WO2011160974A2 - Statin derivatives - Google Patents

Statin derivatives Download PDF

Info

Publication number
WO2011160974A2
WO2011160974A2 PCT/EP2011/059778 EP2011059778W WO2011160974A2 WO 2011160974 A2 WO2011160974 A2 WO 2011160974A2 EP 2011059778 W EP2011059778 W EP 2011059778W WO 2011160974 A2 WO2011160974 A2 WO 2011160974A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
mmol
solution
formula
group
Prior art date
Application number
PCT/EP2011/059778
Other languages
French (fr)
Other versions
WO2011160974A3 (en
Inventor
Laura Storoni
Annalisa Bonfanti
Francesca Benedini
Francesca Oliva
Daniela Miglietta
Ennio Ongini
Original Assignee
Nicox S.A.
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 Nicox S.A. filed Critical Nicox S.A.
Publication of WO2011160974A2 publication Critical patent/WO2011160974A2/en
Publication of WO2011160974A3 publication Critical patent/WO2011160974A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more 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, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/30Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members
    • C07D207/34Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two 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 carbon atoms

Definitions

  • the present invention relates to new statin nitroderivatives . More particularly, the present invention relates to dinitrate statins, pharmaceutical compositions containing them and their use as cholesterol-reducing drugs, as drugs having antioxidant, antithrombotic and anti-inflammatory activity, effects on endothelial function, immunosuppressive properties and for treating and/or preventing acute coronary syndromes, stroke, neurodegenerative disorders, such as Alzheimer's and Parkinson's disease as well as autoimmune diseases, such as multiple sclerosis.
  • Statins is a class of compounds which comprises as main components lovastatin, simvastatin, pravastatin, mevastatin, fluvastatin, atorvastatin, rosuvastatin and cerivastatin (rivastatin) .
  • Statins possess a side group that is structurally similar to HMG-CoA ( 3-hydroxy-3- methylglutaryl coenzyme A) .
  • Fluvastatin, atorvastatin, rosuvastatin and cerivastatin are entirely synthetic compounds containing a heptanoic acid side chain, the remainders being fungal metabolites.
  • statins are inhibitors of HMG-CoA reductase, an enzyme which catalyses an early, rate-limiting step in cholesterol biosynthesis, reduce triglyceride levels and are also indicated for raising HDL- C levels (P.O. Bonetti et al., European Heart Journal (2003) 24, 225-248) .
  • WO 2004/105754 discloses mononitrate statins having improved pharmacological activity and enhanced tolerability and their use for treating and/or preventing several diseases . It has been so surprisingly found that the dinitrate compounds of the invention provide enhanced nitric oxide (NO) release over mononitrate analogs (WO 2004/105754). A consideration of stoichiometry leads to an expectation of a doubling of nitrite levels.
  • Object of the present invention are, therefore, statin nitroderivatives of general formula (I) or pharmaceutically acceptable salts or stereoisomers thereof
  • R is selected from the group consisting of (Ila) (Ilf) :
  • w is an integer selected between 0 and 1 ;
  • R 1 and R 2 can be equal or different and are independently selected from the group consisting of hydrogen and C 1 -C4 alkyl ;
  • Y is selected from the group consisting of:
  • R 3 , R 4 , R 5 , R 6 and R 8 are independently a straight or branched C 1 -C 1 0 alkylene;
  • n is an integer from 0 to 1 ;
  • nl is an integer from 0 to 1 ;
  • p is an integer from 0 to 4 ;
  • pi is an integer from 0 to 4 ;
  • R 7 is selected from the group consisting of hydrogen and
  • R 9 is a saturated, unsaturated or aromatic heterocyclic 5 or 6 members ring, containing one or more heteroatoms selected from nitrogen, oxygen or sulphur.
  • Ci-Cio alkylene refers to branched or straight chain Ci-Cio hydrocarbon, such as methylene, ethylene, propylene, isopropylene, n-butylene, pentylene, n-hexylene and the like.
  • Ci-Cio alkyl refers to branched or straight chain alkyl groups comprising one to ten carbon atoms, including methyl, ethyl, n-propyl, isopropyl, n- butyl, isobutyl, t-butyl, pentyl, hexyl, octyl and the like .
  • heterocyclic refers to saturated, unsaturated or aromatic 5 or 6 members ring, containing one or more heteroatoms selected from nitrogen, oxygen, sulphur, such as for example pyridine, pyrazine, pyrimidine, pyrrolidine, morpholine, imidazole and the like.
  • Preferred compounds according to the invention are the compounds of general formula (I) or pharmaceutically acceptable salts or stereoisomers thereof as defined above wherein R is the group (lid) or (lie) .
  • An embodiment of the invention relates to the compounds of general formula (I) or pharmaceutically acceptable salts or stereoisomers thereof as defined above wherein w is 1.
  • R 1 and R 2 which can be equal or different are independently selected from the group consisting of hydrogen and C3 ⁇ 4.
  • Another preferred embodiment of the invention relates to the compounds of general formula (I) or pharmaceutically acceptable salts or stereoisomers thereof as defined above wherein w is 0.
  • Another preferred embodiment of the invention relates the compounds of general formula (I) or pharmaceutically acceptable salts or stereoisomers thereof as defined above wherein Y of the group A) is selected from the group consisting of:
  • Another embodiment of the invention relates to the compounds of Formula (I) or pharmaceutically acceptable salts or stereoisomers thereof as defined above wherein
  • Y of the roup B is selected from the group consisting of:
  • Another embodiment of the invention relates to the compounds of Formula (I) or pharmaceutically acceptable salts or stereoisomers thereof as defined above wherein Y of the group C) is selected from the group consisting of:
  • Another embodiment of the invention relates to the compounds of Formula (I) or pharmaceutically acceptable salts or stereoisomers thereof as defined above selected from the roup consisting of:
  • Another aspect of the present invention includes the use of the compounds of formula (I) in combination with at least a compound used to treat cardiovascular diseases selected from the group consisting of: ACE inhibitors, renin inhibitors, angiotensin II receptor antagonists, beta-adrenergic blockers, calcium channel blockers, antithrombotics such as aspirin, nitrosated ACE inhibitors, nitrosated renin inhibitors, nitrosated angiotensin II receptor antagonists, nitrosated beta-adrenergic blockers and nitrosated aspirin.
  • ACE inhibitors, angiotensin II receptor antagonists, beta-adrenergic blockers, calcium channel blockers, antithrombotics are described in the literature such as The Merck Index (13 th edition) .
  • Suitable nitrosated compounds are disclosed in WO 98/21193, WO 2005/011646, WO 97/16405, WO 2004/106300, WO 2004/110432 and WO 2007/045551.
  • the present invention also provides pharmaceutical kits comprising one or more containers filled with one or more of the compounds and/or compositions of the present invention and one or more of the compounds used to treat cardiovascular diseases reported above.
  • the invention includes also the pharmaceutically acceptable salts of the compounds of formula (I) and stereoisomers thereof.
  • the compounds according to the present invention when they contain in the molecule one salifiable nitrogen atom, can be transformed into the corresponding salts by reaction in an organic solvent such as acetonitrile, tetrahydrofuran with the corresponding organic or inorganic acids.
  • organic acids examples include oxalic, tartaric, maleic, succinic, citric acids.
  • inorganic acids are: nitric, hydrochloric, sulphuric, phosphoric acids. Salts with nitric acid are preferred.
  • the compounds of the invention which have one or more asymmetric carbon atoms can exist as optically pure enantiomers, pure diastereomers , enantiomers mixtures, diastereomers mixtures, enantiomer racemic mixtures, racemates or racemate mixtures.
  • object of the present invention are also pharmaceutical compositions containing at least a compound of the present invention of formula (I) together with non toxic adiuvants and/or carriers usually employed in the pharmaceutical field.
