 was administered as a solution in the vehicle (PEG-400/ DMSO (90: 10 v/v)), and metformin was administered as a solution in saline.
  • the drugs were orally administered to the mice once a day for 28 consecutive days. Body weight and food consumption were measured during the course of this study. The following data were obtained to confirm the efficacy of the drugs.
  • Oral glucose tolerant tests were conducted on Day 0 (baseline) and Day 28 (endpoint) after treatment. Following oral administration 0.5 g/kg glucose, the blood glucose levels were measured by using Accu-Chek Performa System. Blood samples were collected at 0, 30, 60 and 120 minutes after glucose administration. The results are shown in Figures 6 and 7.
  • AUC Area under curve
  • Serum triglyceride level was measured during the study. Blood samples were collected from the orbital veins at week 4, and then immediately processed by centrifugation at 4°C, 6,000 g for 15 minutes. After the samples were transferred into new test tubes. Triglyceride level was measured by using TOSHIBA TBA-40FR automated biochemical analyzer. The results are shown in Figure 9. The results indicate tendency of strong reduction in triglyceride level observed in the groups administered with the combination of Cmpd 19 and metformin.
  • Hemoglobin Aic (HbAic) level was measured on Day 28. Blood samples were collected from the orbital veins at week 4, and then immediately processed by centrifugation at 4°C, 6,000 g for 15 minutes, and then they were transferred into new test tubes. HbA lc level was measured by using- TOSHIBA TBA-40FR automated biochemical analyzer.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Obesity (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Child & Adolescent Psychology (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

An object of the present invention is to provide a measure for treating or preventing a disease or condition that involves AMPK dysregulation, particularly diabetes, obesity, and hyperlipidemia. The present invention provides a combination therapy using a compound presented by formula I, a pharmaceutically acceptable salt or a solvate thereof with metformin.

Description

DESCRIPTION
Title of Invention: USE OF ASCOCHLORI DERIVATIVE FOR COMBINATION THERAPY Technical Field
[0001] The present invention is directed to a combination therapy using an ascochlorin derivative with metformin for use in the treatment or prevention of diseases or conditions that involve AMPK dysregulation, such as diabetes, obesity, and hyperlipidemia. The invention is also directed to a pharmaceutical composition comprising an ascochlorin derivative for use in combination with administration of metformin, for the treatment or prevention of diseases or conditions that involve AMPK dysregulation such as diabetes, obesity, and hyperlipidemia.
Background Art
[0002] The adenosine monophosphate-activated protein kinase (AMPK) is an important regulatory protein for cellular energy balance and is considered as a master switch of glucose and lipid metabolisms in various organs, especially in skeletal muscle, liver and fat tissue.
[0003] AMPK is also known to have critical roles in regulating cell growth and autophagy through inhibition of m-TOR, as well as mitochondrial biogenesis. [Physiological Rev. 2009, vol. 89, page 1025, Nature Reviews/Molecular Cell Biology, 2012, vol.13 page 251, Nature Cell Biology, 2011, vol.13 (9),1016, Molecular and Cellular Endocrinology, 2013 vol. 366, page 152, J. Physiol., 2006, vol 574.1 page 33.].
[0004] When intracellular ATP levels become lower, AMP or ADP can directly bind to the γ regulatory subunit of AMPK, leading to a conformational change that promotes AMPK phosphorylation at Thrl72 by LKB 1 (known as a tumor suppressor) or C AMKK2 (Ca++ dependent kinase). Further, the conformational change also protects AMPK from dephosphorylation by protein phosphatases 2A and C so that AMPK remains activated.
[0005] The dysregulation of AMPK is considered as a pathogenic factor for the diseases such as cancer, type 2 diabetes, metabolic syndrome-associated diseases (dyslipidemia, adiposity, atherosclerotic cardiovascular disease, hypertension, and nonalcoholic fatty liver disease (NAFLD)), inflammation and Alzheimer disease, and thus AMPK is considered as a target for the prevention and therapy of these diseases [metabolic syndrome: Science in Medicine, 2013, Vol. 123 (7) page 2764, Molecular and Cellular Endocrinology, 2013, vol. 366 page 135; cancer and inflammation: Oncotarget, 2015, vol. 6 (10), page 7365; cardiovascular disease: Circ Res., 2009 vol. 104 (3), page 282; inflammation,
Diabetologia, 2010 vol. 53, page 2406, Am. J. Respir. Cell Mol. Biol., 2010, vol 42, page 676;
Alzheimer disease: J Alzheimers Dis. 2009, vol. 16(4) page 731].
[0006] Metformin is known as an antihyperglycemic agent to indirectly activate AMPK through inhibition of respiratory chain complex 1 [Clinical Science, 2012, vol. 122, page 253]. Metformin, however, is reported to activate AMPK in HepG2 cell only at surprisingly high concentration of mM level for in vitro cellular experiment.
[0007] 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR) is known as an AMP mimic. AICAR directly activates AMPK through binding to a regulatory γ-subunit to promote phosphorylation of AMPK and to protect AMPK from dephosphorylation. A-769662 is reported to be a direct AMPK- activator with a different binding mode, [Cell Metabolism, 2006, vol. 3, page 403]. There are other AMPK-activators reported in literatures, but none of them is clinically used up to now, partly because of the insufficient in vivo efficacy and/or safety profile. Therefore, novel AMPK activators with good efficacy and safety profile would provide therapeutic benefits to the patients with diseases that involve AMPK dysregulations.
[0008] 4-O-Methy-ascochlorin (MAC) is a derivative of a fungal metabolite of ascochlorin, which is reported to have ability for activating AMPK in HepG2 cells [Biochemical and Biophysical Research Communications, 2011 , vol. 406, page 353]. 4-O-Substituted ascochlorin derivatives including MAC are described as an antidiabetic agent in JP H06-305959 A (published in 1994), and prodrugs of MAC that are Schiff base derivatives formed with natural amino acids are described in WO2004/074236. However, the development of any ascochlorin derivatives has not become successful.
Citation List
Patent Documents
[0009] Patent Document 1 : JP H06-305959 A; Patent Document 2 : WO 2004/074236 Non-Patent Documents
[0010] Non-Patent Document 1 : Physiological Rev. 2009, vol. 89, page 1025,
Non-Patent Document 2: Nature Reviews/Molecular Cell Biology, 2012, vol.13 page 251, . Non-Patent Document 3: Nature Cell Biology, 2011 , vol.13 (9), 1016,
Non-Patent Document 4: Molecular and Cellular Endocrinology, 2013 vol. 366, page 152, Non-Patent Document 5: J. Physiol., 2006, vol 574.1 page 33
Non-Patent Document 6 : Science in Medicine, 2013 , 123 (7), 2764;
Non-Patent Document 7: Mol. Cell. Endocrinol., 2013, 366; 135;
Non-Patent Document 8: Oncotarget, 2015, 6 (10), 7365;
Non-Patent Document 9: Circ Res., 2009, 104 (3), page 282;
Non-Patent Document 10: Diabetologia, 2010, 53, 2406;
Non-Patent Document 11 : Am. J. Respir. Cell Mol. Biol., 2010, 42, 676;
Non-Patent Document 12: J Alzheimers Dis. 2009, 16(4), 731;
Non-Patent Document 13: Clinical Science, 2012, 122, 253;
Non-Patent Document 14: Cell Metabolism, 2006, 3, 403.
Non-Patent Document 15: Biochemical and Biophysical Research Communications, 2011, vol.
406, page 353
Summary of Invention
Technical Problem
[0011] The present inventors produced ascochlorin derivatives, which are confirmed to have AMPK activation profile, and significantly improved metabolic stability as compared with MAC (PCT application: PCT/JP2017/000902). Particularly, the inventors found that the ascochlorin derivatives also show good in vivo efficacy in several disease models especially for oncology and metabolic diseases. On the other hand, there are strong needs for measures to treat or prevent a certain number of diseases including diabetes, obesity, and hyperlipidemia.
Solution to Problem
[0012] The present inventors have conducted diligent studies and consequently completed the present invention by finding that a combination of an ascochlorin derivative with metformin has an excellent effect for treating or preventing diabetes, obesity, and hyperlipidemia.
[0013] The present invention is directed to a combination therapy using an ascochlorin derivative with metformin for treating or preventing a disease that involves AMPK dysregulation. The invention is also directed to a pharmaceutical composition comprising an ascochlorin derivative for use in combination with administration of metformin for the treatment or prevention of diseases or conditions that involve AMPK dysregulation.
[0014] According to one aspect of the present invention, there is provided the following inventions:
[0015] (1-1) A pharmaceutical composition comprising a compound represented by formula (I):
Figure imgf000006_0001
wherein
R1 is formyl, or -CH=N-OY, in which Y is a hydrogen atom or Ci-6 alkyl;
R2 is C 1-6 alkyl, in which the alkyl may be substituted with one to five fluorine atoms;
R3 is a hydrogen atom or Ci-6 alkyl;
R4 is hydroxy,€].& alkylamino, C3-7 cycloalkylamino, or 3- to 7-membered
heterocycloalkylamino which contains -0-, -S-, -NR6-, -SO- or -SO2- as a ring atom, or
R3 and R4 together with the carbon atom to which they are attached form >C=N-0-Z;
Z is a hydrogen atom, Ci-e alkyl, -CO(CH2)n-R5 or -(CH2)„-R5;
n is an integer selected from 1 to 4;
R5 is -CO2R6, -CONR7R8, -OCONR7R8, -S02NR7R8, -S02R9, hydroxy, -NHSO2R9, or -
NR7R8;
R6, R7 and R8 are each independently selected from a group consisting of a hydrogen atom, Ci- 6 alkyl, and C3-7 cycloalkyl; or R7 and R8 together with the nitrogen atom to which they are attached form a nitrogen containing heterocyclic ring which may further contain -0-, -S-, -NR6-, -SO- or -S02- as a ring atom, in which the heterocyclic ring may be substituted with one or more substituents selected from hydroxy and Ci-e alkyl,
R9 is Ci-6 alkyl, or C3-7 cycloalkyl;
a pharmaceutically acceptable salt or a solvate thereof,
wherein the composition is for use in combination with administration of metformin.
[0016] (1-2) The pharmaceutical composition according to ( 1 - 1 ), wherein R4 is selected from the group consisting of hydroxy, methylamino, ethylamino, cyclopropylamino, and oxetan-3-ylamino.
[0017] (1-3) The pharmaceutical composition according to ( 1 - 1 ), wherein R3 and R4 together with the carbon atom to which they are attached form >C=N-0-Z in which Z is selected from the group consisting of a hydrogen atom, Ci-e alkyl, -CH2CO2H, -CH2CO2R5, morpholinoethyl, piperazinylethyl, N-methyl-piperazinylethyl, and N,N-dimethylamino-ethyl.
[0018] (1-4) The pharmaceutical composition according to (1-2) or (1-3), wherein R1 is formyl.
[0019] (1-5) The pharmaceutical composition according to (1-2) or (1-3), wherein R1 is -CH=N-OH.
[0020] (1-6) The pharmaceutical composition according to (1-2) or (1-3), wherein R1 is -CH=N- OMe.
[0021 ] (1-7) The pharmaceutical composition according to (1-1), wherein the compound represented by formula (I) is selected from the group consisting of
3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R)-3-hydroxy-l,2,3,6-tetramethylcyclohexyl)-3- methylpenta-2,4-dien-l -yl)-4-methoxy-2-methylbenzaldehyde,
3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)-l,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l -yl)-4-methoxy-2-methylbenzaldehyde,
(E)-3-chloro-6-hydroxy-5-((2E,4E)-5-(( 1 R,2R,6R,E)-3-(hydroxyimino)- 1 ,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l-yl)-4-methoxy-2-methylbenzaldehyde oxime,
3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)-l,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l-yl)-2-methylbenzaldehyde oxime,
3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)-l,2,6- trimethylcyclohexy l)-3 -methylpenta-2,4-dien- 1 -y l)-2-methylbenzaldehyde,
[(l E,2R,3R,4R)-3-[(lE,3E)-5-(3-chloro-5-formyl-6-hydroxy-2-methoxy-4-methylphenyl)-3- methylpenta-l,3-dien-l-yl]-2,3,4-trimethylcyclohexylidene]amino 2-(dimethylamino)acetate, (E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,3S,6R)-3-hydroxy-l,2,3,6- tetramethylcyclohexyl)-3 -methylpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde oxime,
(E)-3-chloro-6-hydroxy-4-methoxy-5-((2E,4E)-5-(( 1 R,2R,6R,E)-3-(methoxyimino)- 1 ,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l -yl)-2-methylbenzaldehyde O-methyl oxime,
(E)-3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(methoxyimino)- 1 ,2,6-trimethylcyclohexyl)-3-methylpenta-2,4-dien-l-yl)-2-methylbenzaldehyde O-methyl oxime,
(E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)-l,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -y])-4-methoxy-2-methylbenzaldehyde O-methyl oxime,
(E)-3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)- 1 ,2,6-trimethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-2-methylbenzaldehyde O-methyl oxime,
3-chloro-4-(fluoromethoxy)-6-hydroxy-2-methyl-5-((2E,4E)-3-methyl-5-((l R,2R,6R,E)-l,2,6- trimethyl-3-(2-mo holinoethoxyimino)cyclohexyl)penta-2,4-dien-l-yl)benzaldehyde,
3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(cyclopropylamino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l-yI)-4-(fluoromethoxy)-6-hydroxy-2-methylbenzaldehyde,
(E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((l R,2R,3S,6R)-3-hydroxy- 1 ,2,3,6- tetramethylcyclohexyl)-3 -methylpenta-2,4-dien- 1 - l)-4-methoxy-2-methy Ibenzaldehyde O-methyl oxime,
(E)-3-chloro-5-((2E,4E)-5-(( 1 R,2R,6R,E)-3-(2-(dimethylamino)ethoxyimino)- 1 ,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l-yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde O- methyl oxime,
(E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-((lR,2R,6R,E)-l,2,6- trimethyl-3-(2-moφholinoethoxyimino)cyclohexyl)penta-2,4-dien- 1 -yl)benzaldehyde O-methyl oxime,
(E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-((l R,2R,6R,E)-l,2,6- trimethyl-3-(2-(4-methylpiperazin-l-yl)ethoxyimino)-cyclohexyl)penta-2,4-dien-l-yl)benzaldehyde O- methyl oxime,
ethyl 2-([[( 1 E,2R,3R,4R)-3-[( 1 E,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[( 1 E)- (methoxy imino)methy 1] -4-methylphenyl] -3 -methy lpenta- 1 ,3 -dien- 1 -y 1] -2,3 ,4- trimethylcyclohexylidene]amino]oxy)acetate, 2- ([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (methoxyimino)methyl]-4-rnethylphenyl]-3-methylpenta-l ,3-dien-l-yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid,
3- chloro-5 (2E,4E)-5-((lR,2R,6R)-3-(ethylamino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l-yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde,
(E)-3-chloro-5-((2E,4E)-5-(( 1 R,2R,6R)-3-(ethylamino)- 1 ,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde O-methyl oxime,
3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(cyc]opropylamino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l-yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde,
(E)-3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(cyclopropylamino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde O-methyl oxime,
2-[(2E,4E)-5-[( 1 R,2R,6R)-3-amino- 1 ,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien- 1 -yl]- 4-chloro-3-methoxy-6-[(l E)-(methoxyimino)methyl]-5-methylphenol,
(E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-(( 1R,2R,6R)- 1,2,6- trimethyl-3-(oxetan-3-ylamino)cyclohexy])penta-2,4-dien-l -yl)benzaldehyde O-methyl oxime,
2-([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-fluoromethoxy-5-formyl-4- methylphenyl]-3-methylpenta-l,3-dien-l-yl]-2,3,4-trimethylcyclohexylidene]-amino]oxy)acetic acid,
2-([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (hydroxyimino)methyl]-4-methylphenyl]-3-methylpenta-l ,3-dien-l -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid,
2-([[( 1 E,2R,3R,4R)-3-[( 1 E,3 E)-5-[3-chloro-2-methoxy-6-hydroxy-5-[(l E)- (hydroxyimino)methyl]-4-methylphenyl]-3-methylpenta- 1 ,3-dien- 1 -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid,
and pharmaceutically acceptable salts thereof.
[0022] ( 1 -8) The pharmaceutical composition according to any one of ( 1 - 1 ) to ( 1 -7), wherein the composition is for use is treating or preventing a disease selected from diabetes, obesity, and hyperlipidemia.
[0023] (1-9) The pharmaceutical composition according to (1-8), wherein the disease is type 2 diabetes.
[0024] (1-10) The pharmaceutical composition according to any one of ( 1 - 1 ) to ( 1 -9), wherein the compound represented by formula (I) is administered in amount of 0.1 to 100 mg/kg/day.
[0025] (1-11) The pharmaceutical composition according to any one of (1-1) to (1-10), wherein metformin is administered in amount of 50 to 1000 mg kg/day.
[0026] (2-1) A pharmaceutical composition comprising metformin for use in combination with administration of a compound represented by formula (I):
Figure imgf000010_0001
wherein
R1 is formyl, or -CH=N-0-Y, in which Y is a hydrogen atom or Ci-6 alkyl;
R2 is Ci-6 alkyl, in which the alkyl may be substituted with one to five fluorine atoms;
R3 is a hydrogen atom or Ci-6 alkyl;
R4 is hydroxy, Ci-6 alkylamino, C3-7 cycloalkylamino, or 3- to 7-membered
heterocycloalkylamino which contains -0-, -S-, -NR6-, -SO- or -SO2- as a ring atom, or
R3 and R4 together with the carbon atom to which they are attached form >C=N-0-Z;
Z is a hydrogen atom, Ci-6 alkyl, -CO(CH2)n-R5 or -(CH2)n-R5;
n is an integer selected from 1 to 4;
R5 is -CO2R6, -CONR7R8, -OCONR7R8, -S02NR7R8, -SO2R9, hydroxy, -NHSO2R9, or -
NR7R8;
R6, R7 and R8 are each independently selected from a group consisting of a hydrogen atom, Ci- 6 alkyl, and C3_7 cycloalkyl; or R7 and R8 together with the nitrogen atom to which they are attached form a nitrogen containing heterocyclic ring which may further contain -0-, -S-, -NR6-, -SO- or -S02- as a ring atom, in which the heterocyclic ring may be substituted with one or more substituents selected from hydroxy and Ci-6 alkyl,
R9 is Ci-6 alkyl, or C3-7 cycloalkyl; a pharmaceutically acceptable salt or a solvate thereof.
[0027] (2-2) The pharmaceutical composition according to (2-1), wherein R4 is selected from the group consisting of hydroxy, methylamino, ethylamino, cyclopropylamino, and oxetan-3-ylamino.
[0028] (2-3) The pharmaceutical composition according to (2-1), wherein R3 and R4 together with the carbon atom to which they are attached form >C=N-0-Z in which Z is selected from the group consisting of a hydrogen atom, Ci-6 alkyl, -CH2CO2H, -CH2CO2R5, morpholinoethyl, piperazinylethyl, N-methyl-piperazinylethyl, and N,N-dimethylamino-ethyl.
[0029] (2-4) The pharmaceutical composition according to (2-2) or (2-3), wherein R1 is formyl.
[0030] (2-5) The pharmaceutical composition according to (2-2) or (2-3), wherein R1 is -CH=N-OH.
[0031] (2-6) The pharmaceutical composition according to (2-2) or (2-3), wherein R1 is -CH=N- OMe.
[0032] (2-7) The pharmaceutical composition according to (2-1), wherein the compound represented by formula (I) is selected from the group consisting of
3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R)-3-hydroxy-l,2,3,6-tetramethylcyclohexyl)-3- methylpenta-2,4-dien- 1 -y l)-4-methoxy-2-methylbenzaldehyde,
3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)- 1,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l-yl)-4-methoxy-2-methylbenzaldehyde,
(E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)-l ,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l-yl)-4-methoxy-2-methylbenzaldehyde oxime,
3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)-l,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-2-methylbenzaldehyde oxime,
3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-(( 1 R,2R,6R,E)-3-(hydroxyimino)- 1 ,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l -yl)-2-methylbenzaldehyde,
[(lE,2R,3R,4R)-3-[(lE,3E)-5-(3-chloro-5-formyl-6-hydroxy-2-methoxy-4-methylphenyl)-3- methylpenta-1 ,3-dien-l -yl]-2,3,4-trimethylcyclohexylidene]amino 2-(dimethylamino)acetate,
(E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,3S,6R)-3-hydroxy-l ,2,3,6- tetramethylcyclohexyl)-3 -methylpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde oxime,
(E)-3-chloro-6-hydroxy-4-methoxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(methoxyimino)-l,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-2-methylbenzaldehyde O-methyl oxime,
(E)-3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((l R,2R,6R,E)-3-(methoxyimino)- 1 ,2,6-trimethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-2-methylbenzaldehyde O-methyl oxime,
(E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)- 1 ,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde O-methyl oxime,
(E)-3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)- 1 ,2,6-trimethy]cyclohexyl)-3-methylpenta-2,4-dien-l -yl)-2-methylbenzaldehyde O-methyl oxime,
3-chloro-4-(fluoromethoxy)-6-hydroxy-2-methyl-5-((2E,4E)-3-methyl-5-((lR,2R,6R,E)-l,2,6- trimethyl-3 -(2-mo holinoethoxyimino)cyclohexyl)penta-2,4-dien- 1 -yl)benzaldehyde,
3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(cyclopropylamino)-l ,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien- 1 -yl)-4-(fluoromethoxy)-6-hydroxy-2-methylbenzaldehyde,
(E)-3-chloro-6-hydroxy-5-((2E,4E)-5-(( 1 R,2R,3S,6R)-3-hydroxy- 1 ,2,3,6- tetramethylcyclohexyl)-3-methylpenta-2,4-dien-l-yl)-4-methoxy-2-methylbenzaldehyde O-methyl oxime,
(E)-3-chloro-5-((2E,4E)-5-((lR,2R,6R,E)-3-(2-(dimethylamino)ethoxyimino)-l,2,6- trimethy Icyclohexy l)-3 -methylpenta-2,4-dien- 1 -yl)-6-hydroxy-4-methoxy-2-methy Ibenzaldehyde O- methyl oxime,
(E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-((lR,2R,6R,E)-l ,2,6- trimethyl-3-(2-mo holinoethoxyimino)cyclohexyl)penta-2,4-dien-l-yl)benzaldehyde O-methyl oxime,
(E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-((l R,2R,6R,E)-l,2,6- trimethyl-3-(2-(4-methylpiperazin-l-yl)ethoxyimino)-cyclohexyl)penta-2,4-dien-l-yl)benzaldehyde O- methyl oxime,
ethyl 2-([[(l E,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(l E)- (methoxyimino)methyl]-4-methylphenyl]-3-methylpenta-l ,3-dien-l -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetate,
2-([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (methoxyimino)methyl]-4-methylphenyl]-3-methylpenta- 1 ,3-dien- 1 -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid, 3-chloro-5-((2E,4E)-5-((l R,2R,6R)^ ethylamino)-l ,2,6-trirnethylcyclohexyl)-3- methylpenta-2,4-dien-l -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde,
(E)-3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(ethylamino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde O-methyl oxime,
3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(cyclopropylamino)-l ,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l-yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde,