  • the daily dose of active ingredient that should be administered can be a single dose or it can be an effective amount divided into several smaller doses that are to be administered throughout the day. Usually, total daily dose may be in amounts preferably from 1 to 500 mg.
  • the dosage regimen and administration frequency for treating the mentioned diseases with the compound of the invention and/or with the pharmaceutical compositions of the present invention will be selected in accordance with a variety of factors, including for example age, body weight, sex and medical condition of the patient as well as severity of the disease, route of administration, pharmacological considerations and eventual concomitant therapy with other drugs. In some instances, dosage levels below or above the aforesaid range and/or more frequent may be adequate, and this logically will be within the judgment of the physician and will depend on the disease state.
  • the compounds of the invention may be administered orally, parenterally, rectally or topically, by inhalation or aerosol, in formulations eventually containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles as desired. Topical administration may also involve the use of transdermal administration such as transdermal patches or iontophoresis devices.
  • transdermal administration such as transdermal patches or iontophoresis devices.
  • parenteral includes subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques.
  • injectable preparations for example sterile injectable aqueous or oleaginous suspensions may be formulated according to known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent.
  • acceptable vehicles and solvents are water, Ringer's solution and isotonic sodium chloride.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono or diglycerides , in addition fatty acids such as oleic acid find use in the preparation of injectables.
  • Suppositories for rectal administration of the drug can be prepared by mixing the active ingredient with a suitable non-irritating excipient, such as cocoa butter and polyethylene glycols.
  • Solid dosage forms for oral administration may include capsules, tablets, pills, powders, granules and gels.
  • the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as in normal practice, additional substances other than inert diluents, e.g. lubricating agents such as magnesium stearate.
  • the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings.
  • Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs containing inert diluents commonly used in the art, such as water. Such compositions may also comprise adjuvants, such as wetting agents, emulsifying and suspending agents, and sweetening, flavouring and the like.
  • statin nitroderivatives possess enhanced anti-inflammatory, antiplatelet and antithrombotic effects as compared to native statins. Moreover, they can be effective also in the other pathologies such as acute coronary syndromes, stroke, peripheral vascular diseases such as peripheral ischemia, all disorders associated with endothelial dysfunctions such as vascular complications in diabetic patients and atherosclerosis, neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD) , autoimmune diseases such as multiple sclerosis .
  • AD Alzheimer's disease
  • PD Parkinson's disease
  • M is an alkali metal or alkaline earth metal such as sodium or calcium and R is as above defined;
  • the reaction is carried out in an aprotic polar/non polar solvent such as DMF, THF, toluene at a temperature ranging from -20°C to 60°C;
  • the reaction is completed within a time ranges from 30 minutes to 24 hours.
  • the reaction may be carried out in presence in the presence of a condensing agent such as dicyclohexylcarbodiimide (DCC) , N' - ( 3-dimethylaminopropyl ) -N-ethylcarbodiimide hydrochloride (EDAC) , N, N' -carbonyldiimidazole (CDI), optionally in the presence of a base, for example DMAP .
  • a condensing agent such as dicyclohexylcarbodiimide (DCC) , N' - ( 3-dimethylaminopropyl ) -N-ethylcarbodiimide hydrochloride (EDAC) , N, N' -carbonyldiimidazole (CDI)
  • DCC dicyclohexylcarbodiimide
  • EDAC 3-dimethylaminopropyl
  • CDI N, N' -carbonyldiimidazole
  • the reaction is carried out in an inert organic solvent dry such as N, N' -dimethylformamide, tetrahydrofuran, benzene, toluene, dioxane, a polyhalogenated aliphatic hydrocarbon at a temperature from -20°C and 50°C.
  • an inert organic solvent dry such as N, N' -dimethylformamide, tetrahydrofuran, benzene, toluene, dioxane, a polyhalogenated aliphatic hydrocarbon at a temperature from -20°C and 50°C.
  • the reaction is completed within a time range from 30 minutes to 36 hours; 2.2) deprotecting the compounds obtained in the step 2.1) .
  • Fluoride ion is the preferred method for removing silyl ether protecting group.
  • the deprotecting of the carboxylic function may be carried out by hydrogenation in presence of Pd/C in an aprotic polar/non polar solvent such as THF at temperature ranging from 0°C to 30 °C and a pressure ranging from 1 to 5 bar;
  • an aprotic polar/non polar solvent such as THF
  • the deprotecting of the carboxylic function may be carried out with palladium tetrakis, in presence of dimedone and triphenylphosphine or in presence of morpholine, in an aprotic polar/non polar solvent such as THF, DMF or CH 2 CI 2 , at temperature ranging from -10 °C to
  • R is as above defined and M is an alkali metal or alkaline earth metal such as sodium or calcium,
  • reaction may be carried out in an aprotic polar/non polar solvent such as THF, DMF or CH 2 CI 2 , at temperature ranging from -80°C to 60°C.
  • an aprotic polar/non polar solvent such as THF, DMF or CH 2 CI 2
  • Hal is selected from Br or CI
  • aprotic polar/non polar solvent such as THF, DMF or CH 2 CI 2 , at temperature ranging from -80°C to 60°C;
  • Y is as above defined in the presence of a condensing agent such as dicyclohexylcarbodiimide (DCC) , N' - ( 3-dimethylaminopropyl ) -N-ethylcarbodiimide
  • DCC dicyclohexylcarbodiimide
  • reaction is carried out in an inert organic solvent dry such as N, N' -dimethylformamide, tetrahydrofuran, benzene, toluene, dioxane, a polyhalogenated aliphatic hydrocarbon at a temperature from -20°C and 50°C.
  • organic solvent dry such as N, N' -dimethylformamide, tetrahydrofuran, benzene, toluene, dioxane, a polyhalogenated aliphatic hydrocarbon at a temperature from -20°C and 50°C.
  • reaction is completed within a time range from 30 minutes to 36 hours; 5) deprotecting of the diol function of compound of formula (VII) wherein R is as above defined in presence of fluoride salt of alkylamine such as triethylamine trihydrofluoride or tetrabutylamonium fluoride and acetic acid, in an aprotic polar/non polar solvent such as THF, at temperature ranging from -10°C to 30°C.
  • fluoride salt of alkylamine such as triethylamine trihydrofluoride or tetrabutylamonium fluoride and acetic acid
  • an aprotic polar/non polar solvent such as THF
  • Another embodiment of the invention relates to compounds of formula (VI) wherein R is (He) .
  • atorvastatin calcium salt 5.0 g, 4.33 mmol
  • DMF 75 mL
  • allyl bromine 2.1 g, 17.30 mmol
  • the solution was stirred at room temperature for 72 hours.
  • the solution was diluted with 3 ⁇ 40 (200mL) and extracted with EtOAc (3 x 100 mL) .
  • the combined organic layers were washed with brine (2 x 100 mL) , dried over a 2 S0 4 and concentrated under reduced pressure.
  • the reaction was stirred at room temperature for 24 hours.
  • the solution was diluted with AcOEt (250 mL) and washed with an aqueous solution of 5 % Na3 ⁇ 4P0 4 (2x100 mL) .
  • the organic layer was washed with 3 ⁇ 40 (4x100 mL) and brine, then dried over Na 2 S0 4 and concentrated under reduced pressure.
  • the residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, eluent: gradient from n-hexane/EtOAc 7/3 to EtOAc during 1.4 L) affording the compound (5) (0.81 g, 58%).
  • the solution was diluted with AcOEt (100 mL) and washed with an aqueous solution of 5 % Na3 ⁇ 4P0 4 (2x50 mL) .
  • the organic layer was washed with 3 ⁇ 40 (4x50 mL) and brine, then dried over Na 2 SC>4 and concentrated under reduced pressure.