(E)-3-chloro-5-((2E,4E)-5-((l R,2R,6R)-3-(cyclopropylamino)- l ,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l-yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde O-methyl oxime,
2-[(2E,4E)-5-[(lR,2R,6R)-3-amino-l,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien-l-yl]- 4-chloro-3-methoxy-6-[(l E)-(methoxyimino)methyl]-5-methylphenol,
(E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-((l R,2R,6R)-l ,2,6- trimethyl-3-(oxetan-3-ylamino)cyclohexyl)penta-2,4-dien-l-yl)benzaldehyde O-methyl oxime,
2-([[( 1 E,2R,3R,4R)-3 -[( 1 E,3 E)-5-[3 -chloro-6-hydroxy-2-fluoromethoxy-5-formyl-4- methylphenyl]-3-methylpenta-l ,3-dien-l -yl]-2,3,4-trimethy]cyclohexylidene]-amino]oxy)acetic acid,
2-([[(l E,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(l E)- (hydroxyimino)methyl]-4-methylphenyl]-3-methylpenta-l ,3-dien-l-yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid,
2-([[(l E,2R,3R,4R)-3-[(l E,3E)-5-[3-chloro-2-methoxy-6-hydroxy-5-[(lE)- (hydroxyimino)methyl]-4-methylphenyl]-3-methylpenta-l ,3-dien-l -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid,
and pharmaceutically acceptable salts thereof.
[0033] (2-8) The pharmaceutical composition according to any one of (2-1) to (2-7), wherein the composition is for use is treating or preventing a disease selected from diabetes, obesity, and hyperlipidemia.
[0034] (2-9) The pharmaceutical composition according to (2-8), wherein the disease is type 2 diabetes.
[0035] (2-10) The pharmaceutical composition according to any one of (2- 1) to (2-9), wherein the compound represented by formula (I) is administered in amount of 0.1 to 100 mg/kg/day. [0036] (2-11) The pharmaceutical composition according to any one of (2-1) to (2-10), wherein metformin is administered in amount of 50 to 1000 mg/kg/day.
[0037] (3-1) A method for treating or preventing a disease that involves AMPK dysregulation comprising:
administering to a subject in need thereof a therapeutically effective amount of a compound represented by formula (I):
Figure imgf000014_0001
wherein
R1 is formyl, or -CH=N-0-Y, in which Y is a hydrogen atom or G-6 alkyl;
R2 is G-6 alkyl, in which the alkyl may be substituted with one to five fluorine atoms;
R3 is a hydrogen atom or G-6 alkyl;
R4 is hydroxy, G-6 alkylamino, C3-7 cycloalkylamino, or 3- to 7-membered
heterocycloalkylamino which contains -0-, -S-, -NR6-, -SO- or -SO2- as a ring atom, or
R3 and R4 together with the carbon atom to which they are attached form >C=N-0-Z;
Z is a hydrogen atom, G.6 alkyl, -CO(CH2)n-R5 or -(CH2)„-R5;
n is an integer selected from 1 to 4;
R5 is -C02R6, -CONR7R8, -OCONR7R8, -S02NR7R8, -S02R9, hydroxy, -NHSO2R9, or -
NR7R8;
R6, R7 and R8 are each independently selected from a group consisting of a hydrogen atom, G- 6 alkyl, and C3-7 cycloalkyl; or R7 and R8 together with the nitrogen atom to which they are attached form a nitrogen containing heterocyclic ring which may further contain -0-, -S-, -NR6-, -SO- or -SO2- as a ring atom, in which the heterocyclic ring may be substituted with one or more substituents selected from hydroxy and G-6 alkyl,
R9 is G-6 alkyl, or C3-7 cycloalkyl; a pharmaceutically acceptable salt or a solvate thereof, in combination with administering a therapeutically effective amount of metformin.
[0038] (3-2) The method according to (3- 1 ), wherein R4 is selected from the group consisting of hydroxy, methylamino, ethylamino, cyclopropylamino, and oxetan-3-ylamino.
[0039] (3-3) The method according to (3-1), wherein R3 and R4 together with the carbon atom to which they are attached form >C=N-0-Z in which Z is selected from the group consisting of a hydrogen atom, Ci-6 alkyl, -CH2CO2H, -CH2CO2R5, morpholinoethyl, piperazinylethyl, N-methyl-piperazinylethyl, and N,N-dimethylamino-ethyl.
[0040] (3-4) The method according to (3-2) or (3-3), wherein R1 is formyl.
[0041] (3-5) The method according to (3-2) or (3-3), wherein R1 is -CH=N-OH.
[0042] (3-6) The method according to (3-2) or (3-3), wherein R1 is -CH=N-OMe.
[0043] (3-7) The method according to (3-1 ), wherein the compound represented by formula (I) is selected from the group consisting of
3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R)-3-hydroxy-l,2,3,6-tetramethylcyclohexyl)-3- methylpenta-2,4-dien- 1 -y l)-4-methoxy-2-methylbenzaldehyde,
3-chloro-6-hydroxy-5-((2E,4E)-5-(( 1 R,2R,6R,E)-3-(hydroxyimino)- 1 ,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l -yl)-4-methoxy-2-methylbenzaldehyde,
(E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)-l,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde oxime,
3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)-l,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l -yl)-2-methylbenzaldehyde oxime,
3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)-l,2,6- trimethylcyclohexy l)-3 -methylpenta-2,4-dien- 1 -yl)-2-methylbenzaldehyde,
[(lE,2R,3R,4R)-3-[(lE,3E)-5-(3-chloro-5-formyl-6-hydroxy-2-methoxy-4-methylphenyl)-3- methylpenta-1 ,3-dien-l -yl]-2,3,4-trimethylcyclohexylidene]amino 2-(dimethylamino)acetate,
(E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,3S,6R)-3-hydroxy-l,2,3,6- tetramethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde oxime,
(E)-3-chloro-6-hydroxy-4-methoxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(methoxyimino)-l,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l -yl)-2-methylbenzaldehyde O-methyl oxime,
(E)-3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(methoxyimino)- l,2,6-trimethylcyclohexyl)-3-methylpenta-2,4-dien-l -yl)-2-methylbenzaldehyde O-methyl oxime,
(E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((l R,2R,6R,E)-3-(hydroxyimino)-l,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yI)-4-methoxy-2-methylbenzaIdehyde O-methyl oxime,
(E)-3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((l R,2R,6R,E)-3-(hydroxyimino)- 1 ,2,6-trimethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-2-methylbenzaldehyde O-methyl oxime,
3-chloro-4-(fluoromethoxy)-6-hydroxy-2-methyl-5-((2E,4E)-3-methyl-5-((lR,2R,6R,E)-l,2,6- trimethyl-3-(2-mo holinoethoxyimino)cyclohexyl)penta-2,4-dien-l-yl)benzaldehyde,
3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(cyclopropylamino)-l ,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien- 1 -yl)-4-(fluoromethoxy)-6-hydroxy-2-methylbenzaldehyde,
(E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,3S,6R)-3-hydroxy-l ,2,3,6- tetramethylcyclohexy])-3-methylpenta-2,4-dien-l -yl)-4-methoxy-2-methylbenzaldehyde O-methyl oxime,
(E)-3-chloro-5-((2E,4E)-5-((lR,2R,6R,E)-3-(2-(dimethylamino)ethoxyimino)-l,2,6- trimethy lcyclohexyl)-3 -methylpenta-2,4-dien- 1 -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde O- methyl oxime,
(E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-((l R,2R,6R,E)-l,2,6- trimethyl-3-(2-morphoIinoethoxyimino)cyclohexyl)penta-2,4-dien- 1 -yl)benzaldehyde O-methyl oxime,
(E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-((l R,2R,6R,E)-l,2,6- trimethyl-3-(2-(4-methylpiperazin-l-yl)ethoxyimino)-cyclohexyl)penta-2,4-dien-l -yl)benzaldehyde O- methyl oxime,
ethyl 2-([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (methoxyimino)methyl]-4-methylphenyl]-3-methylpenta- l,3-dien-l-yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetate,
2-([[( 1 E,2R,3 R,4R)-3-[(l E,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[( 1 E)- (methoxyimino)methyl]-4-methylphenyl]-3-methylpenta-l,3-dien-l -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid, 3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(ethylamino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l-yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde,
(E)-3-chloro-5-((2E,4E)-5-((lR,2R)6R)-3-(ethylamino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien- 1 -yl)-6-hydroxy-4-methoxy-2-methy lbenzaldehyde O-methyl oxime,
3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(cyclopropylamino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien- 1 -yl)-6-hydroxy-4-methoxy-2-methylberizaldehyde,
(E)-3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(cyclopropylamino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien- 1 -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde O-methyl oxime,
2-[(2E,4E)-5-[(lR,2R,6R)-3-amino-l,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien-l -yl]- 4-chloro-3-methoxy-6-[(l E)-(methoxyimino)methyl]-5-methylphenol,
(E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-((lR,2R,6R)-l ,2,6- trimethyl-3-(oxetan-3-ylamino)cyclohexyl)penta-2,4-dien-l -yl)benzaldehyde O-methyl oxime,
2-([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-fluoromethoxy-5-formyl-4- methylphenyl]-3-methylpenta- 1 ,3-dien- 1 -yl]-2,3,4-trimethylcyclohexylidene]-amino]oxy)acetic acid,
2-([[(l E,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (hydroxyimino)methyl]-4-methylphenyl]-3-methylpenta-l,3-dien-l -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid,
2-([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-2-methoxy-6-hydroxy-5-[(lE)- (hydroxyimino)methyl]-4-methylphenyl]-3-methylpenta-l ,3-dien-l -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid,
and pharmaceutically acceptable salts thereof.
[0044] (3-8) The method according to any one of (3-1) to (3-7), wherein the composition is for use is treating or preventing a disease selected from diabetes, obesity, and hyperlipidemia.
[0045] (3-9) The method according to (3-8), wherein the disease is type 2 diabetes.
[0046] (3-10) The method according to any one of (3-1) to (3-9), wherein the compound represented by formula (I) is administered in amount of 0.1 to 100 mg/kg/day.
[0047] (3-11) The method according to any one of (3- 1 ) to (3- 10), wherein metformin is administered in amount of 50 to 1000 mg kg/day. Advantageous Effects of the Invention
[0048] The pharmaceutical composition is useful in treating or preventing a disease selected from diabetes, obesity, hyperlipidemia, cardiovascular diseases, and metabolic syndrome-associated diseases, particularly diabetes, obesity, and hyperlipidemia, more particularly type-2 diabetes.
[0049] The compound of the present invention is useful as a preventive or therapeutic agent for the disease in which AMPK is dysregulated; such as metabolic syndrome, type 2 diabetes, dyslipidemia, adiposity, atherosclerotic cardiovascular disease, hypertension, and nonalcoholic fatty liver disease
(NAFLD), inflammation and Alzheimer disease. According to one aspect of the present invention, the pharmaceutical composition of the present invention is used for preventing or treating a disease in which
AMPK is dysregulated; such as metabolic syndrome, type 2 diabetes, dyslipidemia, adiposity, atherosclerotic cardiovascular disease, hypertension, and nonalcoholic fatty liver disease (NAFLD), inflammation and Alzheimer disease.
Brief Description of the Drawings
[0050] Figure 1 shows the results of western blot analysis to indicate AMPK activation and downstream signal, ACC in HepG2 cell.
Figure 2 shows pharmacokinetic profile of Compound 19 in SD rats.
Figure 3 shows the results of assay indicating the effect of Compound 5 on the level of triglyceride in blood of HFD-STZ model mice.
Figure 4 shows the results of assay indicating the effect of the compounds of formula (I) on the level of total cholesterol in blood of mice.
Figure 5 shows the results of assay indicating the effect of Compound 5 on the blood glucose level of mice (HFD-STZ model) in a fasting state.
Figure 6 shows the results of oral glucose tolerant tests conducted on Day 0 (baseline) in Test Example 7..
Figure 7 shows the results of oral glucose tolerant tests conducted on Day 28 (endpoint) in Test Example 7.
Figure 8 shows fasting blood glucose levels measured in Test Example 7.
Figure 9 shows serum triglyceride levels measured in Test Example 7. Figure 10 shows hemoglobin Aic levels measured in Test Example 7.
Description of Embodiments
[0051 ] In the subject specification, the term "Ci-6 alkyl" refers to a linear or branched saturated hydrocarbon group having 1 to 6 carbon atoms. Examples thereof include methyl, ethyl, n-propyl, iso- propyl, butyl, iso-butyl, tert-butyl, n-pentyl, pentan-3-yl and the like.
[0052] The term "Ci-6 alkylamino" means a group: -NH-(Ci-6 alkyl), in which Ci-6 alkyl is as defined above.
[0053] The term "C3.7 cycloalkyl" refers to a saturated carbocyclic group having 3 to 7 carbon atoms. Examples thereof include cyclopropyl (cPr), cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl and the like.
[0054] The term "C3-7 cycloalkylamino" means a group: -NH-(C3-7 cycloalkyl), in which "C3-7 cycloalkyl" is as defined above. Examples thereof include cyclopropylamino, cyclobutylamino, cyclopentylamino, cyclohexylamino, cycloheptylamino and the like.
[0055] The term "3- to 7-membered heterocycloalkylamino" refers to an amino group substituted with a saturated heterocyclic group having 3 to 7 ring atoms, which includes one or more hetero atoms selected from -0-, -S-, -NR.6-, -SO- and -SO2-. Examples thereof include pyrrolidinylamino, piperidinylamino, piperazinylamino, N-methylpiperazinylamino, morpholinylamino, thiomorpholinylamino,
imidazolidinylamino, homopiperizynylamino, homopiperazinylamino, and the like.
[0056] The term "a nitrogen-containing 3- to 7-membered heterocyclic ring" refers to a heterocyclic ring having 3 to 7 ring atoms, which includes at least one nitrogen atom and one or more hetero atoms selected from -0-, -S-, -NR.6-, -SO- and -SO2-. Examples thereof include pyrrolidine, piperidine, piperazine, N-methylpiperazine, morphorine, thiomorpholine, imidazolidine, imidazoline,
homopiperidine, homopiperazine and the like.
[0057] In one embodiment of the present invention, R1 is selected from aldehyde, -CH=N-OH, - CH=N-OMe, and -CH=N-OEt.
[0058] In another embodiment of the present invention, R2 is Ci-β alkyl which may be substituted with 1 to 3 fluorine atoms, for example, CH3, CH2CH3, CHF2, CH2F, CF3, and CH2CF3, preferably C¾ or CH2F. [0059] In another embodiment of the present invention, R3 is selected from hydrogen, methyl and ethyl.
[0060] In another embodiment of the present invention, R4 is selected from a group consisting of hydroxy, methylamino, ethylamino, cyclopropylamino, oxetan-3-ylamino, (3-morpholinopropyl)amino, pirrolidin-3-ylamino or (3-(2-methlypipelidin-l -yl)propyl)amino. Preferably, R4 is selected from hydroxyl, ethylamino, cyclopropylamino, and oxetan-3-ylamino.
[0061] In another embodiment of the present invention, R3 and R4 form a group =N-0-Z, in which Z is a hydrogen atom, Ci-6 alkyl or,-(CH2)n-R5, and R5 is as defined above. Examples of the group =N-0-Z include hydroxyimino, methoxyimino, (2-(N,N-dimethylamino)ethoxy)imino, (2- morpholinoethoxy)imino, (2-(4-methylpiperazinyl)ethoxy)imino, (ethoxycarbonylmethoxy)imino, (carboxymethoxy)imino, ((N,N-dimethylamino)acetoxy)imino.
[0062] Specifically, the compound of formula (I) is exemplified by the compounds (Compounds 1 to 28) indicated in Table 1.
Table 1
Figure imgf000021_0001
19 -CH=N-OMe (E-form) Me =N-OCH2-C02H (E-form)
20 -CHO Me H NHEt
21 -CH=N-OMe (E-form) Me H NHEt
22 -CHO Me H
H
23 -CH=N-OMe (E-form) Me H
H
24 -CH=N-OMe (E-form) Me H NH2
25 -CH=N-OMe (E-form) Me H
H
26 -CHO CH2F =N-OCH2-C02H (E-form)
27 -CH=N-OH (E-form) CH2F =N-OCH2-C02H (E-form)
28 -CH=N-OH (E-form) Me =N-OCH2-C02H (E-form)
[0063] More specifically, the compound of formula (I) is exemplified by the following compounds:
1) 3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R)-3-hydroxy-l,2,3,6-tetramethylcyclohexyl)-3- methy lpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde;
2) 3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)-l,2,6-trimethylcyclohexyl)- 3-methylpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde;
3) (E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((l R,2R,6R,E)-3 -(hydroxy imino)-l ,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde oxime;
4) (E)-3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)- l,2,6-trimethylcyclohexyl)-3-methylpenta-2,4-dien-l-yl)-2-methylbenzaldehyde oxime;
5) 3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)-l,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l-yl)-2-methylbenzaldehyde;
6) [(lE,2R,3R,4R)-3-[(lE,3E)-5-(3-chloro-5-formyl-6-hydroxy-2-methoxy-4-methylphenyl)-3- methylpenta-1 ,3-dien-l -yl]-2,3,4-trimethylcyclohexylidene]amino 2-(dimethylamino)acetate; 7) (E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R)-3-hydroxy-l ,2,3,6-tetramethylcyclohexyl)- 3-methylpenta-2,4-dien-l -yl)-4-methoxy-2-methylbenzaldehyde oxime;
8) (E)-3-chloro-6-hydroxy-4-methoxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(methoxyimino)-l,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-] -yl)-2-methylbenzaldehyde O-methyl oxime;
9) (E)-3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(methoxyimino)- 1 ,2,6-trimethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-2-methylbenzaldehyde O-methyl oxime;
10) (E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)- 1 ,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde O-methyl oxime;
11) (E)-3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)- 1 ,2,6-trimethylcyclohexyl)-3 -methylpenta-2,4-dien- 1 -yl)-2-methylbenzaldehyde O-methyl oxime;
12) 3-chloro-4-(fluoromethoxy)-6-hydroxy-2-methyl-5-((2E,4E)-3-methyl-5-((lR,2R,6R,E)-l ,2,6- trimethyl-3-(2-morpholinoethoxyimino)cyclohexyl)penta-2,4-dien-l-yl)benzaldehyde;
13) 3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(cyclopropylamino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien- 1 -yl)-4-(fluoromethoxy)-6-hydroxy-2-methylbenzaldehyde;
14) (E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R)-3-hydroxy-l,2,3,6-tetramethylcyclohexyl)- 3-methylpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde O-methyl oxime;
15) (E)-3-chloro-5-((2E,4E)-5-(( 1 R,2R,6R,E)-3-(2-(dimethylamino)ethoxyimino)- 1 ,2,6- trimethylcyclohexyl)-3 -methy lpenta-2,4-dien- 1 -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde O- methyl oxime;
16) (E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-(( 1 R,2R,6R,E)- 1 ,2,6- trimethyl-3-(2-morpholinoethoxyimino)cyclohexyl)penta-2,4-dien-l-yl)benzaldehyde O-methyl oxime;
17) (E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-((lR,2R,6R,E)-l,2,6- trimethyl-3 -(2-(4-methylpiperazin- 1 -y l)ethoxyimino)cyclohexyl)penta-2,4-dien- 1 -y l)benzaldehyde O- methyl oxime;
18) ethyl 2-([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(l E)- (methoxyimino)methyl]-4-methylphenyl]-3-methylpenta-l ,3-dien-l-yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetate;
19) 2-([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (methoxyimino)methyl]-4-methylphenyl]-3-methylpenta-l,3-dien-l -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid;
20) 3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(ethylamino)-l ,2,6-trimethylcyclohexyl)-3-methylpenta- 2,4-dien-l -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde;
21 ) (E)-3 -chloro-5-((2E,4E)-5-(( 1 R,2R,6R)-3-(ethylamino)- 1 ,2,6-trimethylcyclohexyl)-3 - methy lpenta-2,4-dien- 1 -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde O-methyl oxime;
22) 3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(cyclopropylamino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde;
23) (E)-3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(cyclopropylamino)-l ,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde O-methyl oxime;
24) 2-[(2E,4E)-5-[(l R,2R,6R)-3-amino-l,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien-l-yl]-4- chloro-3-methoxy-6-[( 1 E)-(methoxyimino)methyl]-5-methylphenol;
25) (E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-((lR,2R,6R)-l,2,6- trimethyl-3-(oxetan-3-ylamino)cyclohexyl)penta-2,4-dien-l -yl)benzaldehyde O-methyl oxime;
26) 2-([[(l E,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-fluoromethoxy-5-formyl-4- methylphenyl]-3-methylpenta-l,3-dien-l-yl]-2,3,4-trimethylcyclohexylidene]amino]oxy)acetic acid;
27) 2-([[( 1 E,2R,3 R,4R)-3-[( 1 E,3 E)-5-[3 -chloro-6-hydroxy-2-fluoromethoxy-5-[( 1 E)- (hydroxyimino)methyl]-4-methylphenyl]-3-methylpenta-l,3-dien-l-yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid;
28) 2-([[(l E,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-2-methoxy-6-hydroxy-5-[(lE)- (hydroxyimino)methyl]-4-methylphenyl]-3-methylpenta-l,3-dien-l -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid;
and pharmaceutically acceptable salts thereof.
[0064] The compound of formula (I) is exemplified by the following compounds:
7-1) (E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,3S,6R)-3-hydroxy-l,2,3,6- tetramethylcyclohexyl)-3-methylpenta-2,4-dien-l -yl)-4-methoxy-2-methylbenzaldehyde oxime;