  • the residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, eluent: gradient from n- hexane/AcOEt 55/45 to n-hexane/AcOEt 3/7 during 1740 mL) affording the compound (7) (0.31 g, yield 23%).
  • statins dinitrate derivatives disclosed in examples 1-3, 5-8, 10, 11 and 13-16 were tested in vitro in isolated rabbit thoracic aorta preparations (Wanstall J.C. et al . , Br. J. Pharmacol., 134:463-472, 2001) .
  • PSS physiological salt solution
  • Each ring was mounted under 2 g passive tension in 5 ml organ bath. Isometric tension was recorded with a Grass transducer (Grass FT03) attached to a BIOPAC MP150 System. Preparations were allowed to equilibrate for 1 h, then contracted submaximally with noradrenaline (NA, 1 ⁇ ) and, when the contraction was stable, acetylcholine (ACh, 10 ⁇ ) was added. A relaxant response to ACh indicated the presence of a functional endothelium. When a stable precontraction was reached, a cumulative concentration- response curve to each vasorelaxant agent was obtained in the presence of a functional endothelium.
  • Table 1 reports the EC 50 of the tested compounds, the results shown that the dinitrates of the invention showed improved EC 50 values in vessel relaxation assay compared to mononitrate derivatives (Compound A and B) which are greater than two fold. Furthermore, in experiments performed in presence of ODQ (10 ⁇ ) , the vasorelaxant responses to all the tested drugs were inhibited showing that the vasodilative effects are mediated by NO.
  • Example F2 Evaluation of pharmacokinetic parameters
  • Reference compound Atorvastatin .
  • the formulations of the tested compounds were prepared on the day of dosing, immediately before of administration, by dissolving an appropriate amount of the test compound in PEG400/DMA 70/30, in order to obtain the following target concentrations :
  • Test compounds were administered orally to each animal by gavage . Animals were fasted overnight before dosing. On the day of dosing the food was offered about 4 h post dosing. Serial blood samples (about 0.3 mL) were collected from the catheter of each SVC catheterized rat, using syringes containing Na-heparin as anticoagulant, at pre-dose, 15 min, 30 min, 1, 2, 4, 6 and 8 h post-dosing. The samples were then transferred into pre-cooled tubes (containing 7.5 L of a solution of sodium fluorure, 40 mg/mL and DICHLORVOS, 10 mg/mL, in order to inhibit plasma esterase activity, in water for injection), protected from light. The blood was centrifuged at 10000 rpm for 3 min at +4 °C to collect plasma (within 30 min from blood collection) , and plasma samples were stored at -80°C.
  • Plasma samples were thawed in ice bath. Plasma aliquots of 50 yL were protein precipitated in triplicate using 150 yL of acetonitrile to which were also added 10 yL of DMSO, vortex-mixed and centrifuged 10 min at 4°C at 4000 rpm. The supernatant was transferred to a clean plate and injected onto a LC-MS system.
  • Atorvastatin ortho and para hydroxy metabolites levels were quantified using a calibration curve (range 0.01-10 ⁇ ) prepared in rat plasma, spiking an appropriate volume of working solution in 50 yL of plasma and then precipitating the proteins (see below for standard curve preparation) using 150 yL of acetonitrile, vortex-mixed and centrifuged 10 min at 4°C at 4000 rpm. The supernatant was transferred to a clean plate and injected onto a LC-MS system. Analytical conditions were as follows: Column: ACQUITY BEH Phenyl 50 x 2.1 mm (1.7y) @ 40°C
  • Detection was performed using a Waters Quattro Micro API mass spectrometer operated in ESI+ mode.
  • the multiple reaction monitoring (MRM) pair monitored was m/z 559.26- ⁇ 440.2 for atorvastatin and m/z 575.16 -> 440.2 for ortho and para hydroxy metabolites.
  • the autosampler cooler was maintained at 4°C.
  • Table 2 reports a summary of pharmacokinetic parameters of the compounds of the invention and of atorvastatin following single oral administration to SD rats (dose equimolar to atorvastatin 20 mg/kg)
  • Table 3 reports a summary of pharmacokinetic parameters of 2-hydroxy metabolite of atorvastatin formed from the compounds of the invention following single oral administration to SD rats (dose equimolar to atorvastatin 20 mg/kg)

Landscapes

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

Abstract

Dinitrate statins having improved pharmacological activity are described. They can be employed for treating and/or preventing several diseases, in particular acute coronary syndromes, neurodegenerative disorders as well as for reducing cholesterol levels.

Description

STATIN DERIVATIVES
The present invention relates to new statin nitroderivatives . More particularly, the present invention relates to dinitrate statins, pharmaceutical compositions containing them and their use as cholesterol-reducing drugs, as drugs having antioxidant, antithrombotic and anti-inflammatory activity, effects on endothelial function, immunosuppressive properties and for treating and/or preventing acute coronary syndromes, stroke, neurodegenerative disorders, such as Alzheimer's and Parkinson's disease as well as autoimmune diseases, such as multiple sclerosis.
Statins is a class of compounds which comprises as main components lovastatin, simvastatin, pravastatin, mevastatin, fluvastatin, atorvastatin, rosuvastatin and cerivastatin (rivastatin) . Statins possess a side group that is structurally similar to HMG-CoA ( 3-hydroxy-3- methylglutaryl coenzyme A) . Fluvastatin, atorvastatin, rosuvastatin and cerivastatin are entirely synthetic compounds containing a heptanoic acid side chain, the remainders being fungal metabolites.
It is known that the various statins are inhibitors of HMG-CoA reductase, an enzyme which catalyses an early, rate-limiting step in cholesterol biosynthesis, reduce triglyceride levels and are also indicated for raising HDL- C levels (P.O. Bonetti et al., European Heart Journal (2003) 24, 225-248) .
WO 2004/105754 discloses mononitrate statins having improved pharmacological activity and enhanced tolerability and their use for treating and/or preventing several diseases . It has been so surprisingly found that the dinitrate compounds of the invention provide enhanced nitric oxide (NO) release over mononitrate analogs (WO 2004/105754). A consideration of stoichiometry leads to an expectation of a doubling of nitrite levels. Compounds of the invention when tested in vitro in a tissue-based measure of vessel relaxation, in rabbit aortic slices, show significant enhancement in EC50 (molar concentration of compound which produces 50% of the maximum possible response for that compound) compared to mononitrates which are greater than two fold (the increase expected based on the stoichiometric relationship) .
It was object of the present invention to provide new derivatives of statins having an improved pharmacological activity compared to mononitrate derivatives.
Object of the present invention are, therefore, statin nitroderivatives of general formula (I) or pharmaceutically acceptable salts or stereoisomers thereof
Figure imgf000003_0001
(I)
wherein R is selected from the group consisting of (Ila) (Ilf) :
Figure imgf000003_0002
(Ila) (lib)
Figure imgf000004_0001
( H e ) (lid)
Figure imgf000004_0002
( H e ) (Hf)
w is an integer selected between 0 and 1 ;
R1 and R2 can be equal or different and are independently selected from the group consisting of hydrogen and C1-C4 alkyl ;
Y is selected from the group consisting of:
A) [R3-0]p-R4-CH(ON02) - (R5)n-CH(ON02) - [0-R6]pl-R7
B) R4-CH (ON02) -R3-0-R5-CH (ON02) -R7
C) (R8) n-R9- [R3-0] P-R4-CH (ON02) - (R5) ni-CH (ON02) - [O-R6] pl-R7 wherein
R3, R4, R5, R6 and R8 are independently a straight or branched C1-C10 alkylene;
n is an integer from 0 to 1 ;
nl is an integer from 0 to 1 ;
p is an integer from 0 to 4 ;
pi is an integer from 0 to 4 ;
R7 is selected from the group consisting of hydrogen and
C^-Cg alkyl;
R9 is a saturated, unsaturated or aromatic heterocyclic 5 or 6 members ring, containing one or more heteroatoms selected from nitrogen, oxygen or sulphur. The term "Ci-Cio alkylene" as used herein refers to branched or straight chain Ci-Cio hydrocarbon, such as methylene, ethylene, propylene, isopropylene, n-butylene, pentylene, n-hexylene and the like.
The term "Ci-Cio alkyl" as used herein refers to branched or straight chain alkyl groups comprising one to ten carbon atoms, including methyl, ethyl, n-propyl, isopropyl, n- butyl, isobutyl, t-butyl, pentyl, hexyl, octyl and the like .
The term "heterocyclic" as used herein refers to saturated, unsaturated or aromatic 5 or 6 members ring, containing one or more heteroatoms selected from nitrogen, oxygen, sulphur, such as for example pyridine, pyrazine, pyrimidine, pyrrolidine, morpholine, imidazole and the like.
Preferred compounds according to the invention are the compounds of general formula (I) or pharmaceutically acceptable salts or stereoisomers thereof as defined above wherein R is the group (lid) or (lie) .
An embodiment of the invention relates to the compounds of general formula (I) or pharmaceutically acceptable salts or stereoisomers thereof as defined above wherein w is 1.
Preferably R1 and R2 which can be equal or different are independently selected from the group consisting of hydrogen and C¾.
Another preferred embodiment of the invention relates to the compounds of general formula (I) or pharmaceutically acceptable salts or stereoisomers thereof as defined above wherein w is 0.
Another preferred embodiment of the invention relates the compounds of general formula (I) or pharmaceutically acceptable salts or stereoisomers thereof as defined above wherein Y of the group A) is selected from the group consisting of:
Figure imgf000006_0001
Figure imgf000006_0002
Figure imgf000006_0003
Figure imgf000007_0001
5
Figure imgf000007_0002
Figure imgf000008_0001
Figure imgf000008_0002
Another embodiment of the invention relates to the compounds of Formula (I) or pharmaceutically acceptable salts or stereoisomers thereof as defined above wherein
Y of the roup B) is selected from the group consisting of:
Figure imgf000008_0003
Figure imgf000009_0001
Another embodiment of the invention relates to the compounds of Formula (I) or pharmaceutically acceptable salts or stereoisomers thereof as defined above wherein Y of the group C) is selected from the group consisting of:
Figure imgf000009_0002
Another embodiment of the invention relates to the compounds of Formula (I) or pharmaceutically acceptable salts or stereoisomers thereof as defined above selected from the roup consisting of:
Figure imgf000010_0001
(11) (12)
Figure imgf000011_0001
Figure imgf000012_0001
Figure imgf000013_0001

Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000017_0002
Figure imgf000017_0003
Figure imgf000017_0004
(87)
Another aspect of the present invention includes the use of the compounds of formula (I) in combination with at least a compound used to treat cardiovascular diseases selected from the group consisting of: ACE inhibitors, renin inhibitors, angiotensin II receptor antagonists, beta-adrenergic blockers, calcium channel blockers, antithrombotics such as aspirin, nitrosated ACE inhibitors, nitrosated renin inhibitors, nitrosated angiotensin II receptor antagonists, nitrosated beta-adrenergic blockers and nitrosated aspirin. Suitable ACE inhibitors, angiotensin II receptor antagonists, beta-adrenergic blockers, calcium channel blockers, antithrombotics are described in the literature such as The Merck Index (13th edition) .
Suitable nitrosated compounds are disclosed in WO 98/21193, WO 2005/011646, WO 97/16405, WO 2004/106300, WO 2004/110432 and WO 2007/045551.
The administration of the compounds above reported can be carried out simultaneously or successively.
The present invention also provides pharmaceutical kits comprising one or more containers filled with one or more of the compounds and/or compositions of the present invention and one or more of the compounds used to treat cardiovascular diseases reported above.
As stated above, the invention includes also the pharmaceutically acceptable salts of the compounds of formula (I) and stereoisomers thereof.
The compounds according to the present invention, when they contain in the molecule one salifiable nitrogen atom, can be transformed into the corresponding salts by reaction in an organic solvent such as acetonitrile, tetrahydrofuran with the corresponding organic or inorganic acids.
Examples of organic acids are: oxalic, tartaric, maleic, succinic, citric acids. Examples of inorganic acids are: nitric, hydrochloric, sulphuric, phosphoric acids. Salts with nitric acid are preferred.
The compounds of the invention which have one or more asymmetric carbon atoms can exist as optically pure enantiomers, pure diastereomers , enantiomers mixtures, diastereomers mixtures, enantiomer racemic mixtures, racemates or racemate mixtures. Within the object of the invention are also all the possible isomers, stereoisomers and their mixtures of the compounds of formula (I) . As mentioned above, object of the present invention are also pharmaceutical compositions containing at least a compound of the present invention of formula (I) together with non toxic adiuvants and/or carriers usually employed in the pharmaceutical field.
The daily dose of active ingredient that should be administered can be a single dose or it can be an effective amount divided into several smaller doses that are to be administered throughout the day. Usually, total daily dose may be in amounts preferably from 1 to 500 mg. The dosage regimen and administration frequency for treating the mentioned diseases with the compound of the invention and/or with the pharmaceutical compositions of the present invention will be selected in accordance with a variety of factors, including for example age, body weight, sex and medical condition of the patient as well as severity of the disease, route of administration, pharmacological considerations and eventual concomitant therapy with other drugs. In some instances, dosage levels below or above the aforesaid range and/or more frequent may be adequate, and this logically will be within the judgment of the physician and will depend on the disease state.
The compounds of the invention may be administered orally, parenterally, rectally or topically, by inhalation or aerosol, in formulations eventually containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles as desired. Topical administration may also involve the use of transdermal administration such as transdermal patches or iontophoresis devices. The term "parenteral" as used herein, includes subcutaneous injections, intravenous, intramuscular, intrasternal injection or infusion techniques. Injectable preparations, for example sterile injectable aqueous or oleaginous suspensions may be formulated according to known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent. Among the acceptable vehicles and solvents are water, Ringer's solution and isotonic sodium chloride. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono or diglycerides , in addition fatty acids such as oleic acid find use in the preparation of injectables.
Suppositories for rectal administration of the drug can be prepared by mixing the active ingredient with a suitable non-irritating excipient, such as cocoa butter and polyethylene glycols.
Solid dosage forms for oral administration may include capsules, tablets, pills, powders, granules and gels. In such solid dosage forms, the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as in normal practice, additional substances other than inert diluents, e.g. lubricating agents such as magnesium stearate. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. Tablets and pills can additionally be prepared with enteric coatings.
Liquid dosage forms for oral administration may include pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs containing inert diluents commonly used in the art, such as water. Such compositions may also comprise adjuvants, such as wetting agents, emulsifying and suspending agents, and sweetening, flavouring and the like.
As previously reported, in addition to lipid lowering properties the new statin nitroderivatives possess enhanced anti-inflammatory, antiplatelet and antithrombotic effects as compared to native statins. Moreover, they can be effective also in the other pathologies such as acute coronary syndromes, stroke, peripheral vascular diseases such as peripheral ischemia, all disorders associated with endothelial dysfunctions such as vascular complications in diabetic patients and atherosclerosis, neurodegenerative diseases such as Alzheimer's disease (AD) and Parkinson's disease (PD) , autoimmune diseases such as multiple sclerosis .