7-2) (E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,3R,6R)-3-hydroxy-l,2,3,6- tetramethylcyclohexyl)-3 -methylpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde oxime; 13-1) 3-chloro-5-((2E,4E)-5-((lR,2R,3S,6R)-3-(cyclopropylamino)-l ,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien- 1 -y l)-4-(fluoromethoxy)-6-hydroxy-2-methylbenzaldehyde;
13- 2) 3-chloro-5-((2E,4E)-5-((lR,2R,3R,6R)-3-(cyclopropylamino)-l ,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l -yl)-4-(fluoromethoxy)-6-hydroxy-2-methylbenzaldehyde;
14- 1) (E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,3S,6R)-3-hydroxy-l,2,3,6- tetramethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde O-methyl oxime;
14-2) (E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,3R,6R)-3-hydroxy-l,2,3,6- tetramethylcyclohexyl)-3 -methylpenta-2,4-dien- 1 -y l)-4-methoxy-2-methy lbenzaldehyde O-methyl oxime;
20-1) 3-chloro-5-((2E,4E)-5-((lR,2R,3S,6R)-3-(ethylamino)-l ,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde;
20- 2) 3-chloro-5-((2E,4E)-5-((lR,2R,3R,6R)-3-(ethylamino)-l ,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l-yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde;
21- 1) (E)-3-chloro-5-((2E,4E)-5-((lR,2R,3S,6R)-3-(ethy]amino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde O-methyl oxime;
21 -2) (E)-3-chloro-5-((2E,4E)-5-(( 1 R,2R,3R,6R)-3-(ethylamino)- 1 ,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien- 1 -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde O-methyl oxime;
22- 1) 3-chloro-5-((2E,4E)-5-((lR,2R,3S,6R)-3-(cyclopropylamino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde;
22- 2) 3-chloro-5-((2E,4E)-5-((lR,2R,3R,6R)-3-(cyclopropylamino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien- 1 -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde;
23- 1) (E)-3-chloro-5-((2E,4E)-5-((lR,2R,3S,6R)-3-(cyclopropy]amino)-l,2,6-trimethy]cyclohexy!)-3- methylpenta-2,4-dien- 1 -yl)-6-hydroxy-4-methoxy-2-methy lbenzaldehyde O-methyl oxime;
23- 2) (E)-3-chloro-5-((2E,4E)-5-((lR,2R,3R,6R)-3-(cyclopropylamino)-l ,2,6-trimethylcyclohexyl)- 3-methylpenta-2,4-dien- 1 -yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde O-methyl oxime;
24- 1) 2-[(2E,4E)-5-[(lR,2R,3S,6R)-3-amino-l,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien-l- yl]-4-chloro-3-methoxy-6-[(lE)-(methoxyimino)methyl]-5-methylphenol; 24- 2) 2-[(2E,4E)-5-[(l R,2R,3R,6R)-3-amino-l,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien-l - yl]-4-chloro-3-methoxy-6-[(lE)-(methoxyimino)methyl]-5-methylphenol;
25- 1 ) (E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-(( 1 R,2R,3 S,6R)- 1 ,2,6- trimethyl-3-(oxetan-3-ylamino)cyclohexyl)penta-2,4-dien-l -yl)benzaldehyde O-methyl oxime;
25-2) (E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyI-5-((l R,2R,3R,6R)-l,2,6- trimethyl-3-(oxetan-3-ylamino)cyclohexyl)penta-2,4-dien-l-yl)benzaldehyde O-methyl oxime;
[0065] According to one aspect of the present invention, the compound of formula (I) is selected from the compounds of Nos. (5), (8), (9), (10), (11), (12), (14), (15), (16), (19), (21), (24), (25), (26), (27), (28) indicated above, and particularly preferably a compound selected from the compounds of Nos. (5), (10), (12), (15), (16), (19), (21 ), and (25) indicated above.
[0066] The compound of formula (I) has activity to increase phosphorylation of AMPK and its downstream effector, acetyl-CoA carboxylase (ACC) [Test Example 1]. Further, the compound has significantly improved metabolic stability against degradation by liver microsome (human and mouse) [Test Example 3], improved PK profile that allows once a day oral treatment [Test Example 4], and improved solubility [Test Example 2] as compared with 4-O-methyl-ascoclorin (MAC) and its prodrug, a Schiff base of MAC with glycine amide (compound #13 in WO2004/074236).
[0067] The compound of formula (I) exhibited blood glucose lowering activity as well as anti- hyperlipidemic activity (lowering blood triglyceride) [Test Example 5].
[0068] The compounds of formula (I) may be administered in the form of a pharmaceutically acceptable salt. The term "pharmaceutically acceptable salt" refers to a salt, which is non-toxic and is prepared with a pharmaceutically acceptable base or acid. When the compound of formula (I) is basic, the pharmaceutically acceptable salt thereof is generally prepared by adding the compound with a suitable organic or inorganic acid. Examples of the acid-addition salt include acetate, benzenesulfonate, benzoate, citrate, fumarate, glutamate, hydrobromide, hydrochloride, lactate, maleate, mandelate, mesylate, oxalate, palmitate, phosphate/diphosphate, salicylate, stearate, sulfate, succinate, tartrate, and tosylate.
[0069] Furthermore, when the compound of formula (I) is acidic, the pharmaceutically acceptable salt thereof is generally prepared by adding the compound with a suitable organic or inorganic base. Examples of the base-addition salt include salts derived from inorganic bases including aluminum, ammonium, calcium, potassium, sodium and the like. Particularly preferred are the ammonium, calcium, potassium, and sodium salts. Examples of the base-addition salt also include salts derived from primary, secondary, and tertiary amines such as arginine, betaine, 2-diethylaminoethanol, 2- dimethylaminoethanol, ethanolamine, N-ethylmorpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, lysine, methylglucamine, morpholine, piperazine, piperidine, triethylamine, tripropylamine, and the like.
[0070] The compound of formula (I) may be administered as a prodrug. The prodrug can be prepared in line with technical knowledge commonly available to a person skilled in the art. For example, in the case of a carboxy group (-COOH) or hydroxy group being present in the compounds of formula (I), a prodrug can be prepared by converting the carboxy group to a pharmaceutically acceptable ester group, such as methyl, ethyl, or pivaloyloxymethyl ester, or by converting the hydroxy group to a
pharmaceutically acceptable ester group, such as acetate or maleate. Use of a prodrug may be beneficial for modifying the solubility or hydrolysis characteristics for use as sustained-release or prodrug formulations.
[0071] In one embodiment, the compound of formula (I) may be used in an amorphous form or a crystalline form. The crystalline form may include solvent to form a solvate such as a hydrate. It should be understood that the present invention covers any use of such a solvate or any mixture such as a solution, suspension or solid mixture containing the compound of formula (I) or metformin.
[0072] According to one aspect of the present invention, there is provided a method for treating or preventing a disease that involves AMP dysregulation, which comprises administering to a patient in need thereof a therapeutically effective amount of a compound represented by formula (I) or a pharmaceutically acceptable salt thereof in combination with administration of metformin. The disease that involves AMPK dysregulation is exemplified as described above. An appropriate route of the administration of the compound represented by formula (I) may be determined by a person skilled in the art. Examples thereof include oral, endorectal, parenteral (intravenous, intramuscular, or subcutaneous), intracisternal, vaginal, intraperitoneal, intravesical, topical (drip infusion, powder, ointment, gel, or cream) application, and inhalation (intraoral or nasal spray). The dosage form of the compound represented by formula (I) for the administration includes tablets, capsules, granules, powders, pills, aqueous and non-aqueous solutions or suspensions for oral use, and parenteral solutions loaded into containers.
[0073] An appropriate route of the administration of metformin may be determined by a person skilled in the art. Examples thereof include oral, endorectal, parenteral (intravenous, intramuscular, or subcutaneous), intracisternal, vaginal, intraperitoneal, intravesical, topical (drip infusion, powder, ointment, gel, or cream) application, and inhalation (intraoral or nasal spray). The dosage form of metformin for the administration includes tablets, capsules, granules, powders, pills, aqueous and nonaqueous solutions or suspensions for oral use, and parenteral solutions loaded into containers.
[0074] According to one aspect of the present invention, there is provided a pharmaceutical composition comprising a compound represented by formula (I) or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient or additive, for use in combination with
administration of metformin. According to another aspect of the present invention, there is provided a pharmaceutical composition comprising metformin and a pharmaceutically acceptable excipient or additive, for use in combination with administration of a compound represented by formula (I) or a pharmaceutically acceptable salt thereof. The composition may be administered in an appropriate route, for example, oral, endorectal, parenteral (intravenous, intramuscular, or subcutaneous), intracisternal, vaginal, intraperitoneal, intravesical, topical (drip infusion, powder, ointment, gel, or cream) application, and inhalation (intraoral or nasal spray). The dosage form for the administration includes tablets, capsules, granules, powders, pills, aqueous and non-aqueous solutions or suspensions for oral use, and parenteral solutions loaded into containers.
[0075] The pharmaceutical composition may be administered as a pharmaceutical formulation. The pharmaceutical formulation can be produced by well-known methods, using additives such as excipients, coating agents, lubricants, binders, disintegrators, stabilizers, corrigents, diluents, solvent and surfactants or emulsifiers.
[0076] Examples of excipients include starches such as potato starch and cornstarch, lactose, crystalline cellulose, and calcium hydrogen phosphate.
[0077] Examples of coating agents include ethylcellulose, hydroxypropylcellulose, hydroxypropylmethylcellulose, shellac, talc, carnauba wax, and paraffin.
[0078] Examples of lubricants include magnesium stearate, and stearic acid.
[0079] Examples of binders include polyvinylpyrrolidone, macrogol, and the same substances exemplified as the excipients.
[0080] Examples of disintegrators include the same substances exemplified as the excipients and croscarmellose sodium, sodium carboxymethylstarch, and chemically modified starch or celluloses.
[0081] Examples of stabilizers include paraoxybenzoic acid esters such as methylparaben and propylparaben; alcohols such as chlorobutanol, benzylalcohol, and phenylethylalcohol; benzalkonium chloride; phenols such as phenol and cresol; thimerosal; dehydroacetic acid; and ascorbic acid.
[0082] Examples of corrigents include sweeteners which are conventionally used.
[0083] Examples of diluents include the substances exemplified as excipients.
[0084] Examples of solvents include ethanol, phenol, chlorocresol, purified water, and distilled water.
[0085] Examples of surfactants or emulsifiers include polysorbate 80, polyoxyl 40 stearate, lauromacrogol, soybean oil, Maisin 35-1 (Gattefosse, France) and Cremophor (BASF, Germany).
[0086] The dose of the compounds of formula (I) or pharmaceutically acceptable salts thereof differs depending on the symptoms, age, body weight, relative health condition, presence of other medication, method of administration, and the like. For example, a generally effective dose for an oral agent as the active ingredient for a patient (a warmblooded animal, especially a human) is a daily dose of preferably 0.-1 to 1000 mg per kg body weight, more preferably 1 to 30 mg per kg body weight. The daily dose for an adult patient with a normal body weight is preferably in the range of 10 to 1000 mg. For a parenteral agent, the daily dose is preferably 0.1 to 1000 mg per kg body weight, more preferably 10 to 800 mg per kg body weight. Preferably such a dose is administered at one time or in several portions per day, or intermittent schedule depending on the symptoms.
[0087] Metformin may be administered, by a route and in an amount that are commonly known therefor, simultaneously or sequentially with a compound of formula (I). When the compound is administered simultaneously with metformin, these ingredients may be contained in a single unit dosage form or two or more separate dosage forms. In the combination therapy, the compound of formula (I) and metformin are administered on different overlapping schedules. It is also contemplated that in the combination therapy, the compounds of formula (I) and metformin may be administered in lower doses than that administered each alone.
[0088] The dosage of the compounds of formula (I) and metformin used for the subject invention is determined depending on age of a subject, body weight of a subject, symptom, degree of seriousness, therapeutic effect, administration route, administration regimen, and period of the treatment,
[0089] According to one aspect of the subject invention, the compound represented by formula (I) is administered in an amount from 0.1 to 100 mg kg/day, particularly 0.5 to 30 mg/kg/day, more particularly 1 to 16 mg/kg/day.
[0090] According to one aspect of the subject invention, metformin is administered in an amount from 50 to 1000 mg/kg/day, particularly 100 to 500 mg/kg/day, more particularly 250 to 350 mg/kg/day.
[0091 ] According to one aspect of the subject invention, the compound represented by formula (I) is administered in an amount from 0.1 to 100 mg/kg/day, and metformin is administered in an amount from 50 to 1000 mg/kg/day. According to another aspect of the subject invention, the compound represented by formula (I) is administered in an amount from 0.5 to 30 mg/kg/day, and metformin is administered in an amount from 100 to 500 mg/kg/day. According to another aspect of the subject invention, the compound represented by formula (I) is administered in an amount from 1 to 16 mg/kg/day, and metformin is administered in an amount from 250 to 350 mg/kg/day.
[0092] Synthetic processes for preparing the compounds represented by formula (I) are illustrated in the following schemes and examples.
[0093] A starting material, ascochlorin can be obtained by fermentation of Ascochyts viciae Libert as described in JP S45-009832 B published in 1970. In the following schemes, R2, R3, R4, R5, R7, Y, and Z are as defined hereinbefore. Scheme l
Figure imgf000031_0001
[0094] Scheme l shows a method for producing compound (IV), (Via) or (Vlb). The compound (II) in which R2 is methyl (4-O-methyl-ascochlorin) can be prepared by the method described in JP H6- 305959 A. The compound (II) in which R2 is fluorinated alkyl group can be obtained by O-alkylation with a fluoroalkylating agent in the presence of base in an appropriate solvent. Examples of the base include potassium carbonate, cesium carbonate and the like. Examples of fluoroalkylating agents include bromofluoromethane, bis(trifluoromethyl)-bis(trifluoromethyloxy)-sulfane, 5-tert-butyl-2'- (trifluoromethoxy)biphenylyl-2-diazonium hexafluoroantimonate, 2,2,2-tris(fluoranyl)ethyl methanesulfonate, 2-diazo- 1 , 1,1 -tris(fluoranyl)ethane, tris(fluoranyl)methane, 1 -chloranyl-4-[chloranyl- bis(fluoranyl)methyl]sulfonyl-benzene and the like.
[0095] The compound (IV) can be prepared by reacting a compound (II) with NH2OY (III) in the presence of base such as pyridine, sodium carbonate, sodium hydrogencarbonate and the like. The compound (Vlb) can be prepared by reacting compound (IV) with NH2OZ (V) in the presence of base such as pyridine, sodium carbonate, sodium hydrogencarbonate and the like. The compound (Via) can be directly prepared from the compound (II) by reacting two or more equivalents of hydroxyamine (III) in the same reaction condition as described above.
[0096] The compound (Vlb) wherein Z is -<CH2)nC02H can be prepared by basic hydrolysis of the corresponding ester derivative (Vlb) wherein Z is -(CH2)nC02R6. Scheme 2
Figure imgf000032_0001
[0097] Scheme 2 shows a method for producing compound (X). The compound (VIII) in which R2 is methyl or fluorinated alkyl can be prepared from ascochlorin via O-acetyl derivative (VII) wherein R2 is methyl or fluorinated alkyl by the method described in JP 2005- 225851 A. The intermediate (IX) can be prepared by reacting the intermediate (VIII) with NH2O-Z (V) in the presence of base such as pyridine, sodium carbonate, sodium hydrogencarbonate and the like.. The compound (X) can be prepared by acidic hydrolysis of the acetal group of compound (IX) in an appropriate solvent by the method known to a person skilled in the art.
Figure imgf000033_0001
[0098] Scheme 3 shows another method for producing compound (X). The compound (X) can be prepared from the corresponding oxime derivative (VI) by treatment with hydrochloric acid in THF/H2O at 100°C.
Scheme 4
Figure imgf000033_0002
[0099] Scheme 4 shows a method for producing compounds (XII) and (XIII). The intermediate (XI) can be prepared by reacting the intermediate (VIII) with an alkylating agent, such as alkyl lithium, alkyl Grignard agent or alkyl cupper lithium reagent, in an appropriate solvent. The compound (XII) can be prepare by acidic hydrolysis of the acetal group of the intermediate (XI) in an appropriate solvent.
[0100] The compound (XIII) can be prepared by reacting the intermediate (IV) with an alkylating agent stated above in an appropriate solvent. The compound (XIII) can be also prepared by reacting the compound (XII) with NH2-OY (III) in the presence of base such as pyridine, sodium carbonate, sodium hydrogen carbonate and the like. The compound (XII) can be also prepared from the compound (XIII) under similar reaction condition as described in Scheme 3.
Figure imgf000034_0001
[0101] Scheme 5a shows a method for producing compound (XV) and (XVI) wherein R11 is Ci-e alkyl,
C3-7 cycloalkyl, or 3- to 7-membered heterocycloalkyl which contains -0-, -S-, -NR.6-, or -SO2- as a ring atom. The intermediate (XIV) can be prepared by reductive amination of the intermediate (VIII) with a primary amine, R"-NH2, in one step or stepwise via intermediate imine (XVII), using the reducing agent such as sodium cyanoborohydride, sodium triacetoxyborohydride and the like.
[0102] The compound (XV) can be prepared by acidic hydrolysis of the acetal group of compound
(XIV) in an appropriate solvent by the method known to the skilled art. The compound (XVI) can be prepared by reductive amination of the intermediate (IV) with a primary amine, RU-NH2 by the same method as described above either directly or step wise fashion via intermediate (XVIII).
[0103] The compound (XV) can be also prepared from the oxime ether derivative represented by the formula (XVI) under similar reaction condition as described in Scheme 3. Scheme 5b
Figure imgf000035_0001
[0104] The compound (XVI) wherein R11 is Ci-e alkyl, C3-7 cycloalkyl or 3- to 7-membered heterocycloalkylamino which contains -0-, -S-, -NR6-, -SO- or -SO2- as a ring atom such as oxetane can be prepared from the intermediate (IV) in 2 steps: (i) by reductive amination with ammonium acetate/sodium cyanoborohydride in ethanol, (ii) followed by second reductive amination of the resulting intermediate (XIX) with a ketone corresponding to R" such as oxetan-3-one and sodium triacetoxyborohydride in dichloromethane.
Scheme 6
Figure imgf000035_0002
[0105] Scheme 6 shows a method for producing compound (XXI), wherein R11 is C\.e alkyl, C3-7 cycloalkyl, or 3- to 7-membered heterocycloalkyl which contains -0-, -S-, -NR6-, or -SO2- as a ring atom. The intermediate (XX) can be prepared by Scheme 2 or 3. The compound (XXI) can be prepared by esterification of XX with a carboxylic acid of formula R12C02H by an active ester method or with condensation agent that are known to those skilled in the art.
[0106] When the compound of the present invention represented by formula (I) is obtained in a free form, it can be converted to a salt is a conventional method.
Examples [0107] Herein below, the present invention will be more specifically described using Examples, however, it is not to be construed as being limited thereto.
[0108] NMR analysis was performed using Brucker, A VANCE 400MHz or 300MHz, and NMR data were expressed as chemical shifts in ppm (parts per million) (δ), and the deuterium lock signal from the sample solvent was referenced. Mass spectrum data was obtained using a LC-MS, Shimazu LCMS-2020 or -2010 equipped with a photodiode array, SPD-M20A, ESI-SQD 2020 or 2010 (Shimadzu)
[0109] Commercially available reagents were used without further purification "Room temperature" means temperatures ranging from about 20°C to 25°C. All non-aqueous reactions were carried out under a nitrogen atmosphere. Concentration or distillation of solvents under reduced pressure means that a rotary evaporator was used.