Synthesis procedure
1) The compounds of formula (I)
Figure imgf000021_0001
(I) wherein w, R, R1, R2 and Y are as above defined, can be obtained by a process comprising reacting a compound of formula (III)
Figure imgf000021_0002
(III)
wherein M is an alkali metal or alkaline earth metal such as sodium or calcium and R is as above defined;
with a compound of formula (XI)
Figure imgf000022_0001
(XI)
wherein W, R , R and Y are as above defined and Hal is CI, Br or I
The reaction is carried out in an aprotic polar/non polar solvent such as DMF, THF, toluene at a temperature ranging from -20°C to 60°C; The reaction is completed within a time ranges from 30 minutes to 24 hours.
Compounds (XI) can be prepared from the appropriate
compound of formula (X) :
Y-OH
(X)
wherein Y is as above defined, following procedures well known in the literature.
Compounds (X) are known in the literature or can be prepared by known compounds by methods well known in the art .
2) The compounds of formula (I)
Figure imgf000022_0002
(I) wherein w is 0, R, R , R and Y are as above defined, can be obtained
2.1) reacting a compound of formula (XII)
P P
Figure imgf000022_0003
(XII) wherein R is as above defined, P is a hydroxyl protecting group of formula (Xllla) :
Figure imgf000023_0001
(XHIa)
or the P groups taken together form the protecting group of formula (Xlllb) :
Figure imgf000023_0002
(XHIb)
with a compound of formula (X) Y-OH
wherein Y is as above defined,
the reaction may be carried out in presence in the presence of a condensing agent such as dicyclohexylcarbodiimide (DCC) , N' - ( 3-dimethylaminopropyl ) -N-ethylcarbodiimide hydrochloride (EDAC) , N, N' -carbonyldiimidazole (CDI), optionally in the presence of a base, for example DMAP . The reaction is carried out in an inert organic solvent dry such as N, N' -dimethylformamide, tetrahydrofuran, benzene, toluene, dioxane, a polyhalogenated aliphatic hydrocarbon at a temperature from -20°C and 50°C. The reaction is completed within a time range from 30 minutes to 36 hours; 2.2) deprotecting the compounds obtained in the step 2.1) . Fluoride ion is the preferred method for removing silyl ether protecting group. Triethylamine trihydrofluoride or tetrabutylamonium fluoride and acetic acid, in an aprotic polar/non polar solvent such as THF, at temperature ranging from -10°C to 30°C are the preferred method for removing the silyl ether group.
Compounds of formula (XII) can be prepared by hydrolysis of compounds of formula (XIV) ? P
0 0 0
o-x
(XIV)
wherein R and P are as above defined and X is selected from the formulas (IXa)-(IXb)
Figure imgf000024_0001
(IXa) (IXb) when X is (IXa) the deprotecting of the carboxylic function may be carried out by hydrogenation in presence of Pd/C in an aprotic polar/non polar solvent such as THF at temperature ranging from 0°C to 30 °C and a pressure ranging from 1 to 5 bar;
when X is (IXb) the deprotecting of the carboxylic function may be carried out with palladium tetrakis, in presence of dimedone and triphenylphosphine or in presence of morpholine, in an aprotic polar/non polar solvent such as THF, DMF or CH2CI2, at temperature ranging from -10 °C to
30°C.
Compounds of formula (XIV) can be prepared by reaction of compound of formula (IV)
Figure imgf000024_0002
(IV) wherein R and X are as above defined,
- when P is (Xllla) with tertbutyldimethylchlorosilane and imidazole in in an aprotic polar/non polar solvent such as THF, DMF or CH2CI2, at temperature ranging from -20 °C to 60°C; - when P groups taken together form the group (Xlllb) with 1, 3-dichloro-l, 1, 3, 3-tetraisopropyldisiloxane in presence of imidazole in an aprotic polar/non polar solvent such as THF, DMF or CH2CI2, at temperature ranging from -20 °C to
60°C.
Compounds of formula (IV) can be prepared by reaction compound of formula (III)
Figure imgf000025_0001
(III)
wherein R is as above defined and M is an alkali metal or alkaline earth metal such as sodium or calcium,
with a compound of formula X-Hal wherein X is as above defined and Hal is selected from Br or CI,
the reaction may be carried out in an aprotic polar/non polar solvent such as THF, DMF or CH2CI2, at temperature ranging from -80°C to 60°C.
In another embodiment of the invention, there is provided a process for the preparation of compounds of formula (I)
Figure imgf000025_0002
(I)
wherein w is 0, R, R1, R2 and Y are as above defined;
said process, as depicted in Scheme 1
Figure imgf000026_0001
(V) (vi :
Figure imgf000026_0002
(VII) (VIII)
Scheme 1 comprising
1) reacting compounds of formula (III), wherein M is an alkali metal or alkaline earth metal such as sodium or calcium and R is as above defined;
with a compound of formula X-Hal wherein X is selected from the formulas (IXa)-(IXb)
Figure imgf000026_0003
(IXa) (IXb)
and Hal is selected from Br or CI,
in an aprotic polar/non polar solvent such as THF, DMF or CH2CI2, at temperature ranging from -80°C to 60°C;
2) reacting a compound of formula (IV) wherein R and X are as above defined, with 1, 3-dichloro-l, 1, 3, 3- tetraisopropyldisiloxane in presence of imidazole in an aprotic polar/non polar solvent such as THF, DMF or CH2CI2, at temperature ranging from -80°C to 60°C; 3) deprotecting of the carboxylic function of compound of formula (V) wherein R and X are as above defined:
when X is (IXa) by catalytic hydrogenation in presence of Pd/C in an aprotic polar/non polar solvent such as THF at room temperature and pressure;
when X is (IXb) with palladium tetrakis, in presence of dimedone and triphenylphosphine or in presence of morpholine, in an aprotic polar/non polar solvent such as THF, DMF or CH2CI2, at temperature ranging from -10 °C to 30°C;
4) reacting a compound of formula (VI) wherein R is as above defined with a compound of formula (X)
HO-Y
(X)
wherein Y is as above defined in the presence of a condensing agent such as dicyclohexylcarbodiimide (DCC) , N' - ( 3-dimethylaminopropyl ) -N-ethylcarbodiimide
hydrochloride (EDAC) , N, N' -carbonyldiimidazole (CDI), optionally in the presence of a base, for example DMAP . The reaction is carried out in an inert organic solvent dry such as N, N' -dimethylformamide, tetrahydrofuran, benzene, toluene, dioxane, a polyhalogenated aliphatic hydrocarbon at a temperature from -20°C and 50°C. The reaction is completed within a time range from 30 minutes to 36 hours; 5) deprotecting of the diol function of compound of formula (VII) wherein R is as above defined in presence of fluoride salt of alkylamine such as triethylamine trihydrofluoride or tetrabutylamonium fluoride and acetic acid, in an aprotic polar/non polar solvent such as THF, at temperature ranging from -10°C to 30°C. Another embodiment of the invention relates to compounds of formula (VI) wherein R is selected from the group consisting of (Ila)-(IIf). Another embodiment of the invention relates to compounds of formula (VI) wherein R is (lid) .
Another embodiment of the invention relates to compounds of formula (VI) wherein R is (He) .
The following examples are to further illustrate the invention without limiting it.
Example 1
Synthesis of (3R, 5R) -3, 3-dimethyl-5, 6-bis (nitrooxy) hexyl 7- (2- (4-fluorophenyl) -5-isopropyl-3-phenyl-4- (phenyl
carbamoyl) -lH-pyrrol-l-yl) -3, 5-dihydroxyheptanoate
(Compound ( 1 ) )
Figure imgf000028_0001
(1)
A) Methyl 2- ( (4R, 6R) -6- (2- (2- (4-fluorophenyl) -5-isopropyl- 3-phenyl-4- (phenylcarbamoyl ) -lH-pyrrol-l-yl) ethyl) -2, 2- dimethyl-1, 3-dioxan-4- l) acetate
Figure imgf000028_0002
To a solution of tert-butyl 2- ( (4R, 6R) -6- (2- (2- (4- fluorophenyl ) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl) -1H- pyrrol-l-yl) ethyl) -2, 2-dimethyl-l, 3-dioxan-4-yl) acetate ( 2.0 g, 3.05 mmol) in MeOH (20 mL) , NaOH (0.25 g, 4.58 mmol) was added. The solution was stirred at reflux for 7 hours. The solution was concentrated under vacuum , diluted with an aqueous solution of 5 % Na¾P04 (100 mL) and extracted with AcOEt ( 3x200 mL) . The combined organic layers were washed with brine (2x100 mL) , dried over Na2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, gradient method calculated from TLC: n-hexane/EtOAc 8/2, Rf product: 0.15) affording the title compound (0.7 g, yield 37%).