[0110] In the preparation of compounds, a functional group was protected with a protecting group as required to obtain a target compound, followed by removal of the protecting groups. Selection of the protecting group, as well as procedure for the introduction or the removal of the protecting group were carried out, for example, according to a method described in Greene and Wuts, "Protective groups in Organic Synthesis" (second edition, John Wiley & Sons, 1991).
Example 1
[0111] Synthesis of 3-chloro-4-(fluoromethoxy)-6-hydroxy-2-methyl-5-[(2E,4E)-3-methyl-5- lR,2R,6R)-l ,2,6-trimethyl-3-oxocyclohexyl]penta-2,4-dien-l-yl]benzaldehyde.
Figure imgf000036_0001
[0112] A solution of 3-chloro-4,6-dihydroxy-2-methyl-5-[(2E,4E)-3-methyl-5-[(l R,2R,6R)-l,2,6- trimethyl-3-oxocyclohexyl]penta-2,4-dien-l-yl]benzaldehyde, (ascochlorine, 6 g, 14.82 mmol) in acetonitrile (120 mL), bromofluoromethane (excess), cesium carbonate (2.4 g, 7.38 mmol) was placed in a 250-mL sealed tube. The resulting mixture was stirred for 16 h at 25°C. The mixture was filtered, and the filtrate was concentrated under vacuum. The resulting residue was purified by silica gel column chromatography with Petroleum ether/ethyl acetate (5:1). The product was recrystallized from dichloromethane/hexane in the ratio of 1 :5 to give the titled compound (4.5 g, 70%) as an off-white solid.
LC-MS (ES, m/z): 435 [M-H]"
H-NMR (400MHz, DMSO, ppm): δ 12.60 (s, 1H), 10.30 (s, 1H), 5.86 (m, 2H), 5.72 (s, 1H), 5.42 (m, 2H), 3.50 (d, J = 7.2 Hz, 2H), 2.64 (s, 3H), 2.54-2.49 (m, 2H), 2.17 (m, 1H), 2.05 (m, 1H), 1.84 (s, 4H), 1.52 (m, 1H), 0.73-0.67 (m, 6H), 0.59 (s, 3H)
F-NMR (400MHz, DMSO, ppm): δ -148.31.
Example 2
[0113] Synthesis of (2R,3R,4R)-3-[(l E,3E)-5-[3-chloro-5-(diethoxymethyl)-2-(fluoromethoxy)-6- h droxy-4-methylphenyl]-3-methylpenta-l ,3-dien-l-yl]-2,3,4-trimethylcyclohexan-l -one
Figure imgf000037_0001
[0114] Step l
A solution of 3-chloro-4-(fluoromethoxy)-6-hydroxy-2-methyl-5-[(2E,4E)-3-methyl-5- [(lR,2R,6R)-l,2,6-trimethyl-3-oxocyclohexyl]penta-2,4-dien-l-yl]benzaldehyde [Example 1] (600 mg, 1.37 mmol) in pyridine (12 mL) was placed in a 50-mL round-bottom flask purged and maintained with an atmosphere of nitrogen, and acetic anhydride (196 mg, 1.92 mmol) was added at 25°C. The resulting solution was stirred for 1.5 h at 25°C. Then the mixture was diluted with of dichloromethane (50 mL) and washed with aqueous hydrochloric acid (0.3mol/L, 20 mL x5). The organic layer was dried with anhydrous sodium sulfate and concentrated under vacuum to give 4-chloro-3- (fluoromethoxy)-6-formyl-5-methyl-2-[(2E,4E)-3-methyl-5-[(l R,2R,6R)-l,2,6-trimethyl-3- oxocyclohexyl]penta-2,4-dien-l -yl]phenyl acetate (500 mg, 76%) as a white solid.
[0115] Step 2
Into a solution of 4-chloro-3-(fluoromethoxy)-6-formyl-5-methyl-2-[(2E,4E)-3-methyl-5- [(lR,2R,6R)-l ,2,6-trimethyl-3-oxocyclohexyl]penta-2,4-dien-l-yl]phenyl acetate (500 mg, 1.04 mmol) in ethanol (22.5 mL) was placed in a 250-mL round-bottom flask purged and maintained with an atmosphere of nitrogen, and triethylamine (0.02 mL) was added at 25°C. The solution was stirred for 5 h at 25°C, and then concentrated under vacuum to give (2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-5- (diethoxymethyl)-2-(fluoromethoxy)-6-hydroxy-4-methylphenyl]-3-methylpenta-l ,3-dien-l-yl]-2,3,4- trimethylcyclohexan-l-one (410 mg, 77%) as a yellow oil.
Example 3
[0116] Synthesis of 3-chloro-6-hydroxy-5-[(2E,4E)-5-[(l R,2R,3S,6R)-3-hydroxy-l,2,3,6- tetramethylcyclohexyl]-3-methylpenta-2,4-dien- 1 -yl]-4-methoxy-2-methylbenzaldehyde (Compound 1 - 1)
Figure imgf000038_0001
[0117] The intermediate (VIII) wherein R2 is methyl: (4R,5R,6S)-5-[(l E,3E)-5-[3-chloro-5- (diethoxymethyl)-6-hydroxy-2-methoxy-4-methylphenyl]-3-methylpenta-l,3-dien-l -yl]-4,5,6- trimethyloxan-3-one was prepared from ascochiorin by the procedures described in JP 2005-225851 A.
[01 18] Stepl
A solution of (2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-5-(diethoxymethyl)-6-hydroxy-2-methoxy- 4-methylphenyl]-3-methylpenta-l,3-dien-l -yl]-2,3,4-trimethylcyclohexan-l-one (1 g, 2.03 mmol) in tetrahydrofuran (30 mL) was placed in a 50-mL 3-necked round-bottom flask purged and maintained with an atmosphere of nitrogen, followed by addition of methylmagnesium bromide (2 mL, 3.00 equiv, 3 ) dropwise with stirring at -78°C. The resulting solution was stirred for 3 h at 0°C. The reaction was then quenched by addition of sat. aqueous ammonium chloride (30 mL). The resulting mixture as extracted with diethyl ether (30 mL x3) and the organic layers were combined and washed with saturated aqueous sodium chloride (50 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under vacuum. The resulting residue was purified by Flash-Prep- HPLC with the following condition to give 4-chloro-2-(diethoxymethyl)-6-[(2E,4E)-5-[(lR,2R,3S,6R)- 3-hydroxy-l,2,3,6-tetramethylcyclohexyl]-3-methylpenta-2,4-dien-l-yl]-5-methoxy-3-methylphenol (0.6 g, 60%) as a light yellow solid (mp. 39-40°C).
Flash-Prep-HPLC condition (IntelFlash-1): Column, CI 8 silica gel; mobile phase, acetonitrile/water =70/30 increasing to C¾CN /water = 90/10 within 10 min and then CH3CN within 10 min; Detector, UV 210 nm. LC-MS (ESI, m/z): 507 [M-H]-
H-NMR (CD3CN, 400 MHz, ppm): δ 5.91 (d, J = 16.4 Hz, 1H), 5.83 (s, 1H), 5.38-5.43 (m, 1H), 5.30 (d, J = 16 Hz, 1H), 3.72-3.79 (m, 5H), 3.62-3.68 (m, 2H), 3.46 (d, J = 5.6 Hz, 2H), 2.33 (s, 3H), 1.87 (s, 3H), 1.45-1.70 (m, 3H), 1.29-1.35 (m, 2H), 1.20-1.25 (m, 7H), 1.12 (s, 3H), 0.90 (s, 3H), 0.80 (d, J = 5.6 Hz, 3H), 0.70 (d, J = 5.4 Hz, 3H).
[0119] Step 2
A solution of 4-chloro-2-(diethoxymethyl)-6-[(2E,4E)-5-[(lR,2R,3S,6R)-3-hydroxy- 1,2,3,6- tetramethylcyclohexyl]-3-methylpenta-2,4-dien-l-yl]-5-methoxy-3-methylphenol (355 mg, 0.699 mmol) in tetrahydrofuran/water (10 mL /20 mL) was placed in a 50-mL round-bottom flask purged and maintained with an atmosphere of nitrogen, followed by addition of acetic acid (5 mL). The resulting solution was stirred for 5 h at 25°C. The resulting solution was concentrated under vacuum. The resulting residue was purified by Flash-Prep-HPLC with the following condition to give 3-chloro-6- hydroxy-5-[(2E,4E)-5-[(lR,2R,3S,6R)-3-hydroxy-l,2,3,6-tetramethylcyclohexyl]-3-methylpenta-2,4- dien-l-yl]-4-methoxy-2-methylbenzaldehyde (0.26 g, 76%, Compound 1) as an off-white solid (mp. 42- 43°C).
Flash-Prep-HPLC condition (IntelFlash-1): Column, C18 silica gel; mobile phase,
CH3CN/water = 70/30 increasing to CH3CN /water =90/10 within 10 min; Detector, UV 210 nm.
LC-MS (ES m/z): 433 [M-H]"
H-NMR (CD3CN, 400 MHz, ppm): δ 10.25 (s, 1H), 5.89 (d, J = 16.0 Hz, 1H), 5.40 (t, J = 1.2 Hz, 1H), 5.29 (d, J = 16.0 Hz, 1H), 3.84 (s, 3H), 3.48 (d, J = 7.2 Hz, 2H), 2.63 (s, 3H), 1.87 (s, 3H), 1.57-1.78 (m, 3H), 1.33-1.47 (m, 3H), 1.09 (s, 3H), 0.79 (d, J = 3.2 Hz, 3H), 0.68 (d, J = 6.8 Hz, 3H).
Example 4
[0120] Synthesis of 3-chloro-6-hydroxy-5-[(2E,4E)-5-[(lR,2R,3E,6R)-3- (hydroxyimino)- 1,2,6- trimethylcyclohexyl]-3-methylpenta-2,4-dien-l -yl]-4-methoxy-2-methylbenzaldehyde (Compound 2)
Figure imgf000040_0001
[0121] Stepl
A solution of (2R,3R,4R)-3-[(l E,3E)-5-[3-chloro-5-(diethoxymethyl)-6-hydroxy-2-methoxy- 4-methylphenyl]-3-methylpenta-l,3-dien-l-yl]-2,3,4-trimethylcyclohexan-l-one (1 g, 2.03 mmol) in pyridine (30 mL) was placed in a 50-mL 3-necked round-bottom flask purged and maintained with an inert atmosphere of nitrogen, followed by addition of hydroxyamine hydrochloride (200 mg, 2.88 mmol). The mixture was stirred for 16 h at 25°C. The reaction mixture was concentrated under vacuum. The residue was purified by Flash-Prep-HPLC with the following condition to give 4-chloro-2- (diethoxymethyl)-6-[(2E,4E)-5-[(lR,2R,3E,6R)-3-(hydroxyimino)-l,2,6-trimethylcyclohexyl]-3- methylpenta-2,4-dien-l-yl]-5-methoxy-3-methylphenol (0.40 g, 39%) as an off-white solid (mp. 68- 70°C).
Flash-Prep-HPLC condition (IntelFlash-1 ): Column, CI 8 silica gel; mobile phase, CH3CN /water =70/30 increasing to CH3CN /water = 90/10 within 15 min and then acetonitrile within 10 min; Detector, UV 210 nm.
LC-MS: (ESI, m/z): 506 [M-H]"
H-NMR: (CD3CN, 400 MHz, ppm): δ 5.95 (d, J = 16.0 Hz, 1H), 5.81 (s, 1H), 5.42 (q, J = 12.8 Hz, 4.8 Hz, 2H), 3.69-3.77 (m, 5H), 3.59-3.66 (m, 2H), 3.45 (d, J = 7.2 Hz, 2H), 3.27-3.32 (m, 1H), 2.47 (s, 3H), 2.27 (d, J = 6.8 Hz, 1H), 1.88 (s, 3H), 1.60-1.72 (m, 3H), 1.19-1.29 (m, 7H), 0.82 (d, J = 6.8 Hz, 3H), 0.72 (d, J = 6.8 Hz, 3H), 0.67 (s, 3H).
[0122] Step 2
A solution of 4-chloro-2-(diethoxymethyl)-6-[(2E,4E)-5-[(lR,2R,3E,6R)-3-(hydroxyimino)- l,2,6-trimethylcycIohexyl]-3-methyIpenta-2,4-dien-l-yl]-5-methoxy-3-methylphenol (370 mg, 0.726 mmol) in tetrahydrofuran/water (10 mL /20 mL), was placed in a 100-mL 2-necked round-bottom flask purged and maintained with an atmosphere of nitrogen, followed by addition of acetic acid (10 mL). The mixture was stirred for 5 h at 25°C. The reaction mixture was concentrated under vacuum. The residue was purified by Flash-Prep-HPLC with the following condition to give 3-chloro-6-hydroxy-5- [(2E,4E)-5-[(lR,2R,3E,6R)-3-(hydroxyimi
4-methoxy-2-methylbenzaldehyde (220 mg, 70%, Compound 2) as a light yellow solid (mp. 158-160°C).
Flash-Prep-HPLC condition (IntelFlash-1 ): Column, C18 silica gel; mobile phase, CH3CN / water = 70/30 increasing to CH3CN /water = 90/10 within 10 min; Detector, UV 210 run.
LC-MS (ESI, m/z): 432 [M-H]"
H-NMR (CD3CN, 400 MHz, ppm): δ 10.25 (s, 1H), 5.94 (d, J = 16.4 Hz, 1H), 5.44 (q, J = 15.6 Hz, 6.8 Hz, 2H), 3.85 (s, 3H), 3.50 (d, J = 7.2 Hz, 2H), 3.29 (t, J = 8.4 Hz, 1H), 2.63 (s, 3H), 2.21 (q, J = 13.6 Hz, 6.8 Hz, 1H), 1.90 (s, 3H), 1.60-1.69 (m, 3H), 1.20-1.33 (m, 1H), 0.81 (d, J = 6.8 Hz, 3H), 0.71 (d, J = 6.8 Hz, 3H), 0.66 (s, 3H).
Example 5
[0123 ] Synthesis of 4-chloro-2-[(2E,4E)-5-[( 1 R,2R,3E,6R)-3 -(hydroxy imino)- 1 ,2,6- trimethylcyclohexyl]-3-methylpenta-2,4-dien-l-yl]-6-[(lE)-(hydroxyimino)methyl]-3-methoxy-5- methylphenol (Compound 3)
Figure imgf000041_0001
[0124] A solution of 3-chloro-6-hydroxy-4-methoxy-2-methyl-5-[(2E,4E)-3-methyl-5-[(lR,2R,6R)- l,2,6-trimethyl-3-oxocyclohexyl]penta-2,4-dien-l -yl]benzaldehyde (700 mg, 1.67 mmol, ascochlorine) in pyridine (14 ml) was placed in a 25-mL round-bottom flask, followed by addition of hydroxyamine hydrochloride (290 mg, 4.17 mmol) at 25°C. The mixture was stirred for 16 h at 25°C, and then concentrated under vacuum. After dilution with dichloromethane (100 ml), the resulting solution was washed with aqueous hydrochloric acid (IN, 50 mLx2) and brine (50 mL). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under vacuum. The resulting residue was purified by Flash-Prep-HPLC with the following condition to give the titled compound (221.8 mg, 29.6%) as a white solid.
Flash-Prep-HPLC condition (IntelFlash-1): Column, silica gel; mobile phase, CH3CN
/water(0.08% ammonium bicarbonate) = 50/50 increasing to a CH3CN / water (0.08% ammonium bicarbonate) = 80/20 within 25 min; Detector, UV 254 nm.
LC-MS (ES, m/z): 449 [M+H]+
H-NMR (CD3OD, 400MHz, ppm): 8.56 (s, IH), 5:92 (d, J=16 Hz, IH), 5.46 (m,
2H), 3.78 (s, 3 H), 3.52 (d, J=7.2 Hz, 2H), 3.41 (m, IH), 3.31 (m, IH), 2.43 (s, 3H), 2.20 (m, IH), 3H), 1.67 (m, 3H), 1.33 (m, IH), 0.91-0.69 (m, 9H)
Example 6
[0125] Synthesis of 3-chloro-4-(fluoromethoxy)-6-hydroxy-5-[(2E,4E)-5-[(l R,2R,3E,6R)-3- (hydroxyimino)-l,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien-l -yl]-2-methylbenzaldehyde (Compound 5)
Figure imgf000042_0001
[0126] Step 1
A solution of (2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-5-(diethoxymethyl)-2-(fluoromethoxy)-6- hydroxy-4-methylphenyl]-3-methylpenta-l ,3-dien-l -yl]-2,3,4-trimethylcyclohexan- 1 -one [Example 2] (410 mg, 0.80 mmol) in pyridine (12.3 mL) was placed in a 25-mL round-bottom flask, followed by addition of hydroxyamine hydrochloride (83.7 mg, 1.20 mmol, 1.50 equiv) at 25°C. The mixture was stirred overnight at 25°C, and then diluted with dichloromethane (300 mL). The organic layer was washed with aqueous hydrogen chloride (0.3mol/L, 200 mLx5). The organic layer was combined and concentrated under vacuum to give 4-chloro-2-(diethoxymethyl)-5-(fluoromethoxy)-6-[(2E,4E)-5- [(lR,2R,3E,6R)-3-(hydroxyimino)-l,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien-l-yl]-3- methylphenol (330 mg, 78%) as a yellow solid.
[0127] Step 2
A solution of 4-chloro-2-(diethoxymethyl)-5-(fluoromethoxy)-6-[(2E,4E)-5-[(lR,2R,3E,6R)- 3-(hydroxyimino)-l,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien-l -yl]-3-methylphenol (330 mg, 0.63 mmol) in tetrahydrofuran (6.6 mL) was placed in a 250-mL round-bottom flask, and then a solution of glacial acetic acid (6.6 mL, 5.56 equiv) in water (13.2 mL) was added at 25°C. The mixture was stirred for 4 h at 25°C, and then extracted with of dichloromethane (100 mLx5). The organic layer was combined and the pH value of the solution was adjusted to 7-8 with aqueous sodium bicarbonate. The resulting organic layer was separated, dried and concentrated under vacuum. The resulting residue was purified by Flash-Prep-HPLC with the following condition to give 3-chloro-4-(fluoromethoxy)-6- hydroxy-5-[(2E,4E)-5-[(l R,2R,3E,6R)-3-(hydroxyimino)- l ,2,6-trimethylcyclohexyl]-3-methylpenta- 2,4-dien-l-yl]-2-rriethylbenzaldehyde (200 mg, 71%, Compound 5) as a yellow solid.
Flash-Prep-HPLC condition (IntelFlash-1): Column, C 18 silica gel; mobile phase,
CH3CN/H2O = 50/50 increasing to CH3CN/H2O = 80/20 within 28 min; Detector, UV 254 nm.
LC-MS (ES, m/z): 452 [M+H] +
H-NMR (DMSO, 300MHz, ppm): 12.59(s, IH) , 10.34 (s, IH), 10.30 (s, IH), 5.85-5.90 (t, 2H), 5.70 (s, IH), 5.43 (s, I H), 5.38 (d, J=4.5Hz, 2H) , 3.48-3.51 (d, J=7.5Hz,2H), 3.31-3.20 (m, I H), 2.72-2.64 (d, J=24.9Hz, IH), 2.26-2.18 (m, IH), 1.84 (s, 3H), 1.63-1.54 (m, 3H), 1.24-1.19(d, J=13.2Hz, IH)
Example 7
[0128] Synthesis of [(lE,2R,3R,4R)-3-[(l E,3E)-5-(3-chloro-5-formyl-6-hydroxy-2-methoxy-4- methylphenyl)-3-methylpenta-l ,3-dien-l -yl]-2,3,4-trimethylcyclohexylidene]amino 2-(dimethylamino)- acetate (compound 6)
Figure imgf000043_0001
[0129] Step 1
A solution of 2-(dimethylamino)acetic acid (15 mg, 0.15 mmol) in dichloromethane (0.4 mL) was placed in a 25-mL round-bottom flask, followed by addition of l-(3-dimethylaminopropyl)-3- ethylcarbodiimide methiodide (45 mg, 0.15 mmol), 1 -hydroxybenzotrizole (21 mg, 0.16 mmol) and N- methylmorpholine (15 mg, 0.15 mmol) at 25°C. The mixture was stirred for 30 min at 25°C. Then 3- chloro-6-hydroxy-5-[(2E,4E)-5-[(lR,2R,3E,6R)-3-(hydroxyimino)-l ,2,6-trimethylcyclohexyl]-3- methylpenta-2,4-dien-l -yl]-4-methoxy-2-methylbenzaldehyde, [Compound 2, Example 3] (60 mg, 0.14 mmol) was added. The resulting mixture was stirred for 2 h at 25°C, then washed with water (10 mL) and brine (10 ml). The organic layer was dried over anhydrous sodium sulfate, filtered and concentrated under vacuum. The resulting residue was purified by Prep-HPLC with the following condition to give the titled compound (30 mg, 42%) as a yellow solid.
Prep-HPLC condition: Column, X Bridge C I 8, 19* 150mm 5um; mobile phase, CH3CN / water (0.05% ammonium bicarbonate)=50/50 increasing to CH3CN /water (0.05% ammonium bicarbonate)= 100/0 within 30 min. Detector, UV 254nm.
LC-MS: (ES, m/z): 519 [M+H]+
H-NMR: (CD3OD, 400MHz, ppm): 10.30 (s, IH), 5.96 (d, J=16Hz, IH), 5.46 (m, 2H), 3.83 (s, 3H), 3.52 (d, J=7.2Hz, 2H), 3.34(s, 2H), 3.31 (m, I H), 2.64 (s, 3H), 2.47-2.32 (m, 7H), 1.95 (m, 4H), 1.76 (m, 2H), 1.39 (m, IH), 0.97 (d, J=6.8Hz, 3H), 0.75-0.88 (m, 6H).
[0130] Step 2
A solution of hydrogen chloride in tetrahydrofuran(0.0225M, 13 mL) was added to
[(lE,2R,3R,4R)-3-[(lE,3E)-5-(3-chloro-5-formyl-6-hydroxy-2-methoxy-4-methylphenyl)-3- methylpenta-l ,3-dien-l-yl]-2,3,4-trimethylcyclohexylidene]amino 2-(dimethylamino)acetate (150 mg, 0.29 mmol) in a 50-mL round-bottom flask. The mixture was stirred for 5 min at 25°C, then concentrated under vacuum to give [(lE,2R,3R,4R)-3-[(l E,3E)-5-(3-chloro-5-formyl-6-hydroxy-2- methoxy-4-methylphenyl)-3-methylpenta-l ,3-dien-l -yl]-2,3,4-trimethylcyclohexylidene]amino 2- (dimethylamino)acetate hydrochloride (101.6 mg, 63%) as a light yellow solid.
LC-MS: (ES, m/z):519 [M+H]+
H-NMR: (CD3OD, 300MHz, ppm): 10.30 (s, IH), 5.96 (d, J=17.7Hz, 1H),4.34 (m, 2H), 3.83 (s, 3H), 3.52 (m, 2H), 2.81-3.15 (m, 6H), 3.31 (m, I H), 2.64 (s, 3H), 2.45 (m, I H), 1.60-2.11 (m, 7H), 1.39 (m, 1 H), 0.97 (m, 3H), 0.78 (m, 6H).
Example 8
[0131] Synthesis of 4-chloro-3-(fluoromethoxy)-2-[(2E,4E)-5-[(l R,2R,3E,6R)-3-(hydroxyimino)- 1 ,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien- 1 -yl]-6-[( 1 E)-(hydroxyimino)methyl]-5- methylphenol (Compound 4)
Figure imgf000045_0001
[0132] A solution of 3-chloro-4-(fluoromethoxy)-6-hydroxy-2-methyl-5-[(2E,4E)-3-methyl-5- [(lR,2R,6R)-l,2,6-trimethyl-3-oxocyclohexyl]penta-2,4-dien-l-yl]benzaldehyde [Example 1] (250 mg, 0.57 mmol) in pyridine (7.5 mL) was placed in a 25 mL round-bottom flask, followed by addition of hydroxyamine hydrochloride (80 mg, 1.15 mmol) at 25°C. The mixture was stirred for 16 h at 25°C, and concentrated under vacuum. The resulting residue was purified by Flash-Prep-HPLC with the following condition to give the titled compound (201.7 mg, 75%, Compound 4) as a white solid.
Flash-Prep-HPLC condition (Intel Flash- 1) to give: Column, silica gel; mobile phase, CH3CN /water (0.08% ammonium bicarbonate) = 70/30 increasing to CH3CN /water (0.08% ammonium bicarbonate) = 90/10 within 15 min; Detector, UV 220 nm.
LC-MS: (ES, m/z): 467 [M+H]+
H-NMR: (CD3OD, 400MHz,ppm): 8.55 (s, 1H), 5.90 (d, J=16Hz, 1H), 5.72 (s, 1H), 5.58 (s, 1H), 5.39-5.47 (m, 1H), 3.58 (d, J=7.2Hz, 2H), 3.36-3.42 (m, 1H), 2.43 (s, 3H), 2.18 (t, 1H), 1.89 (s, 3H), 1.61-1.70 (m, 3H), 1.29-1.35 (m, 1H).
Example 9
[0133] Synthesis of 4-chloro-2-[(2E,4E)-5-[(lR,2R,3S,6R)-3-hydroxy-l,2,3,6-tetramethylcyclohexyl]- 3-methylpenta-2,4-dien-l-yl]-6-[(l E)-(hydroxyimino)methyl]-3-methoxy-5-methylphenol (Compound 7-1
Figure imgf000045_0002
[0134] A solution of 3-chloro-6-hydroxy-5-[(2E,4E)-5-[(lR,2R,3S,6R)-3-hydroxy-l,2,3,6- tetramethylcyclohexyl]-3-methylpenta-2,4-dien-l -yl]-4-methoxy-2-methylbenzaldehyde [Compound 1 , Example 2] (160 mg, 0.37 mmol) in pyridine (9.6 mL) was placed in a 25-mL round-bottom flask, followed by addition of hydroxyamine hydrochloride (25.6 mg, 0.37 mmol, 1.00 equiv) at 25°C. The mixture was stirred for 16 h at 25°C and then concentrated under vacuum. The resulting residue was purified by Flash-Prep-HPLC with the following condition to give the titled compound (114.0 mg, 69%, Compound 7-1) as a white solid.
Flash-Prep-HPLC condition (IntelFlash-1): Column, silica gel; mobile phase, C¾CN /water (0.08% NH4HCO3) = 70/30 increasing to CH3CN /water (0.08% NH4HCO3) = 90/10 within 15 min; Detector, UV 254 nm.
LC-MS: (ES, m/z): 450 [M+H]+
H-NMR: (CD3OD, 400MHz, ppm): 8.55 (s, IH), 5.87 (d, J=16Hz, IH), 5.42 (t, IH), 5.24 (d, J=16Hz, IH), 3.78 (s, 3H), 3.47-3.53 (m, 2H), 2.44 (s, 3H), 1.88 (s, 3H), 1.46-1.72 (m, 3H), 1.30-1.37 (m, 2H), 1.21 (m, 3H), 1.15 (s, IH), 0.92 (s, 3H), 0.84 (d, J=7.2Hz, 3H), 0.72 (d, J=6.8Hz, 3H).
Example 10
[0135] Synthesis of 4-chloro-3-methoxy-2-[(2E,4E)-5-[( 1 R,2R,3E,6R)-3-(methoxyimino)- 1 ,2,6- trimethylcyclohexyl]-3-methylpenta-2,4-dien-l-yl]-6-[(lE)-(methoxyimino)methyl]-5-methylphenol (Compound 8)
Figure imgf000046_0001
[0136] The titled compound was prepared from 4-O-methyl-ascochlorine by the similar manner as described in Example 4 except that O-methyl hydroxyamine hydrochloride was used instead of hydroxyamine hydrochloride.
LC-MS: (ES, m/z): 477 [M+H]+
H-NMR: (DMSO, 400MHz, ppm): δ 10.80 (s, IH), 8.64 (s, IH), 5.89(d, J = 16.1 Hz, IH), 5.46- 5.37(m, 2H), 3.96 (s, 3H), 3.75 (s, 3H), 3.71 (s, 3H), 3.45 (d, J=7.2Hz, 2H), 3.15-3.11 (m, IH), 2.41 (s, 3H), 2.223 (q, J=6.8Hz, IH), 1.84 (s, 3H), 1.66 (m, 3H), 1.20 (td, J = 14.4, 13.1, 6.3 Hz, IH), 0.80 (d, J = 6.8 Hz, 3H), 0.65 (m, 6H).
Example 11
[0137] Synthesis of 4-chloro-3-(fluoromethoxy)-2-[(2E,4E)-5-[(lR,2R,3E,6R)-3-(methoxyimino)- l ,2,6-trimethylcyclohexyl]-3-meth^
methylphenol (Compound 9)
Figure imgf000047_0001
[0138] The titled compound was prepared from 4-O-fluoromethyl-ascochlorine by the similar manner as described in Example 8 except that O-methyl hydroxyamine hydrochloride was used instead of hydroxyamine hydrochloride.
LC-MS (ES, m/z): [M+H]+: 495.
Ή NMR (300 MHz, Methanol-d4) δ 8.60 (s, 1H), 5.89 (d, J = 16.2 Hz, 1H), 5.64 (d, J = 54.3 Hz, 2H), 5.42 (t, J = 7.2 Hz, 1H), 5.36 (d, J = 16.2 Hz, 1H), 3.97 (s, 3H), 3.73 (s, 3H), 3.58 (d, J = 7.2 Hz, 2H), 3.23-3.26 (m, 1H), 2.43 (s, 3H), 2.17 (q, J = 6.9 Hz, 1H), 1.87 (s, 3H), 1.60-1.71 (m, 3H), 1.30- 1.33 (m, 1H), 0.86 (d, J = 6.9 Hz, 3H), 0.73 (d, J = 6.6 Hz, 3H), 0.67 (s, 3H).
Example 12
[0139] Synthesis of 4-chloro-2-[(2E,4E)-5-[( 1 R,2R,3E,6R)-3-(hydroxyimino)-l ,2,6- trimethylcyclohexyl]-3-methylpenta-2,4-dien-l-yl]-3-methoxy-6-[(lE)-(methoxyimino)methyl]-5- methylphenol (Compound 10)