B) 2- ( (4R, 6R) -6- (2- (2- (4-fluorophenyl) -5-isopropyl-3-phenyl -4- (phenylcarbamoyl ) -lH-pyrrol-l-yl) ethyl) -2, 2-dimethyl- 1, 3-dioxan-4-yl) acetic acid
Figure imgf000029_0001
To a solution of compound A) ( 0.74 g, 1.04 mmol) in THF (7 mL) , an aqueous solution of IN KOH (1.26 mL, 1.26 mmol) was added. The solution was stirred at reflux for 6 hours. The solution was concentrated under vacuum, diluted with an aqueous solution of 5 % Naf^PC^ (70 mL) and extracted with AcOEt ( 3x100 mL) . The combined organic layers were washed with brine (2x100 mL) , dried over Na2S04 and concentrated under reduced pressure affording the title compound (0.61 g, yield 98%) .
C) 3, 3-dimethyl-5, 6-bis (nitrooxy) hexyl 2- ( (4R, 6R) -6- (2- (2- (4-fluorophenyl) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl) - lH-pyrrol-l-yl) ethyl) -2, 2-dimethyl-l , 3-dioxan-4-yl) acetate
Figure imgf000030_0001
To a solution of compound B) (0.54 g, 0.9 mmol) in CH2CI2 (10 mL) , 6-hydroxy-4 , 4-dimethylhexane-l , 2-diyl dinitrate (0.35 g, 1.4 mmol) was added. The solution was cooled at 0°C and DMAP (22 mg, 0.18 mmol) and EDAC (0.27 g, 1.4 mmol) were added. The reaction was stirred at room temperature for 4 hours. The solution was diluted with CH2CI2 (30 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x60 mL) . The organic layer was washed with ¾0 (4x40 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, gradient method calculated from TLC: n-hexane/EtOAc 7/3, Rf product: 0.46) affording the title compound (0.620 g, yield 93%) .
D) (3R, 5R) -3, 3-dimethyl-5, 6-bis (nitrooxy) hexyl 7- (2- (4- fluorophenyl ) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -1H- pyrrol-l-yl) -3, 5-dihydroxyheptanoate (Compound (1))
To a suspension of p-toluensulfonic acid silica supported (0.44 g, 0.28 mmol) in methylene chloride (5 mL) cooled at 0°C, compound C) (0.21 g, 0.28 mmol) in CH2CI2 (10 mL) was added dropwise. The reaction was stirred at 0°C for 1 hour. The solution was filtered and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, Cartridge column FLASH 12+M™ KP-Sil, gradient method calculated from TLC: n-hexane/EtOAc 1/1, Rf product: 0.31) affording the compound (1) (0.03 g, yield 15%) . NMR (CDCI3) : 7.26-6.94 (14H, m) , 6.87 (1H, s), 5.46 (1H, m) , 4.73 (1H, dd) , 4.43 (1H, dd) , 4.24-4.05 (3H, m) , 3.97(1H, m) , 3.76 (1H, m) , 3.59 (2H, m) , 3.45 (1H, s), 2.40 (2H, d) , 1.85-1.41 (6H, m) , 1.55 (6H, m) , 1.27 (2H, t) , 1.02 (6H,s)
Example 2
Synthesis of (3R, 5R) -2- (2, 3-bis (nitrooxy) propoxy) ethyl 7- (2- (4-fluorophenyl) -5-isopropyl-3-phenyl-4- (phenyl
carbamoyl) -lH-pyrrol-l-yl) -3, 5-dihydroxyheptanoate
(Compound (2))
Figure imgf000031_0001
(2)
E) (3R, 5R) -allyl-7- (2- (4-fluorophenyl) -5-isopropyl-3-phenyl -4- (phenylcarbamoyl ) -lH-pyrrol-l-yl) -3, 5- dihydroxyheptanoate
Figure imgf000031_0002
To a solution of atorvastatin calcium salt (5.0 g, 4.33 mmol) in DMF (75 mL) , was added allyl bromine (2.1 g, 17.30 mmol) . The solution was stirred at room temperature for 72 hours. The solution was diluted with ¾0 (200mL) and extracted with EtOAc (3 x 100 mL) . The combined organic layers were washed with brine (2 x 100 mL) , dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 340 g, gradient method calculated from TLC: n-hexane/EtOAc 1/1, Rf product: 0.42) affording the title compound (2.60 g, 50%).
F) allyl 2- ( ( 6R, 8R) -8- (2- (2- (4-fluorophenyl) -5-isopropyl-3- phenyl-4- (phenylcarbamoyl ) -lH-pyrrol-l-yl) ethyl) -2,2,4,4- tetraisopropyl-1 , 3,5,2, 4-trioxadisilocan-6- l) acetate
Figure imgf000032_0001
To a solution of compound E) (3.00 g; 5.01 mmol) in DMF (30 mL) , was added imidazole (1.91 g; 28.05 mmol). A solution of 1, 3-dichloro-l, 1, 3, 3-tetraisopropyldisiloxane (3.20 mL; 10.00 mmol) in DMF (5mL) was then added dropwise. The reaction was stirred at room temperature for 20 hours. The solution was diluted with an aqueous solution of 5 % NaH2P04 (50 mL) and extracted with AcOEt (3 x 25 mL) . The combined organic layers were washed with ¾0 (4 X 20 mL) and brine, then dried over Na2SC>4 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 340 g, eluent: gradient method calculated from TLC: n- hexane/EtOAc 9/1, Rf product: 0.30) affording the title compound (3.04 g, 72%) .
G) 2- ( (6R, 8R) -8- (2- (2- (4-fluorophenyl) -5-isopropyl-3- phenyl-4- (phenylcarbamoyl) -lH-pyrrol-l-yl) ethyl) -2,2,4,4- tetraisopropyl-1 , 3,5,2, 4-trioxadisilocan-6-yl) acetic acid
Figure imgf000033_0001
To a solution of compound F) (0.83 g; 0.99 mmol) in dry CH2CI2 (20 mL) , was added dimedone (0.21 g; 1.48 mmol), followed by triphenylphosphine (0.39 g; 1.48 mmol) and palladium tetrakis (0.05 g; 0.04 mmol) . The solution was stirred for 3 hours and then concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 340 g, eluent: gradient method calculated from TLC: n-hexane/EtOAc 66/33, Rf product: 0.38) affording the title compound (4.67 g, 93%) .
1H-NMR (DMSO-de) : 9.80 (1H, m) , 7.51 (2H, m) , 7.22 (6H, m) , 7.02 (6H, m) , 4.28 (1H, m) , 4.08-3.77 (3H, m) , 3.19 (1H, m) , 2.32 (2H, m) , 1.81-1.18 (10H, m) , 1.18-0.64 (28H) .