Figure imgf000047_0002
[0140] Step 1
A mixture of 3-chloro-6-hydroxy-4-methoxy-2-methyl-5-[(2E,4E)-3-methyl-5-[(l R,2R,6R)- l,2,6-trimethyl-3-oxocyclohexyl]penta-2,4-dien-l-yl]benzaldehyde (4-O-methyl-ascochlorine, 350 mg, 0.84 mmol) and NH20Me.HCl (69.3 mg, 0.83 mmol) in pyridine (4 mL) was placed in a 50-mL round- bottom flask purged and maintained with an atmosphere of argon. The mixture was stirred overnight at room temperature and concentrated under vacuum. The residue (400 mg) was purified by Prep-HPLC with the following condition to give (2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (methoxyimino)methyl]-4-methylphenyl]-3-methylpenta- 1 ,3-dien- 1 -yl]-2,3,4-trimethylcyclohexan- 1 - one (200.9 mg, 54%) as an off-white solid.
Prep-HPLC condition (2#-AnalyseHPLC-SHIMADZU(HPLC-10)): Column, XBridge Prep CI 8 OBD Column, 5 urn, 19* 150mm; mobile phase, Water with 10mmolNH4HCO3 and CH3CN (hold 80.0% CH3CN in 12 min); Detector, U V 254nm.
LC-MS (ES, m/z): 448 [M+H]+
H-NMR (DMSO, 300MHz, ppm): δ 10.80 (s, 1H), 8.64 (s, 1 H), 5.88 (d, J= 16.0 Hz, 1H), 5.37-5.51 (m, 2H), 3.96 (s, 3H), 3.75 (s, 3H), 3.46 (d, J=6.9Hz, 2H) , 2.56 (m, 2H), 2.41 (s, 3H), 2.15 (m, 1 H), 1.80-2.06 (m, 4H), 1.41-1.59 (m, 1H), 0.70 (dd, J= 15.0, 6.7 Hz, 6H), 0.60 (s, 3H).
[0141 ] Step 2
A mixture of (2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (methoxyimino)methyl]-4-methylphenyl]-3-methylpenta- 1 ,3-dien- 1 -yl]-2,3,4-trimethylcyclohexan- 1 - one (500 mg, 1.12 mmol) and NH20H.HC1 (84.9 mg, 1.23 mmol, 1.10 equiv) in pyridine (5 mL) was placed in a 50-mL round-bottom flask purged and maintained with an atmosphere of argon. The mixture was stirred overnight at room temperature and then concentrated under vacuum. The residue was purified by Prep-HPLC with the following condition to give 4-chloro-2-[(2E,4E)-5-[(lR,2R,3E,6R)- 3-(hydroxyimino)-l,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien-l-yl]-3-methoxy-6-[(lE)- (methoxyimino)methyl]-5-methylphenol (246.5 mg, 48%, Compound 10) as a white solid.
Prep-HPLC condition: Column: X Bridge CI 8, 19*250 mm, 5 urn; Mobile Phase
A:Water/10mm NH4HCO3, Mobile Phase B: CH3CN; Flow rate: 30 mL/min; Gradient: 88%B to 88%B in 8 min; 254nm; Detector.
LC-MS (ES, m/z): 463 [M+H]+
H-NMR (DMSO, 300MHz, ppm):5 10.80 (s, 1H), 10.35 (s, 1H), 8.65 (s, 1H), 5.90 (d, J = 16.1 Hz, 1H), 5.42 (m, 2H), 3.96 (s, 3H), 3.75 (s, 3H), 3.46 (d, J = 7.1 Hz, 2H), 3.23 (m, 1 H), 2.41 (s, 3H), 2.23 (t, J = 6.8 Hz, 1H), 1.86 (s, 3H), 1.62 (d, J = 11.9 Hz, 3H), 1.18 (d, J = 10.5 Hz, 1H), 0.80 (d, J = 6.8 Hz, 3H), 0.68 (d, J = 6.6 Hz, 3H), 0.61 (s, 3H).
Example 13 [0142] Synthesis of 4-chloro-3-(fluoromethoxy)-2-[(2E,4E)-5-[(lR,2R,3E,6R)-3-(hydroxyimino)- l,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien-l -yl]-6-[(lE)-(methoxyimino)methyl]-5- methylphenol (Compound 11).
Figure imgf000049_0001
[0143] The titled compound was prepared from 4-O-fluoromethyl-ascochlorine by the similar manner as described in Example 12.
LC-MS (ES, m/z): [M+H]+: 481.
¾ NMR (300 MHz, DMSO-cfe) δ 10.36 (s, 1H), 8.66 (s, 1H), 5.88 (d, J= 16.2 Hz, 1H), 5.72 (d, J= 54.3 Hz, 2H) 5.42 (t, J= 7.5 Hz, 1H), 5.37 (d, J= 16.2 Hz, 1H), 3.95 (s, 3H) 3.50 (m, 2H), 3.23 (m, 2H), 2.43 (s, 3H), 2.21 (q, J- 6.6 Hz, 1H), 1.84 (s, 3H), 1.59 (m, 3H), 1.25 (m, 1H), 0.80 (d, J 6.6 Hz, 3H), 0.68 (d, J= 6.9 Hz, 3H), 0.61 (s, 3H).
Example 14
[0144] Synthesis of 4-chloro-2-[(2E,4E)-5-[( 1 R,2R,3E,6R)-3-[[2-(dimethylamino)ethoxy]imino]- l,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien-l -yl]-3-methoxy-6-[(l E)-(methoxyimino)methyl]-5- methylphenol (Compound 15)
Figure imgf000049_0002
[0145] The titled compound was prepared by the method similar to that described in Example 12 except that 0-(2-(dimethylamino)ethyl)hydroxyamine hydrochloride was used instead of hydroxyamine hydrochloride.
LC-MS (ES, m/z): 534 [M+H]+
H-NMR (DMSO, 400MHz, ppm): 510.79 (s, 1H), 8.63 (s, 1 H), 5.89 (d, J = 16.1 Hz, 1H), 5.33 - 5.48 (m, 2H), 3.98 (m, 5H), 3.74 (s, 3H), 3.45 (d, J = 7.2 Hz, 2H), 3.13 (m, 1H), 2.46 (m, 2H), 2.39 (s,3H), 2.23 (q, J = 6.8 Hz, 1 H), 2.14 (s, 6H), 1.85 (d, J = 1.3 Hz, 3H), 1.57 - 1.74 (m, 3H), 1.19 (m, 0.78 (d, J = 6.7 Hz, 3H), 0.66 (d, J = 6.5 Hz, 3H), 0.61 (s, 3H).
Example 15
[0146] Synthesis of 4-chloro-3-methoxy-6-[(l E)-(methoxyimino)methyl]-5-methyl-2-[(2E,4E)-3- methyl-5-[(lR,2R,3E,6R)-l,2,6-trimethyl-3-[[2-(mo holin-4-yl)ethoxy]imino]cyclohexyl]penta-2,4- dien-l-yl]phenol (Compound 16).
Figure imgf000050_0001
[0147] The titled compound was prepared by the method similar to that described in Example 12 except that 0-(2-(morpholin-4-yl)ethyl)hydroxyamine hydrochloride was used instead of hydroxyamine hydrochloride.
LC-MS (ES, m/z): 576 [M+H]+
H-NMR (CD3CN, 300MHz, ppm): δ 8.55 (s, 1H), 5.93 (d, J= 16.1 Hz, 1H), 5.36-5.50 (m, 2H), 4.07 (t, J= 5.7 Hz, 2H), 3.94 (s, 3H), 3.75 (s, 3H), 3.51 (m, 6H), 3.14-3.28 (m, 1 H), 2.55 (m, 2H), 2.46 (s, 7H), 2.20 (t, 7= 6.8 Hz, 1H), 1.89 (d, J= 1.1 Hz, 3H), 1.58-1.72 (m, 3H), 1.25 (m, 1 H), 0.81 (d, y= 6.8 Hz, 3H), 0.67 (m, 6H).
Example 16
[0148] Synthesis of 4-chloro-3-methoxy-6-[(lE)-(methoxyimino)methyl]-5-methyl-2-[(2E,4E)-3- methyl-5-[( 1 R,2R,3E,6R)- 1 ,2,6-trimethyl-3-[[2-(4-methylpiperazin- 1 - yl)ethoxy]imino]cyclohexyl]penta-2,4-dien-l -yl]phenol (Compound 17).
Figure imgf000050_0002
[0149] The titled compound was prepared by the method similar to that described in Example 12 except that 0-(2-(N-methylpipearzin-4-yl)ethyl)hydroxyamine hydrochloride was used instead of hydroxyamine hydrochloride.
LC-MS (ES, m/z): 589 [M+H]+
H-NMR (CDCb, 400MHz, ppm): δ 10.80 (s, 1H), 8.50 (s, 1 H), 5.92 (t, J=16.0Hz, 1H), 5.51 (m, l H), 5.33 (m, 1H), 4.19 (m, 2H), 3.99 (s, 3H), 3.83 (s, 3H), 3.56 (d, J=7.2Hz, 2H), 3.28 (m, 1H), 2.42-2.70 (m, 13H), 2.32 (s, 3H), 2.14 (m, 1H), 1.93 (s, 3H), 1.63 (m, 3H), 1.29 (m, 1H), 0.88 (d, J=6.8Hz, 3H), 0.71 (m, 6H).
Example 17
[0150] Synthesis of ethyl 2-([[( 1 E,2R,3R,4R)-3-[( 1 E,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (methoxyimino)methyl]-4-methylphenyl]-3-methylpenta-l ,3-dien-l-yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetate (Compound 18).
Figure imgf000051_0001
[0151] The titled compound was prepared by the method similar to that described in Example 12 except that ethyl 2-(aminooxy)acetate was used instead of hydroxyamine hydrochloride.
LC-MS (ES, m/z): [M+H]+: 549
Ή NMR (300 MHz, DMSO-c/6): δ 10.74 (s, 1H), 8.60 (s, 1H), 5.86 (d, J= 16.2 Hz, 1H), 5.41 (t, J= 7.0 Hz, 1H), 5.35 (d, J= 16.2 Hz, 1H), 4.51 (s, 2H), 4.06 (q, J= 7.1 Hz, 2H), 3.71 (s, 3H), 3.42 (d, J= 7.2 Hz, 2H), 3.28 (s, 1 H), 3.18 (dd, J= 12.9, 4.1 Hz, 1 H), 2.37 (s, 3H), 2.24 (q, J= 6.8 Hz, 1H), 1.81 (d, J= 1.3 Hz, 3H), 1.79 - 1.56 (m, 3H), 1.31 - 1.17 (m, 1H), 1.14 (t, 7= 7.1 Hz, 3H), 0.71 (d, J= 6.7 Hz, 3H), 0.64 (d, J= 6.5 Hz, 3H), 0.58 (s, 3H).
Example 18
[0152] Synthesis of 4-chloro-2-[(2E,4E)-5-[(l R,2R,3S,6R)-3-hydroxy-l ,2,3,6-tetramethylcyclohexyl]- 3-methylpenta-2,4-dien-l-yl]-3-methoxy-6-[(lE)-(methoxyimino)methyl]-5-methylphenol (Compound 14-1).
Figure imgf000052_0001
[0153] A mixture of 3-chloro-6-hydroxy-5-[(2E,4E)-5-[(lR,2R,3S,6R)-3-hydroxy-l,2,3,6- tetramethylcyclohexyl]-3-methylpenta-2,4-dien-l-yl]-4-methoxy-2-methylbenzaldehyde [Compound 1, Example 2] (350 mg, 0.80 mmol) and methoxylamine hydrochloride (85 mg, 1.02 mmol) in pyridine (5 mL) was placed in a 50-mL round-bottom flask. The mixture was stirred for 12 h at 10 to 15°C and concentrated under vacuum. The residue was purified by Prep-HPLC with the following condition to give the titled compound (209.3 mg, 55%) as a light yellow solid.
Prep-HPLC condition (2#-AnalyseHPLC-SHIMADZU(HPLC-10)): Column, XBridge BEH CI 8 OBD Prep Column, 5 μιη, 19 mm 250 mm; mobile phase, Water with lOmmol NH4HCO3 and CH3CN (hold 90.0% CH3CN in 9 min); Detector, uv 254&220nm.
LC-MS (ES, m/z) [M-H] 462
H-NMR (DMSO, 400MHz ppm): δ 10.79 (s,lH), 8.64 (s,lH),5.82 (m,lH), 5.39 (m.lH), 5.22 (m,lH), 3.96 (s.3H), 3.74 (m,4H), 3.32-3.45 (m,2H), 2.50 (s,3H), 1.81 (m,3H), 1.47-1.59 (m,2H), 1.21- 1.38 (m,3H), 1.13 (m, lH), 1.04 (s,3H), 0.85 (m,3H), 0.74 (m,3H), 0.64 (m,3H).
Example 19
[0154] Synthesis of 2-([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (methoxyimino)methyl]-4-methylphenyl]-3-methylpenta-l,3-dien-l-yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid (Compound 19).
Figure imgf000052_0002
A solution of ethyl 2-([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (methoxyimino)methyl]-4-methylphenyl]-3-methylpenta-l ,3-dien-l -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetate [Compound 18, Example 17] (350 mg, 0.64 mmol) was placed in a 25-mL round-bottom flask, followed by addition of a solution of lithium hydroxide (76 mg, 3.17 mmol) in water/THF (0.5/1 mL). The mixture was stirred overnight at room temperature. The pH value of the mixture was adjusted to 4 with aqueous hydrochloride acid (IN). The reaction mixture was extracted with dichloromethane (2 mL x2). The organic layer was combined, and then concentrated under vacuum. The residue was purified by Prep-HPLC with the following condition to give 2-([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (methoxyimino)methyl]-4-methylphenyl]-3-methylpenta-l,3-dien-l-yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid (300 mg, 90%, Compound 19) as a white solid.
Prep-HPLC (2#-AnalyseHPLC-SHIMADZU(HPLC-10)): Column, XBridge Shield RP18 OBD Column, 5um,19*150mm; mobile phase, WATER WITH 0.05%TFA and CH3CN (hold 72.0% CH3CN in 11 min); Detector, UV 254nm.
LC-MS: (ES, m/z): [M+H]+: 521.
lH NMR (300 MHz, DMSO-</6) δ 10.76 (s, 1H), 8.60 (s, 1H), 5.86 (d, J= 16.2 Hz, 1H), 5.41 (t, J= 6.9 Hz, 1H), 5.35 (d, J= 16.2 Hz, 1H), 4.42 (s, 2H), 3.92 (s, 3H), 3.42 (d, J= 7.1 Hz, 2H), 3.13 - 3.25 (m, 1H), 2.37 (s, 3H), 2.23 (q, J= 6.8 Hz, 1H), 1.82 (d, J= 1.4 Hz, 3H), 1.56 - 1.76 (m, 3H), 1.22 (s, 1H), 0.71 (d, J= 6.8 Hz, 3H), 0.64 (d, J= 6.5 Hz, 3H), 0.59 (s, 3H).
Example 20
[0155] Synthesis of 3-chloro-4-(fluoromethoxy)-6-hydroxy-2-methyl-5-[(2E,4E)-3-methyl-5- [(1 R,2R,3E,6R)- 1 ,2,6-trimethyl-3-[[2-(mo holin-4-yl)ethoxy]imino]cyclohexyl]penta-2,4-dien- 1 - yljbenzaldehyde (Compound 12)
Figure imgf000053_0001
[0156] The titled compound was prepared by the similar method as described in Example 6 except that 0-2-(morpholin-4-yl)ethyl hydroxyamine hydrochloride was used instead of hydroxyamine hydrochloride.
LC-MS (ES, m/z): [M+H]+: 565 Ή NMR (300 MHz, Chloroform-c δ 12.55 (s, 1H), 10.29 (s, 1H), 5.91 (d, J= 16.0 Hz, 1H), 5.70 (d, J= 54.3 Hz, 1H), 5.46 (t, J= 7.4 Hz, 1H), 5.34 (d, J= 16.0 Hz, 1H), 4.19 (t, J= 5.8 Hz, 2H), 3.72 (m, 4H), 3.61 (d, J= 7.3 Hz, 2H), 3.21 - 3.36 (m, 1H), 2.67 (d, J= 9.0 Hz, 5H), 2.55 (m, 4H), 2.15 (q, J= 6.7 Hz, 1H), 1.52 - 1.74 (m, 3H), 1.30 (m, 1 H), 0.89 (d, J= 6.7 Hz, 3H), 0.73 (d, J= 6.4 Hz, 3H), 0.69 (s, 3H).
Example 21
[0157] Synthesis of 4-chloro-2-[(2E,4E)-5-[(l R,2R,6R)-3-(ethylamino)-l,2,6-trimethylcyclohexyl]-3- methylpenta-2,4-dien-l-yl]-3-methoxy-6-[(lE)-(methoxyimino)methyl]-5-methylphenol (Compound 21).
Figure imgf000054_0001
[0158] Step 1
A mixture of placed (2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (methoxyimino)methyl]-4-methylphenyl]-3-methylpenta- 1 ,3-dien- 1 -yl]-2,3,4-trimethylcyclohexan- 1 - one [Example 12, step 1 ] (3 g, 6.70 mmol), Ti(i-PrO)4 (15.2 g, 53.52 mmol), ethylamine hydrochloride (2.2 g, 26.83 mmol) and MS 4A (lg) in THF (30 mL) was placed in a 250-mL round-bottom flask. The mixture was stirred overnight at 68°C in an oil bath. The resulting mixture was diluted with 200 mL of ethyl acetate followed by addition of 10 mL of saturated ammonium chloride. The mixture was filtered, and the filtrate was dried over anhydrous magnesium sulfate and concentrated under vacuum to give 4- chloro-2-[(2E,4E)-5-[(lR,2R,3E,6R)-3-(ethylimino)-l ,2,6-trimethylcyclohexyl]-3-methylpenta-2,4- dien-l-yl]-3-methoxy-6-[(lE)-(methoxyimino)methyl]-5-methylphenol (1.2 g, crude) as a brown solid.
[0159] Step 2
A solution of 4-chloro-2-[(2E,4E)-5-[(lR,2R,3E,6R)-3-(ethylimino)-l ,2,6- trimethylcyclohexyl]-3-methylpenta-2,4-dien-l-yl]-3-methoxy-6-[(lE)-(methoxyimino)methyl]-5- methylphenol (1.2 g, 2.53 mmol) and sodium triacetoxyborohydride (1.07 g, 5.05 mmol, 2.00 equiv) in dichloromethane (20 mL) was placed in a 100-mL round-bottom flask. The mixture was stirred for 5 h at room temperature. The mixture was filtered, and the filtrate was concentrated under vacuum. The residue was purified by Flash-Prep-HPLC with the following condition to give a crude product (350 mg), which was purified by Chiral-Prep-HPLC with the following condition to give 4-chloro-2-[(2E,4E)-5- [( 1 R,2R,6R)-3-(ethylamino)- 1 ,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien- 1 -yl]-3-methoxy-6- [(lE)-(methoxyimino)methyl]-5-methylphenol (211.4 mg, 18%, Compound 21) as an orange solid.
Flash-Prep-HPLC condition (CombiFlash-1): Column, CI 8 silica gel; mobile phase, water/acetonitrile=l :0 increasing to water/acetonitrile = 0: 1 within 40 min; Detector, UV 254 nm.
Chiral-Prep-HPLC condition: Column: XBridge Shield RP18 OBD Column, 5um, 19* 150mm; Mobile Phase A: Water with lOmmol NH4HCO3, Mobile Phase B: ACN; Flow rate: 20 mL/min;
Gradient: 75% B to 95% B in 10 min; Detector 254 nm.
LC-MS (ES, m/z): 477 [M+H]+
H-NMR (DMSO, 300MHz, ppm): δ 10.71-10.87 (br, IH), 8.644 (s, IH), 5.82 (t, J=16.2Hz, IH), 5.39 (m, IH), 5.25 (m, IH), 3.96 (s, 3H), 3.75 (s, 3H), 3.44 (d, J=6.9Hz, 2H), 2.57-2.66 (m, 2H), 2.51 (s, 3H), 2.28-2.36 (m, IH), 1.74-1.82 (m, 4H), 1.12-1.50 (m, 6H), 0.98 (m, 3H), 0.86 (m, 3H), 0.77 (m, 3H), 0.63 (m, 3H).
Example 22
[0160] Synthesis of 4-chloro-2-[(2E,4E)-5-[(lR,2R,6R)-3-(cyclopropylamino)-l ,2,6- trimethylcyclohexyl]-3-methylpenta-2,4-dien-l -yl]-3-methoxy-6-[(lE)-(methoxyimino)methyl]-5- methylphenol (Compound 23)
Figure imgf000055_0001
[0161] The titled compound was prepared by the similar method described in Example 21 except that cyclopropyl amine was used instead of ethylamine.
LC-MS (ES, m/z): 489 [M+H]+
H-NMR (DMSO, 300MHz, ppm): δ 10.79 (s, IH), 8.64 (s, IH), 5.80 (t, J=16.2Hz,lH), 5.37 (m, IH), 5.24 (m, IH), 3.96 (s, 3H), 3.74 (s, 3H), 3.44 (d, J=6.3Hz, 2H), 2.74 (m, IH), 2.41 (s, 3H), 1.96 (m, 2H), 1.82 (s, 3H), 1.41 -1.57 (m, 3H), 1.15-1.40 (m, 3H), 0.80 (s, 3H), 0.73 (m, 3H), 0.64 (m, 3H), 0.39 (m, 1H), 0.25 (m, 2H), 0.15 (m, 1 H).
Example 23
[0162] Synthesis of 2-[(2E,4E)-5-[( 1 R,2R,6R)-3 -amino- 1 ,2,6-trimethylcyclohexyl]-3-methylpenta-2,4- oxyimino)methyl]-5-methylphenol (Compound 24)
Figure imgf000056_0001
[0163] A mixture of (2R,3R,4R)-3-[(l E,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (methoxyimino)methyl]-4-methylphenyl]-3-methylpenta-l ,3-dien-l -yl]-2,3,4-trimethylcyclohexan-l - one (3 g, 6.70 mmol) [Example 12, Step 1], NaBH3CN (2.75 g, 43.76 mmol) and NH4OAc (20 g, 40.00 equiv) in ethanol (100 mL) was placed in a 250-mL round-bottom flask purged and maintained with an atmosphere of argon. The resulting mixture was stirred overnight at 70°C in an oil bath. The resulting mixture was concentrated under vacuum. The residue was diluted with H20 (80 mL). The mixture was extracted with ethyl acetate (200 mL x3) and the organic layer was combined, then concentrated under vacuum. The residue was purified by Flash-Prep-HPLC with the following condition to give the titled compound ( 1.5 g, 50%, Compound 24) as a yellow solid.
Flash-Prep-HPLC condition (IntelFlash-1): Column, C I 8 silica gel; mobile phase, CH3CN: H2O=0:95 increasing to CH3CN: H20 = 95:5 within 55 min; Detector, UV 254 nm.
LC-MS: (ES, m/z): [M+H]+ 449
H-NMR: (CDCl3,400MHz ppm): 510.77 (s, 1 H), 8.50 (s, 1H), 5.93 (d, J=16.4Hz ,1 H), 5.52 (m, l H), 5.19 (m, lH), 3.99 (s, 3H), 3.82 (s, 3H), 3.54 (m, 2H), 2.42 (s, 3H), 2.04-2.23 (m, 1 H), 1.90 (m, 3H), 1.25-1.85 (m, 6H), 0.91-1.04 (m, 5H), 0.70-0.78 (m, 4H).
Example 24
[0164] Synthesis of 4-chloro-3-methoxy-6-[(l E)-(methoxyimino)methyl]-5-methyl-2-[(2E,4E)-3- methyl-5-[(lR,2R,6R)-l,2,6-trimethyl-3-[(oxetan-3-yl)amino]cyclohexyl]penta-2,4-dien-l -yl]phenol (Compound 25)
Figure imgf000057_0001
[0165] Into a 50-mL sealed tube, a solution of 2-[(2E,4E)-5-[(lR,2R,6R)-3-amino-l,2,6- trimethy lcyclohexy 1] -3 -methylpenta-2,4-dien- 1 -y 1] -4-chloro-3 -methoxy-6- [( 1 E)- (methoxyimino)methyl]-5-methylphenol (Compound 24; 1 g, 2.23 mmol), sodium
triacetoxyborohydride (STAB, 709.8 mg, 3.35 mmol) and oxetan-3-one (353.6 mg, 4.91 mmol) in DCM (12mL) were placed. The mixture was stirred for 18 h at room temperature and then concentrated under vacuum. The residue was purified by Prep-HPLC with the following condition to give the titled compound (239.2 mg, 21%, Compound 25) as an off-white solid.
Prep-HPLC condition (2#-AnalyseHPLC-SHIMADZU(HPLC-10)): Column, XBridge Shield RP18 OBD Column, 5um, 19* 150mm; mobile phase, Water with 10mmolNH4HCO3 and CH3CN (hold 82.0% ACN in 10 min); Detector, UV 254nm.
LC-MS: (ES, m/z): [M+H]+ 505
H-NMR: (DMSO,400MHz ppm): δ 10.80 (s, 1H), 8.66 (s, 1H), 5.84 (d, J=16.4Hz ,1H), 5.41 (m,lH), 5.25 (d, J=15.6Hz, lH), 4.60 (m,2H), 4.27-4.36(m, 2H), 4.38 (s, 3H), 3.83 (m, 1H), 3.76 (s, 3H), 3.46 (d, J=7.2Hz, 2H), 2.42 (s, 3H), 1.84 (s, 3H), 1.45-1.58 (m, 3H), 1.35 (m, 2H), 1.21 (m, 1 H), 0.89 (s, 3H), 0.81 (d, J=7.2Hz, 3H), 0.67 (m, 3H).
Example 25
[0166] Synthesis of 3-chloro-5-[(2E,4E)-5-[(lR,2R,6R)-3-(ethylamino)-l,2,6-trimethylcyclohexyl]-3- methylpenta-2,4-dien-l -yl]-6-hydroxy-4-methoxy-2-methylbenzaldehyde (Compound 20).
Figure imgf000057_0002
[0167] A solution of 4-chloro-2-[(2E,4E)-5-[( 1 R,2R,6R)-3-(ethylamino)- 1 ,2,6-trimethylcyclohexyl]-3- methylpenta-2,4-dien-l -yl]-3-methoxy-6-[( 1 E)-(methoxyimino)methyl]-5-methylphenol [Compound 21 , Example 21] (500 mg, 1.05 mmol), formaldehyde (425 mg, 5.25 mmol, 5.00 equiv, 37%) and hydrochloride acid (500 mg, 5.3 mmol, 5.00 equiv, 38%) in THF/water (20 mL/2 mL) was placed in a 30-mL sealed tube. The mixture was stirred overnight at 100°C and then concentrated under vacuum. The residue was purified by Prep-HPLC with the following condition to give the titled compound (55.4 mg, 9.4%, Compound 20) as an off-white solid.
Prep-HPLC condition (2#-AnalyseHPLC-SHIMADZU(HPLC-10)): Column, Gemini-NX 5u C18 11 OA, AXIA Packed, 150 x 21.2 mm; mobile phase, Column: Mobile Phase A: Water with lOmmol CF3COOH, Mobile Phase B: CH3CN; Flow rate: 30 mL/min; Gradient: 60% B to 95% B in 10 min; 254 nm; Detector, uv 254nm.
LC-MS (ES, m/z): 448 [M+H]+
H-NMR (DMSO, 300 MHz, ppm) 12.62 (br, I H), 10.27 (br, IH), 7.90 (br, IH), 7.43 (br, IH), 5.94 (d, J=16.2Hz, IH), 5.43 (t, J=7.5Hz, IH), 5.34 (d, J=15.9Hz, IH), 3.82 (s, 3H), 3.47 (d, J=7.5Hz, 2H), 3.28 (m, IH), 2.96 (m, 2H), 2.62 (s, 3H), 1.85 (m, 5H), 1.70 (m, IH), 1.50 (s, 3H), 1.20 (m, 3H), 0.88 (m, 3H), 0.83 (m, 3H), 0.74 (m, 3H).
Example 26
[0168] Synthesis of 3-chloro-5-[(2E,4E)-5-[(lR,2R,6R)-3-(cyclopropylamino)-l , 2,6- trimethylcyclohexyl]-3-methylpenta-2,4-dien-l-yl]-6-hydroxy-4-methoxy-2-methylbenzaldehyde
(Compound 22)
Figure imgf000058_0001
[0169] The titled compound was prepared from Compound 23 (Example 22) by the similar method as described in Example 25.
LC-MS (ES, m/z): 460 [M+H]+
H-NMR (DMSO, 300 MHz, ppm): 12.60 (br, IH), 10.27 (s, I H), 5.80 (d, J=16.2Hz, IH), 5.36 (t, J=7.2Hz, IH), 5.25 (d, J=15.9Hz, IH), 3.81 (s, 3H), 3.42 (d, J=6.9Hz, 2H), 2.75 (m, IH), 2.62 (s, 3H), 1.96 (m, 2H), 1.82 (s, 3H), 1.20-1.61 (m, 6H), 0.80 (m, 3H), 0.71 (m, 3H), 0.63 (m, 3H), 0.35-0.47 (m, 1H), 0.24-0.31 (m, 2H), 0.06-0.18 (m, 1H).
Example 27
[0170] Synthesis of 3-chloro-5-[(2E,4E)-5-[( 1 R,2R,6R)-3-(cyclopropylamino)- 1 ,2,6- trimethylcyclohexyI]-3-methylpenta-2,4-dien-l-yl]-4-(fluoromethoxy)-6-hydroxy-2- methylbenzaldeh de (Compound 13)
Figure imgf000059_0001
[0171] Step 1
Into a 50-mL round-bottom flask, was placed a solution of (2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-5- (diethoxymethyl)-2-(fluoromethoxy)-6-hydroxy-4-methylphenyl]-3-methylpenta- 1 ,3-dien- 1 -yl]-2,3,4- trimethylcyclohexan-l-one [Example 2] (900 mg, 1.76 mmol), MS 4A (200 mg) and cyclopropanamine (2 g, 35.03 mmol, 20.00 equiv) in ethanol (10 mL). The resulting solution was stirred overnight at 30°C. The resulting mixture was concentrated under vacuum to give 4-chloro-2-[(2E,4E)-5-[(lR,2R,3E,6R)-3- (cyclopropylimino)-l,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien-l-yl]-6-(diethoxymethyl)-3- (fluoromethoxy)-5-methylphenol (0.9 g, 93%) as a solid.
[0172] Step 2
A solution of 4-chloro-2-[(2E,4E)-5-[(lR,2R,3E,6R)-3-(cyclopropylimino)-l ,2,6- trimethylcyclohexyl]-3-methylpenta-2,4-dien-l-yl]-6-(diethoxymethyl)-3-(fluoromethoxy)-5- methylphenol (1.1 g, 2.00 mmol) and sodium borohydride acetate (464 mg, 2.19 mmol) in ethanol (20 mL) was placed in a 100-mL round-bottom flask. The mixture was stirred for 3 h at room temperature and concentrated under vacuum. The residue was purified by silica gel column chromatography with ethyl acetate/petroleum ether (1 :10) to give 4-chloro-2-[(2E,4E)-5-[(lR,2R,6R)-3-(cyclopropylamino)- l ,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien-l -yl]-6-(diethoxymethyl)-3-(fluoromethoxy)-5- methylphenol (460 mg, 42%) as a solid.