H) 2- (2, 3-dinitrooxypropoxy) ethyl 2- ( ( 6R, 8R) -8- (2- (2- (4- fluorophenyl ) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -1H- pyrrol-l-yl) ethyl) -2, 2, 4, 4-tetraisopropyl-l, 3, 5, 2, 4- trioxadisilocan- 6- l ) acetate
Figure imgf000033_0002
To a solution of compound G) (0.64 g, 0.79 mmol) in CH2CI2 (8 mL) , 2-ethoxy-3- (2-nitrooxyethoxy) propyl nitrate (0.2 g, 0.79 mmol) was added. The solution was cooled at 0°C and DMAP (19 mg, 0.16 mmol) and EDAC (0.23 g, 1.2 mmol) were added. The reaction was stirred at room temperature for 6 hours. The solution was diluted with CH2CI2 (20 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x50 mL) . The organic layer was washed with ¾0 (4x30 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 50 g, gradient method calculated from TLC: n-hexane/EtOAc 7/3, Rf product: 0.45) affording the title compound (0.480 g, yield 63%).
I) (3R, 5R) -2- (2, 3-bis (nitrooxy) propoxy) ethyl 7- (2- (4- fluorophenyl ) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -1H- pyrrol-l-yl) -3, 5-dihydroxyheptanoate (Compound (2))
To a solution of compound H) (0.43 g, 0.42 mmol) in dry THF (8 mL) , cooled at 0°C, acetic acid (0.18 mL, 2.86 mmol) and a 1M THF solution of TBAF (2.6 mL, 2.55 mmol) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with AcOEt (50 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x30 mL) . The organic layer was washed with ¾0 (4x20 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 50 g, eluent: gradient method calculated from TLC: n-hexane/EtOAc 4/6, Rf product: 0.25) affording the Compound (2) (0.2 g, 51%) .
1H-NMR (DMSO-de) : 9.78 (1H, s), 7.51 (2H, d) , 7.21 (6H, m) , 7.01 (6H, m) , 5.58 (1H, m) , 4.91 (1H, dd) , 4.81-4.51 (3H, m) , 4.11 (2H, t) , 3.98-3.58 (7H, m) , 3.51 (1H, m) , 3.23 (1H, m) , 2.44-2.21 (2H, m) , 1.67-1.45 (4H, m) , 1.37 (6H, m) . Example 3
Synthesis of ( 3R, 5R) -(( 4R, 5R) -3-methyl-4 , 5-bis (nitrooxy) hexyl) -7- (2- (4-fluorophenyl) -5-isopropyl-3-phenyl-4- (phenyl carbamoyl) -lH-pyrrol-l-yl) -3, 5-dihydroxy heptanoate
(Compound (3) )
Figure imgf000035_0001
(3)
J) (3R, 5R) -benzyl-7- (2- (4-fluorophenyl) -5-isopropyl-3-phenyl -4- (phenylcarbamoyl ) -lH-pyrrol-l-yl) -3, 5-dihydroxy
heptanoate
Figure imgf000035_0002
To a solution of atorvastatin calcium salt (5.0 g, 4.33 mmol) in DMF (25 mL) , was added benzyl bromine (1.54 g, 9.0 mmol) . The solution was stirred at room temperature for 48 hours. The solution was diluted with ¾0 (70 mL) and extracted with EtOAc (3 x 30 mL) . The combined organic layers were washed with brine (2 x 20 mL) , dried over a2S04 and concentrated under reduced pressure. The crude (4.5 g) was used in the next step without further purification.
K) Benzyl 2- ( ( 6R, 8R) -8- (2- (2- (4-fluorophenyl) -5-isopropyl- 3-phenyl-4- (phenylcarbamoyl) -lH-pyrrol-l-yl) ethyl) -2,2,4,4- tetraisopropyl-1 , 3,5,2, 4-trioxadisilocan-6-yl) acetate
Figure imgf000036_0001
To a solution of compound J) (2.5 g; 3.8 mmol) in DMF (20 mL) , was added imidazole (1.55 g; 22.8 mmol). A solution of 1, 3-dichloro-l, 1, 3, 3-tetraisopropyldisiloxane (2.3 mL; 7.32 mmol) in DMF (5mL) was then added dropwise. The reaction was stirred at room temperature for 24 hours. The solution was diluted with an aqueous solution of 5 % Na¾P04 (60 mL) and extracted with AcOEt (3 x 30 mL) . The combined organic layers were washed with ¾0 (4 X 20 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 340 g, eluent: gradient method calculated from TLC: n-hexane/EtOAc 9/1, Rf product: 0.30) affording the title compound (2.8 g, 85%).
G) 2- ( (6R, 8R) -8- (2- (2- (4-fluorophenyl) -5-isopropyl-3- phenyl-4- (phenylcarbamoyl ) -lH-pyrrol-l-yl) ethyl) -2,2,4,4- tetraisopropyl-1 , 3,5,2, 4-trioxadisilocan-6-yl) acetic acid
Figure imgf000036_0002
To a solution of compound K) (2.5 g; 3.35 mmol) in THF (30 mL) , Pd/C (0.23 g) was added. The solution was treated with hydrogen under the pressure of 4 bar and at room temperature for 5.5 hours. The reaction was filtered and concentrated under reduced pressure. The residue was (4.5 g) was crystallized from n-hexane (20 mL) affording the title compound (2.25 g, yield 84%) L) (4R, 5R) -4, 5-dinitrooxy-3-methylhexyl 2- ( ( 6R, 8R) -8- (2- (2- (4-fluorophenyl) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) - lH-pyrrol-l-yl) ethyl) -2, 2, 4, 4-tetraisopropyl-l, 3, 5, 2, 4- trioxadisilocan- 6- l ) acetate
Figure imgf000037_0001
To a solution of compound G) (1.15 g, 1.43 mmol) in CH2CI2 (15 mL) , (2R, 3R) -6-hydroxy-4-methylhexane-2, 3-diyl-di nitrate (0.40 g, 1.67 mmol) was added. The solution was cooled at 0°C and DMAP (41 mg, 0.33 mmol) and EDAC (0.48 g, 2.52 mmol) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with CH2CI2 (30 mL) and washed with an aqueous solution of 5 % aH2 P04 (2x50 mL) . The organic layer was washed with ¾0 (4x30 mL) and brine, then dried over a2 S 04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, gradient method calculated from TLC: n-hexane/EtOAc 8/2, Rf product: 0.33) affording the title compound (1.15 g, yield 79%). M) (3R, 5R) - ( (4R, 5R) -3-methyl-4, 5-bis (nitrooxy) hexyl) 7- (2- (4-fluorophenyl) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl) - lH-pyrrol-l-yl) -3, 5-dihydroxyheptanoate (Compound (3)) To a solution of compound L) (1.04 g, 1.02 mmol) in dry THF (16 mL) , cooled at 0°C, acetic acid (0.41 mL, 6.72 mmol) and a 1M THF solution of TBAF (6.1 mL, 6.11 mmol) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with AcOEt (100 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x70 mL) . The organic layer was washed with ¾0 (4x70 mL) and brine, then dried over Na2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, eluent: gradient method calculated from TLC: n-hexane/EtOAc 1/1, Rf product: 0.25) affording the compound (3) (0.31 g, 40%). 1H-NMR (DMSO-de) : 9.78 (1H, s), 7.50 (2H, d) , 7.19 (6H, m) , 6.99 (6H, m) , 5.41 (2H, m) , 4.68 (2H, dd) , 4.19-3.67 (5H, m) , 3.51 (1H, m) , 3.23 (1H, m) , 2.31 (2H, m) , 1.80-1.39 (10H, m) , 1.38 (6H, m) , 0.96 (3H, dd) .