[0173] Step 3 A solution of 4-chloro-2-[(2E,4E)-5-[(l R,2R,6R)-3-(cyclopropylamino)-l ,2,6- trimethylcyclohexyl]-3-methylpenta-2,4-dien-l -yl]-6-(diethoxymethyl)-3-(fluorometh^
methylphenoi (460 mg, 0.83 mmol) in THF/water/acetic acid (10/20/10 mL) was placed in a 100-mL round-bottom flask. The mixture was stirred for 3 h at room temperature and concentrated under vacuum. The residue was dissolved in water (10 mL). The mixture was extracted with ethyl acetate (20 mL) and the organic layer was combined, then concentrated under vacuum. The residue was purified by Prep-HPLC with the following condition of 3-chloro-5-[(2E,4E)-5-[(l R,2R,6R)-3- (cyclopropylamino)-l ,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien-l -yl]-4-(fluoromethoxy)-6- hydroxy-2-methylbenzaldehyde (206.7 mg, 52%, Compound 13) as a light yellow solid.
Prep-HPLC condition (2#-AnaIyseHPLC-SHIMADZU(HPLC- 10)): Column, Gemini-NX C 18 AXAI Packed, 21.2 x 150mm 5um 1 l nm; mobile phase, Water with l Ommol NH4HCO3 and CH3CN (hold 95.0% CH3CN in 8 min); Detector, uv 254nm.
LC-MS (ES, m/z): [M+H]+: 478.
'H NMR (300 MHz, DMSO-C&) δ 10.26 (s, 1 H), 5.79 (d, J= 154.3 Hz, 2H), 5.78 (d, J= 16.2 Hz, 1H) 5.34 (t, /= 7.2 Hz, 1 H), 5.25 (d, J= 16.2 Hz, 1H), 3.48 (d, J= 6.9 Hz, 2H), 2.76 (m, 1 H), 2.64 (s, 3H), 1.95-2.08 (m, 2H), 1.80(s, 3H), 1.20- 1.48 (m, 5H), 0.79 (s, 3H), 0.69 (d, J= 7.2 Hz, 3H), 0.59 (d, J= 6.3 Hz, 3H), 035 (m, 1 H), 0.24 (m, 1H),0.1 1 (m, 1 H).
Example 28
[0174] Synthesis of 2-([[( 1 E,2R,3R,4R)-3-[( 1 E,3E)-5-[3-chloro-2-(fluoromethoxy)-5-formyl-6- hydroxy-4-methyIphenyl]-3-methylpenta- 1 ,3-dien- 1 -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid (Compound 26)
Figure imgf000060_0001
[0175] Step 1
A solution of (2R,3R,4R)-3-[(l E,3E)-5-[3-chloro-5-(diethoxymethyl)-2-(fluoromethoxy)-6- hydroxy-4-methylphenyl]-3-methylpenta-l ,3-dien-l -yl]-2,3,4-trimethylcyclohexan-l -one [Example 2] (1.5 g, 2.94 mmol), CS2CO3 (479 mg, 1.47 mmol) and ethyl 2-(aminooxy)acetate oxalic acid salt (605 mg, 2.89 mmol) in ethanol (10 mL) was placed in a 100-mL round-bottom flask. The mixture was stirred for 2 h at room temperature. The reaction mixture was diluted with water (10 mL), and extracted with ethyl acetate (10 mL x2). The organic layer was combined, and then concentrated under vacuum. The residue was purified by Flash-Prep-HPLC with the following condition to give ethyl 2- ([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-5-(diethoxymethyl)-2-(fluoromethoxy)-6-hydroxy-4- methylphenyl]-3-methylpenta-l,3-dien-l-yl]-2,3,4-trimethylcyclohexylidene]amino]oxy)acetate (1.0 g, 56%) as a light yellow solid.
Flash-Prep-HPLC condition (IntelFlash-1): Column, CI 8 silica gel; mobile phase,
CH3CN/H2O = 60:40 increasing to CH3CN/H2O = 95:5 within 40 min; Detector, UV 254 nm.
LC-MS (ES, m/z): 612 [M+H]+
[0176] Step 2
A mixture of ethyl 2-([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-5-(diethoxymethyl)-2- (fluoromethoxy)-6-hydroxy-4-methylphenyl]-3-methylpenta-l,3-dien-l -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetate (1 g, 1.63 mmol), lithium hydroxide (117 mg, 3.00 equiv) in THF H2O (1 : 1 , 30 mL) was placed in a 100-mL round-bottom flask. The mixture was stirred for 2 h at room temperature. Then (15 mL) was added to the solution. The resulting solution was stirred for an additional 6 h at room temperature. The resulting mixture was concentrated under vacuum and diluted with 20 mL of water. The mixture was extracted with of ethyl acetate (20 mL x2). The organic layer was combined, and then concentrated under vacuum. The residue was purified by Flash- Prep-HPLC with the following condition to give 2-([[(lE,2R,3R,4R)-3-[(l E,3E)-5-[3-chloro-2- (fluoromethoxy)-5-formyl-6-hydroxy-4-methylphenyl]-3-methylpenta-l ,3-dien-l-yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)-acetic acid (533.3 mg, 64%, Compound 26) as a light yellow solid.
Flash-Prep-HPLC condition (IntelFlash-1): Column, C18 silica gel; mobile phase,
CH3CN/H20=55:45 increasing to CH3CN/H2O = 95:5 within 45 min; Detector, UV 254 nm.
LC-MS (ES, m/z): [M+H]+ : 510. H-NMR (300 MHz, DMSO-i¼) 512.58 (s, 2H), 10.30 (s, 1H), 5.88 (d, J= 16.2 Hz, 1H), 5.79 (d, J= 53.7 Hz, 2H), 5.40 (d, J= 16.2 Hz, 1H), 5.38 (m, 1H), 4.46 (s, 2H), 3.50 (d, J= 7.2 Hz, 2H), 3.23 (dd, J= 12.7, 4.0 Hz, 1H), 2.64 (s, 3H), 2.27 (q, J= 6.7 Hz, 1H), 1.84 (s, 3H), 1.82-1.59 (m, 3H), 1.35- 1.19 (m, 1H), 0.75 (d, J= 6.6 Hz, 3H), 0.68 (d, J=.6.6 Hz, 3H), 0.62 (s, 3H).
Example 29
[0177] Synthesis of 2-([[( 1 E,2R,3R,4R)-3-[( 1 E,3 E)-5-[3 -chloro-2-(fiuoromethoxy)-6-hydroxy-5-[( 1 E)- (hydroxyimino)methyl]-4-methylphenyl]-3-methylpenta-l ,3-dien-l -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid (Compound 27)
Figure imgf000062_0001
[0178] Step 1
A solution of 3-chloro-4-(fluoromethoxy)-6-hydroxy-2-methyl-5-[(2E,4E)-3-methyl-5- [(lR,2R,6R)-l ,2,6-trimethyl-3-oxocyclohexyl]penta-2,4-dien-l -yl]benzaldehyde [Example 1] (600 mg, 1.37 mmol) and hydroxyamine hydrochloride (93.5 mg, 1.35 mmol, 0.98 equiv) in pyridine (6 mL) was placed in a 100-mL round-bottom flask. The mixture was stirred for 2 h at room temperature. The resulting mixture was concentrated under vacuum to give (2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-2- (fluoromethoxy)-6-hydroxy-5-[(lE)-(hydroxyimino)methyl]-4-methylphenyl]-3-methylpenta-l,3-dien- l-yl]-2,3,4-trimethylcyclohexan-l-one (500 mg, crude) as yellow oil.
[0179] Step 2
A solution of (2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-2-(fluoromethoxy)-6-hydroxy-5-[(lE)- (hydroxyimino)methyl]-4-methylphenyl]-3-methylpenta-l,3-dien-l -yl]-2,3,4-trimethylcyclohexan-l-one (500 mg, 1.11 mmol) and ethyl 2-(aminooxy)acetate oxalic acid salt (925 mg, 4.42 mmol, 4.00 equiv) in pyridine (6 mL) was placed in a 100-mL round-bottom flask. The mixture was stirred for 1 h at room temperature. The resulting mixture was concentrated under vacuum to give resulted in of ethyl 2- ([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-2-(fluoromethoxy)-6-hydroxy-5-[( lE)- (hydroxyimino)methyl]-4-methylphenyl]-3-methylpenta-l,3-dien-l-yl]-2,3
trimethylcyclohexylidene]amino]oxy)acetate (550 mg, crude) as yellow solid.
[0180] Step 3
A mixture of ethyl 2-([[(lE,2R,3R,4R)-3-t(l E,3E)-5-[3-chloro-2-(fiuoromethoxy)-6-hydroxy- 5-[(lE)-(hydroxyimino)methy]]-4-methylphenyl]-3-rnethylpenta-l,3-dien-l-yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetate (550 mg, 0.99 mmol), lithium hydroxide (853 mg, 35.62 mmol) in tetrahydrofuran (6 mL) and water (2 mL) was placed in a 100-mL round-bottom flask. The resulting solution was stirred for 1 h at room temperature. The mixture was concentrated under vacuum. The resulting mixture was extracted with ethyl acetate (10 mL x3) and the organic layer was combined, then concentrated under vacuum. The residue was purified by Prep-HPLC with the following condition to give 2-([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-2-(fluoromethoxy)-6-hydroxy-5- [(lE)-(hydroxyimino)methyl]-4-methylphenyl]-3-methylpenta-l,3-dien-l -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid (242.2 mg, 46%, Compound 27) as a solid.
Prep-HPLC condition (Prep HPLCJvlC 1 ): Column: XBridge Shield RP18 OBD Column, 5um, 19* 150mm; Mobile Phase A: Waters(0.1 %FA), Mobile Phase B: ACN; Flow rate: 20 mL/min; Gradient: 56% B to 80% B in 8 min; Detector: 254 nm,.
LC-MS (ES, m/z): [M+H]+: 525, [M+Na-H]+: 547
H-NMR (300MHz, CD3OD, ppm): δ 8.56(S, IH), 5.89(d, J=16.2 Hz, IH), 5.72(s, IH), 5.54(s, IH), 5.44-5.33(m, 2H), 4.48(s, 2H), 3.56(d, J=7.2 Hz, 2H), 3.39-3.32(m, IH), 2.43(s, 3H), 2.19(q, J=6.6 Hz, IH), 1.86(s, 3H), 1.77-1.59(m, 3H), 1.25-1.48(m, I H), 0.84(d, J=6.0 Hz, 3H), 0.74-0.69(m, 6H).
Test Example 1
Effect on phosphorylation level of AMPK and ACC in HepG2 cells.
[0181] p-AMPK and p-ACC levels were measured by Western blot analysis using the antibodies for p- AMPK and p-ACC. Following reagents were purchased from the vendors, and the instruments listed below were used.
[0182] Reagents: AMP a (D5A2) rabbit mAb, phospho-acetyl-CoA carboxylase (Ser79) (D7D11) rabbit mAb, anti-rabbit IgG, HRP-linked antibody, and beta-actin (13E5) rabbit mAb were purchased from Cell Signaling Technology. ECL Western Blotting Detection Reagents were purchased from Thermo.
[0183] Instruments: Western Blot System (Invitrogen / Bio-Rad), and Imaging System (Cell
Biosciences)
Compound Treatment
[0184] HepG2 cells were cultured in MEM medium supplemented with 10% FBS at 37°C and under 5% CO2 condition. HepG2 cells (1 X 106 cells/well) were seeded into 6-well cell culture plates for 24hr. The cells were starved in serum-free MEM medium (3ml/well) for 24hr.
Test Compounds Preparation:
[0185] The compounds were dissolved in DMSO to make the stock solution of 5, 25, 50mM respectively. The compounds were diluted to final concentration in serum-free MEM medium to be 5, 25, and 50 μΜ.
Metformin Preparation:
[0186] Metformin was dissolved in PBS to make the stock concentration as 1M. Metformin was diluted to final concentration in serum-free MEM medium to be 5, and 50 mM
The serum-free MEM medium was discarded from each well. 3ml of 1 x compounds was added to each well. Cells were incubated in a 37°C, 5% CO2 incubator for 6 h (Compound 5,
Compound 19 and Metformin).
Western Blot Analysis
[0187] Total proteins (10 μg) from each sample was loaded to sample well of SDS-PAGE.
Electrophoresis was run at 120 V constant voltage until the blue marker reaches the end of the gel. The proteins were transferred to a PVDF membrane using Bio-Rad's Trans blot for 40 min at 300 mA.
After transfer, the membrane was blocked with blocking buffer for 2hr at room temperature. The membrane was incubated with the corresponding primary antibody solution at 4°C overnight. The membrane was washed for 5 min χ 3 with 1 xTBST and incubated with secondary antibody solution at room temperature for 45 min. The membrane was washed for 5min χ 3 with 1 xTBST and the detection was conducted with chemiluminescence detection reagents. The results are indicated in Figure 1. Test Example 2
Measurement of solubility in pH 4.0 and pH 7.4 phosphate buffer solution
1) Preparation of Stock Solutions
[0188] The stock solutions of test compounds and positive control compound were prepared in DMSO at the concentration of 30 mM. Diclofenac was used as positive control in the assay.
2) Procedures for Solubility Determination
[0189] The stock solution of each compound (10 μί) was placed in order into their proper 96-well rack, followed by adding 990 μΐ, of PBS at pH 4.0 and pH 7.4 into each vial of the cap-less Solubility Sample plate. This study was performed in duplicate. One stir stick was added to each vial and then vials were sealed using a molded PTDE/SIL 96-Well Plate Cover. The Solubility Sample plate was transferred to the Thermomixer Comfort plate shaker and incubated at 25 °C for 2 hours with shaking at 1 , 100 rpm. After 2 hours incubation, stir sticks were removed using a big magnet and all samples from the Solubility Sample plate were transferred into the filter plate. All the samples were filtered by using the vacuum manifold. The filtered samples were diluted with methanol.
3) Samples analyzed by LC-MS/MS :
[0190] The LC system comprised a Shimadzu liquid chromatograph separation system equipped with degasser DGU-20A3, solvent delivery unit LC-20AD, system controller CBM-20A, column oven CTO- 1 OAS VP and CTC Analytics HTC PAL System. Mass spectrometric analysis was performed using an API 4000 instrument from AB Inc (Canada) with an ESI interface. The data acquisition and control system were created using Analyst 1.6 software from ABI Inc. The results of the measurements are indicated in Table 2.
Table 2
Figure imgf000066_0001
Test Example 3
Determination of stability against mouse and human microsomes
[0191] Pooled human liver microsomes (Cat. 452161 ; Lot. 23418) were purchased from Corning and pooled male mouse liver microsomes (Cat. l 000; Lot. 1210302) were purchased from Xenotech. Microsomes were stored at - 80 °C prior to use. Ultra-pure H2O (40 μί) was used instead of NADPH solution in the negative control. The negative control was used to exclude the misleading factor that was resulted from instability of chemical itself. Samples with NADPH were prepared in duplicate. Negative controls were prepared in singlet.
[0192] The reaction was started with the addition of 4 of 200 μΜ compound solution to each master solution to get the final concentration of 2 μΜ. Verapamil was used as positive control in the assay. Aliquots of 50 μΐ. were taken from the reaction solution at 0, 15, 30, 45 and 60 min. The reaction was stopped by addition of 4 volumes of cold acetonitrile with IS (200 nM imipramine, 200 nM labetalol and 2 μΜ ketoprofen). Samples were centrifuged at 3,220 g for 40 minutes. Aliquot of 90 μΐ. of the supernatant was mixed with 90 ih of ultra-pure H2O and then was used for LC-MS/MS analysis. Results are shown in Table 3 by indicating remaining % of the test compound after 60 minutes incubation with liver microsomes from human and mouse in the presence of NADPH.
Table 3
Figure imgf000067_0001
Test Example 4
[0193] PK profiles of Compound 19 in the blood were analyzed by LC/MS/MS after single dose administration of the test compounds in SD rats.
Test animal: SD rat (n=3 /group)
Vehicle: 10 v/v% DMSO and 90 v/v% PEG400
Dose (single): iv I 1 and 5mg/kg, /?o / 5 and 25 mg/kg
Analytical instrument and condition: LC/MS/MS instrument [LC: Shimazu, MS: AB API4000 (ion source: ESI)], LC column: XSELECT™ CSH™ C I 8 2.5μπι (2.1 χ 50 mm), mobile phase: 5% acetonitrile (0.1 % formic acid) and 95% acetonitrile (0.1 % formic acid).
The results are shown in Figure 2. Test Example 5
In vivo efficacy in HFD-STZ C57BL/6J mice model
Diet Induced Obesity Model Establishment:
[0194] C57BL/6J mice were fed with high fat diet (Research Diets, D 12492) for 64 days, and streptozotocin (10 mg/kg) was given orally on day 22 and day 29. Compound treatment was done from day 36 to day 63.
Groups and Treatments
[0195] Based on the animal's body weight and oral glucose tolerance test (OGTT) on week 5, mice were randomly assigned to respective groups using a computer-generated randomization procedure.
Vehicle group: HFD/STZ + vehicle
Treatment group: HFD/STZ + test compound
Treatment dose: Compound 5 (po. qd x 28: 0.2mg/kg, 1 mg/kg and 5mg/kg)
Pioglitazone, a positive control (po. qd x 28: 30mg/kg)
Normal diet group: normal diet + vehicle
Number of animals per group: 8
Vehicle for Compound 5: PEG-400 (Sigma) / Tween 80 (Sigma) buffer = 95:5
Vehicle for pioglitazone: saline
Administration of test compounds:
[0196] Compound 5 and pioglitazone were administered orally once a day for 4 weeks with the dose indicated above.
Glucose Determination:
Blood glucose level was measured with the blood from tail vein every 2nd week by using Accu-Chek Aviva System.
Blood Chemistry Analysis:
[0197] The lipid profile of every 2nd week and terminal blood bio-chemistry parameters were tested as listed in Table 2. Blood samples were obtained from collection on Day 49 and 65, the samples were immediately processed by centrifugation at 4°C, 4,000 g for 15 minutes, and then they were transferred^ into a new test tube. Lipid profile and full panel blood bio-chemistry parameters were measured by using TOSHIBA TBA-40FR automated biochemical analyzer.
[0198] Figures 3 and 4 show the effect of the compounds of the present invention on the levels of triglyceride and total cholesterol in blood of mice. Figure 5 shows the effect of the compounds of the present invention on the blood glucose level of mice in fasting state.
Test Example 6
[0199] In a similar manner to Test Example 5, in vivo efficacy study of Compound 19 was carried out with HFD-STZ C57BL/6J mice model. As a result, Compound 19 (once a day oral treatment for 28 days at 0.25 to 16 mg/kg) significantly decreased blood glucose level (-68%), and HbAlC level (-31%). In OGTT assay, a group treated with Compound 19 showed significantly improved glucose absorption, specifically blood glucose level after 2 hours of glucose administration was 30% lower and glucose AUC was 25% lower than control group (a group treated with vehicle).
Test Example 7
[0200] An efficacy study was conducted by using Compound 19 (Cmpd 19) with db/db mice. Male db/db mice (7 to 8 weeks old) were purchased. The mice were kept in laminar flow rooms at constant temperature and humidity with one animal in each cage. Animals were housed in polycarbonate cages and in an environmentally monitored, well-ventilated room maintained at a temperature of (22 ± 2°C) and a relative humidity of 40%-70%. Fluorescent lighting provided illumination approximately 12 hours per day. The bedding material was soft wood, which was changed once per week. Animals had free access to normal diet (Beijing Keaoxieli Feed Co., Ltd) during the entire study period except for time periods specified by the protocol. Sterile drinking water in a bottle was available to all animals ad libitum during the quarantine and study periods.
[0201] After 5 days of acclimation period, the mice were randomly assigned to 8 groups, based on body weight and fasting blood glucose level thereof on day 0. The study design is shown in Table 4. Table 4. Study design
Figure imgf000070_0001
[0202] Cmpd 19 was administered as a solution in the vehicle (PEG-400/ DMSO (90: 10 v/v)), and metformin was administered as a solution in saline. The drugs were orally administered to the mice once a day for 28 consecutive days. Body weight and food consumption were measured during the course of this study. The following data were obtained to confirm the efficacy of the drugs.
1. Results of Oral Glucose Tolerant Test (OGTT)
[0203] Oral glucose tolerant tests were conducted on Day 0 (baseline) and Day 28 (endpoint) after treatment. Following oral administration 0.5 g/kg glucose, the blood glucose levels were measured by using Accu-Chek Performa System. Blood samples were collected at 0, 30, 60 and 120 minutes after glucose administration. The results are shown in Figures 6 and 7.
[0204] The results on Day 28 indicate that administration of Cmpd 19 exhibits tendency of dose- dependent reduction in glucose level, comparing with the result of the control group. Further, the results significantly indicate that administration of combination of Cmpd 19 with metformin exhibit reduction of the glucose level.
[0205] Area under curve (AUC) of blood glucose level was calculated based on the foregoing results and shown in Table 5. The results of Table 5 shows that administration of Cmpd 19 alone (16 mg/kg) exhibits reduction of AUC with significance. Further, administration of Cmpd 19 in combination with metformin exhibits remarkable reduction of AUC.
Table 5. AUC of blood glucose level in OGTT on Day 0 and Day 28
Figure imgf000071_0001
Note:* p<0.05, ** p<0.01 compared with vehicle control group
2. Fasting blood glucose level
[0206] Fasting blood glucose level was measured via tail vein at week 0, 2, 3 and 4 by using Accu- Chek Performa System. Animals were fasted overnight (16 hours) before measurement. The results are shown in Figure 8. The results indicate that fasting blood glucose level reduced dose-dependently by administration of Cmpd 19 alone or combination of Cmpd 19 and metformin.
3. Serum triglyceride level
[0207] Serum triglyceride level was measured during the study. Blood samples were collected from the orbital veins at week 4, and then immediately processed by centrifugation at 4°C, 6,000 g for 15 minutes. After the samples were transferred into new test tubes. Triglyceride level was measured by using TOSHIBA TBA-40FR automated biochemical analyzer. The results are shown in Figure 9. The results indicate tendency of strong reduction in triglyceride level observed in the groups administered with the combination of Cmpd 19 and metformin.
4. Hemoglobin Aic level
[0208] Hemoglobin Aic (HbAic) level was measured on Day 28. Blood samples were collected from the orbital veins at week 4, and then immediately processed by centrifugation at 4°C, 6,000 g for 15 minutes, and then they were transferred into new test tubes. HbA lc level was measured by using- TOSHIBA TBA-40FR automated biochemical analyzer.
[0209] The results are shown in Figure 10. The results indicate that reduction of HbAic level in the group administered with a combination of Cmpd 19 and metformin was observed with significance.

Claims

Claims
[Claim 1] A pharmaceutical composition comprising a compound represented by formula I:
Figure imgf000073_0001
wherein
R1 is formyl, or -CH=N-0-Y, in which Y is a hydrogen atom or Ci-6 alkyl;
R2 is Ci-6 alkyl that may be substituted with 1 to 5 fluorine atoms;
R3 is a hydrogen atom or Ci-6 alkyl;
R4 is hydroxy, Ci-6 alkylamino, C3-7 cycloalkylamino, or 3- to 7-membered
heterocycloalkylamino which contains -0-, -S-, -NR6-, -SO- or -SO2- as a ring atom, or R3 and R4 together with the carbon atom to which they are attached form >C=N-0-Z;
Z is a hydrogen atom, Ci-6 alkyl, -CO(CH2)„-R5 or -(CH2)n-R5;
n is an integer selected from 1 to 4;
R5 is -CO2R6, -CONH2, -CONR7R8, -OCONR7R8, -S02NR7R8, -S02R9, hydroxy, -NHS02R9, or -NR7R8;
R6, R7 and R8 are each independently selected from a group consisting of a hydrogen atom, Ci-6 alkyl, and C3-7 cycloalkyl; or R7 and R8 together with the nitrogen atom to which they are attached form a nitrogen-containing 3- to 7-membered heterocyclic ring which may further contain -0-, -S-, -NR6-, -SO- or -S02- as a ring atom, in which the heterocyclic ring may be substituted with one or more substituents selected from hydroxy and Ci-6 alkyl;
R9 is Ci-6 alkyl, or C3-7 cycloalkyl;
a pharmaceutically acceptable salt or a solvate thereof,
wherein the composition is for use in combination with administration of metformin.
[Claim 2] The pharmaceutical composition according to claim 1, wherein R4 is selected from the group consisting of hydroxy, methylamino, ethylamino, cyclopropylamino, and oxetan-3-ylamino.
[Claim 3] The pharmaceutical composition according to claim 1, wherein R3 and R4 together with the carbon atom to which they are attached form >C=N-0-Z in which Z is selected from the group consisting of a hydrogen atom, Ci-6 alkyl, -CH2CO2R6, morpholinoethyl, piperazinylethyl, N-methyl- piperazinylethyl, and N,N-dimethylamino-ethyl.
[Claim 4] The pharmaceutical composition according to claim 2 or 3, wherein R1 is formyl.
[Claim 5] The pharmaceutical composition according to claim 2 or 3, wherein R1 is -CH=N-OH.
[Claim 6] The pharmaceutical composition according to claim 2 or 3, wherein R1 is -CH=N- OMe.
[Claim 7] The pharmaceutical composition according to claim 1, wherein the compound represented by formula (I) is selected from the group consisting of
3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R)-3-hydroxy-l ,2,3,6-tetramethylcyclohexyl)-3- methylpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde,
3-chloro-6-hydroxy-5-((2E,4E)-5-((l R,2R,6R,E)-3-(hydroxyimino)- 1,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde,
(E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)-l,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l -yl)-4-methoxy-2-methylbenzaldehyde oxime,
3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxyimino)-l ,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l-yl)-2-methylbenzaldehyde oxime,
3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((l R,2R,6R,E)-3-(hydroxyimino)-l,2,6- trimethylcyclohexyl)-3 -methylpenta-2,4-dien- 1 -y l)-2-methy lbenzaldehyde,
[(lE,2R,3R,4R)-3-[(l E,3E)-5-(3-chloro-5-formyl-6-hydroxy-2-methoxy-4-methylphenyl)-3- methylpenta-l,3-dien-l-yl]-2,3,4-trimethylcycIohexylidene]amino 2-(dimethylamino)acetate,
(E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,3S,6R)-3-hydroxy-l ,2,3,6- tetramethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde oxime,
(E)-3-chloro-6-hydroxy-4-methoxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(methoxyimino)- 1,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l -yl)-2-methylbenza!dehyde O-methyl oxime,
(E)-3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(methoxyimino)-
1 ,2,6-trimethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-2-methylbenzaldehyde O-methyl oxime, (E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((l R,2R,6R,E)-3-(hydroxyimino)-l ,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -y l)-4-methoxy-2-methy lbenzaldehyde O-methyl oxime,
(E)-3-chloro-4-(fluoromethoxy)-6-hydroxy-5-((2E,4E)-5-((lR,2R,6R,E)-3-(hydroxytmino)- l,2,6-trimethylcyclohexyl)-3-methylpenta-2,4-dien-l-yl)-2-methylbenzaldehyde O-methyl oxime,
3-chloro-4-(fluoromethoxy)-6-hydroxy-2-methyl-5-((2E,4E)-3-methyl-5-((lR,2R,6R,E)-l,2,6- trimethyl-3 -(2-morpholinoethoxyim ino)cyclohexyl)penta-2,4-dien- 1 -y l)benzaldehyde,
3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(cyclopropylamino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien-l-yl)-4-(fluoromethoxy)-6-hydroxy-2-methylbenzaldehyde,
(E)-3-chloro-6-hydroxy-5-((2E,4E)-5-((lR,2R,3S,6R)-3-hydroxy-l,2,3,6- tetramethylcyclohexyl)-3-methylpenta-2,4-dien- 1 -yl)-4-methoxy-2-methylbenzaldehyde O-methyl oxime,
(E)-3-chloro-5-((2E,4E)-5-((lR,2R,6R,E)-3-(2-(dimethylamino)ethoxyimino)-l ,2,6- trimethylcyclohexyl)-3-methylpenta-2,4-dien-l-yl)-6-hydroxy-4-methoxy-2-methylbenzaldehyde O- methyl oxime,
(E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-((lR,2R,6R,E)-l,2,6- trimethyl-3-(2-moφholinoethoxyimino)cyclohexyl)penta-2,4-dien- 1 -yl)benzaldehyde O-methyl oxime,
(E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-((l R,2R,6R,E)-l,2,6- trimethyl-3-(2-(4-methylpiperazin- 1 -yl)ethoxyimino)-cyclohexyl)penta-2,4-dien- 1 -yl)benzaldehyde O- methyl oxime,
ethyl 2-([[(lE,2R,3R,4R)-3-[(l E,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (methoXyimino)methyl]-4-methylphenyl]-3-methylpenta-l ,3-dien-l-yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetate, 2- ([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(l E)- (methoxyirnino)rnethyl]-4-methylphenyl]-3-methyIpenta- 1 ,3-dien- 1 -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid,
3- chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(ethylamino)-l,2,6-trimethylcyclohexyl)-3- methyIpenta-2,4-dien-l-yI)-6-hydroxy-4-methoxy-2-rnethylbenzaldehyde,
(E)-3-chloro-5-((2E,4E)-5-((l R,2R,6R)-3-(ethylarnino)-l ,2,6-trimethylcyclohexyl)-3- methy lpenta-2,4-dien- 1 -y l)-6-hydroxy-4-methoxy-2-methy lbenzaldehyde O-methyl oxime,
3-chloro-5-((2E,4E)-5-((lR,2R,6R)-3-(cyclopropylamino)-l,2,6-trimethylcyclohexyl)-3- methylpenta-2,4-dien- 1 -yl)-6-hydroxy-4-methoxy-2-methy lbenzaldehyde,
(E)-3-chloro-5-((2E,4E)-5-(( 1 R,2R,6R)-3-(cyclopropylamino)-l ,2,6-trimethylcyclohexyl)-3- methy lpenta-2,4-dien- 1 -yl)-6-hydroxy-4-methoxy-2-methy lbenzaldehyde O-methyl oxime,
2-[(2E,4E)-5-[(l R,2R,6R)-3-amino-l,2,6-trimethylcyclohexyl]-3-methylpenta-2,4-dien-l -yl]- 4-chloro-3-methoxy-6-[(l E)-(methoxyimino)methyl]-5-methylphenol,
(E)-3-chloro-6-hydroxy-4-methoxy-2-methyl-5-((2E,4E)-3-methyl-5-((l R,2R,6R)-1 ,2,6- trimethyl-3-(oxetan-3-ylamino)cyclohexyl)penta-2,4-dien- 1 -yl)benzaldehyde O-methyl oxime,
2-([[(lE,2R,3R)4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-fluoromethoxy-5-formyl-4- methylphenyl]-3-methylpenta-l,3-dien-l -yl]-2,3,4-trimethylcyclohexylidene]-amino]oxy)acetic acid,
2-([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-6-hydroxy-2-methoxy-5-[(lE)- (hydroxyimino)methyl]-4-methylphenyl]-3-methylpenta-l ,3-dien-l -yl]-2,3,4- trimethylcyclohexylidene]amino]oxy)acetic acid,
2-([[(lE,2R,3R,4R)-3-[(lE,3E)-5-[3-chloro-2-methoxy-6-hydroxy-5-[(lE)- (hydroxyimino)methy l]-4-methylphenyl]-3 -methyl penta- 1 ,3 -dien- 1 -y l]-2,3 ,4- trimethylcyclohexylidene]amino]oxy)acetic acid,
and pharmaceutically acceptable salts thereof.
[Claim 8] The pharmaceutical composition according to any one of claims 1 to 7, wherein the composition is for use is treating or preventing a disease selected from diabetes, obesity, and hyperlipidemia.
[Claim 9] The pharmaceutical composition according to claim 8, wherein the disease is type 2 diabetes.
[Claim 10] The pharmaceutical composition according to any one of claims 1 to 9, wherein the compound represented by formula (I) is administered in amount of 0.1 to 100 mg/kg/day.
[Claim 11] The pharmaceutical composition according to any one of claims 1 to 10, wherein metformin is administered in amount of 50 to 1000 mg kg/day.
PCT/JP2018/020870 2017-05-23 2018-05-23 Use of ascochlorin derivative for combination therapy WO2018216821A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2017-101585 2017-05-23
JP2017101585 2017-05-23

Publications (1)

Publication Number Publication Date
WO2018216821A1 true WO2018216821A1 (en) 2018-11-29

Family

ID=64395776

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2018/020870 WO2018216821A1 (en) 2017-05-23 2018-05-23 Use of ascochlorin derivative for combination therapy

Country Status (1)

Country Link
WO (1) WO2018216821A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994005274A1 (en) * 1992-09-10 1994-03-17 Immuno Japan Inc. Glycation inhibitor
EP1176134A1 (en) * 1999-03-11 2002-01-30 Nuclear Receptor Research Limited Ligands of nuclear receptors ppar's
EP1616856A1 (en) * 2003-02-24 2006-01-18 NRL Pharma, Inc. Novel transcriptional factor, process for producing the same and use thereof
WO2007060976A1 (en) * 2005-11-25 2007-05-31 Yokohama Tlo Company, Ltd. Novel ascochlorin derivative compounds and medicinal compositions containing the same
WO2017119515A1 (en) * 2016-01-05 2017-07-13 Nrl Pharma, Inc. Ascochlorin derivative and use thereof as ampk activator

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994005274A1 (en) * 1992-09-10 1994-03-17 Immuno Japan Inc. Glycation inhibitor
EP1176134A1 (en) * 1999-03-11 2002-01-30 Nuclear Receptor Research Limited Ligands of nuclear receptors ppar's
EP1616856A1 (en) * 2003-02-24 2006-01-18 NRL Pharma, Inc. Novel transcriptional factor, process for producing the same and use thereof
WO2007060976A1 (en) * 2005-11-25 2007-05-31 Yokohama Tlo Company, Ltd. Novel ascochlorin derivative compounds and medicinal compositions containing the same
WO2017119515A1 (en) * 2016-01-05 2017-07-13 Nrl Pharma, Inc. Ascochlorin derivative and use thereof as ampk activator

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
JEONG, JI-HAK ET AL.: "4-0-methylascochlorin, methylated derivative of ascochlorin, stabilizes HIF-1alpha via AMPK activation", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, vol. 406, no. 3, 15 February 2011 (2011-02-15), pages 353 - 358, XP028166930 *

Similar Documents

Publication Publication Date Title
EP3400209B1 (en) Ascochlorin derivative and use thereof as ampk activator
JP6637136B2 (en) 2,2-Difluoropropionamide derivatives of bardoxolone methyl, polymorphs and methods of use
EP2934503B1 (en) Deuterium substituted fumarate derivatives
EA021538B1 (en) Hepatitis c virus inhibitors
CA2969803A1 (en) Isoxazole hydroxamic acid compounds as lpxc inhibitors
CN112105601B (en) Benzenesulfonamide derivatives and related FERROSTATIN-1 analogs as cell death inhibitors
KR20210102887A (en) Crystalline spirocyclic compound inhibitors of tryptophan hydroxylase 1 (TPH1) for treating diseases or disorders associated with peripheral serotonin
AU2019344905A1 (en) Crystalline forms of a farnesoid X receptor agonist
KR20160038079A (en) Benzothiazolone compound
CN110938094A (en) Preparation method and application of thyroid hormone receptor β subtype agonist derivative
US11298358B2 (en) Combination therapy using ascochlorin derivative
WO2018216821A1 (en) Use of ascochlorin derivative for combination therapy
KR20200128096A (en) Amide prodrugs of small molecule nuclear receptor modulators
JP7282036B2 (en) novel aromatic compounds
WO2005016331A1 (en) Therapeutic agent for neuropathic pain containing n-(benzoyl)amino acid derivative as active ingredient
CN116514902A (en) Deuterated peptidomimetic compounds and application thereof
JP2024516851A (en) Combining brain-targeted activity with fatty acid amide hydrolase (FAAH) cleavable prodrugs and peripherally restricted FAAH inhibitors
EP3134398A1 (en) Autotaxin inhibitors

Legal Events

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

Ref document number: 18805592

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

NENP Non-entry into the national phase

Ref country code: JP

122 Ep: pct application non-entry in european phase

Ref document number: 18805592

Country of ref document: EP

Kind code of ref document: A1