Example 4
Synthesis of ( 3R, 5R) -2- ( ( S ) -2 , 3-bis (nitrooxy) propoxy) ethyl 7- (2- (4-fluorophenyl) -5-isopropyl-3-phenyl-4- (phenyl carbamoyl) -lH-pyrrol-l-yl) -3, 5-dihydroxyheptanoate
(Compound (2 ) )
Figure imgf000038_0001
(2)
N) 2- ( (R) -2, 3-dinitrooxypropoxy) ethyl-2- ( (6R, 8R) -8- (2- (2 (4-fluorophenyl) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) - lH-pyrrol-l-yl) ethyl) -2, 2, 4, 4-tetraisopropyl-l, 3, 5, 2, 4- trioxadisilocan- 6-yl ) acetate
Figure imgf000039_0001
To a solution of compound G) (0.73 g, 0.91 mmol) in CH2CI2 (15 mL) , (R) 1 , 2-dinitrooxy-3- (2-hydroxyethoxy) propane (0.23 g, 1.02 mmol) was added. The solution was cooled at 0°C and DMAP (20 mg, 0.15 mmol) and EDAC (0.22 g, 1.15 mmol) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with CH2CI2 (40 mL) and washed with an aqueous solution of 5 % aH2P04 (2x50 mL) . The organic layer was washed with ¾0 (4x30 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, gradient method calculated from TLC : n-hexane/EtOAc 75/25, Rf product: 0.33) affording the title compound (0.76 g, yield 83%) .
0) (3R, 5R) -2- ( (S) -2, 3-bis (nitrooxy) propoxy) ethyl 7- (2- (4- fluorophenyl ) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -1H- pyrrol-l-yl) -3, 5-dihydroxyheptanoate (Compound (2))
To a solution of compound N) (0.75 g, 0.74 mmol) in dry THF (10 mL) , cooled at 0°C, acetic acid (0.28 mL, 4.55 mmol) and a 1M THF solution of TBAF (4.2 mL, 4.2 mmol) were added. The reaction was stirred at room temperature for 6 hours. The solution was diluted with AcOEt (100 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x70 mL) . The organic layer was washed with ¾0 (4x70 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, eluent: gradient from n-hexane/EtOAc 75/25 to n-hexane/EtOAc 20/80 during 1.4 L) affording the compound (2) (0.43 g, 75%) .
1H-NMR (DMSO-de) : 9.78 (1H, s), 7.51 (2H, d) , 7.21 (6H, m) , 7.01 (6H, m) , 5.58 (1H, m) , 4.93 (1H, dd) , 4.82-4.71 (2H, m) , 4.62 (1H, d) , 4.11 (2H, t) , 3.98-3.58 (7H, m) , 3.51 (1H, m) , 3.23 (1 H, m) , 2.44-2.21 (2H, m) , 1.67-1.45 (4H, m) , 1.37 (6H, m) .
Example 5
Synthesis of (3R, 5R) -( (R) -5, 6-bis (nitrooxy) hexyl) -7- (2- (4- fluorophenyl ) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -1H- pyrrol-l-yl) -3 5-dihydroxyheptanoate (Compound (4))
Figure imgf000040_0001
(4)
P) (R) -5, 6-dinitrooxyhexyl-2- ( (6R, 8R) -2,2,4, 4-tetraisopro pyl-8- (2- (2-isopropyl-4 , 5-diphenyl-3- (phenylcarbamoyl) -1H- pyrrol-l-yl) eth l) -1,3,5,2, 4-trioxadisilocan-6-yl) acetate
Figure imgf000040_0002
To a solution of compound G) (2.5 g, 3.16 mmol) in CH2CI2 (50 mL) , (R) -1, 2-dinitrooxyhexan-6-ol (0.8 g, 3.2 mmol) was added. The solution was cooled at 0°C and DMAP (154 mg, 1.2 mmol) and EDAC (1.21 g, 6.32 mmol) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with CH2CI2 (80 mL) and washed with an aqueous solution of 5 % Naf^PC^ (2x50 mL) . The organic layer was washed with ¾0 (4x30 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, gradient from n-hexane to n- hexane/EtOAc 60/40 during 1.7 L) affording the title compound (2.93 g, yield 92%) .
Q) (3R, 5R) - ( (R) -5, 6-bis (nitrooxy) hexyl) 7- (2- (4-fluoro phenyl) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -lH-pyrrol- 1-yl) -3, 5-dihydroxyheptanoate (Compound (4))
To a solution of compound P) (2.93 g, 2.91 mmol) in dry THF (45 mL) , cooled at 0°C, acetic acid (1.18 mL, 19.2 mmol) and a 1M THF solution of TBAF (17.5 mL, 17.5 mmol) were added. The reaction was stirred at room temperature for 24 hours. The solution was diluted with AcOEt (150 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x100 mL) . The organic layer was washed with ¾0 (4x100 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, eluent: gradient from n-hexane/EtOAc 9/1 to n-hexane/EtOAc 20/80 during 1.4 L) affording the compound (4) (1.86 g, 70%) .
1H-NMR (DMSO-de) : 9.74 (1H, s), 7.50 (2H, d) , 7.19 (6H, m) , 6.99 (6H, m) , 5.41 (1H, m) , 4.92 (1H, dd) , 4.70 (2H, m) , 4.59 (1H, d) , 3.98-3.66 (5H, m) , 3.51 (1H, m) , 3.23 (1H, m) , 2.36 (1H, dd) , 2.25 (1H, dd) , 1.83-1.39 (6H, m) , 1.38 (6H, m) .
Example 6
Synthesis of ( 3R, 5R) -3- (( S ) -2 , 3-bis (nitrooxy) propoxy) propyl 7- (2- (4-fluorophenyl) -5-isopropyl-3-phenyl-4- (phenyl carbamoyl) -lH-pyrrol-l-yl) -3, 5-dihydroxyheptanoate
(Compound (5) )
Figure imgf000042_0001
(5)
R) 3- ( (R) -2, 3-dinitrooxypropoxy) propyl 2- ( ( 6R, 8R) -2 , 2 , 4 , 4- tetraisopropyl-8- (2- (2-isopropyl-4 , 5-diphenyl-3- (phenyl carbamoyl) -lH-pyrrol-l-yl) ethyl) -1,3,5,2, 4-trioxadisilocan- 6-yl ) acetate
Figure imgf000042_0002
To a solution of compound G) (2.06 g, 2.57 mmol) in CH2CI2 (30 mL) , (R) -1, 2-dinitrooxyhexan-6-ol (0.57 g, 2.57 mmol) was added. The solution was cooled at 0°C and DMAP (94 mg, 0.77 mmol) and EDAC (0.74 g, 3.85 mmol) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with CH2CI2 (100 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x50 mL) . The organic layer was washed with ¾0 (4x30 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, gradient from n- hexane/AcOEt 95/5 to n-hexane/EtOAc 60/40 during 1.4 L) affording the title compound (1.96 g, yield 74%) .
S) ( 3R, 5R) -3- ( ( S ) -2 , 3-bis (nitrooxy) propoxy) propyl 7- (2 fluorophenyl) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) - pyrrol-l-yl) -3, 5-dihydroxyheptanoate (Compound (5)) To a solution of compound R) (1.96 g, 1.91 mmol) in dry THF (50 mL) , cooled at 0°C, acetic acid (0.78 mL, 12.64 mmol) and a 1M THF solution of TBAF (11.5 mL, 11.5 mmol) were added. The reaction was stirred at room temperature for 24 hours. The solution was diluted with AcOEt (250 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x100 mL) . The organic layer was washed with ¾0 (4x100 mL) and brine, then dried over Na2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, eluent: gradient from n-hexane/EtOAc 7/3 to EtOAc during 1.4 L) affording the compound (5) (0.81 g, 58%).
1H-NMR (DMSO-de) : 9.75 (1H, s), 7.50 (2H, d) , 7.19 (6H, m) , 6.99 (6H, m) , 5.56 (1H, m) , 4.93 (1H, dd) , 4.77 (1H, dd) , 4.74 (1H, d) , 4.62 (1H, d) , 4.04 (2H, t), 3.96-3.70 (5H, m) , 3.51 (3H, m) , 3.23 (1H, m) , 2.37 (1H, dd) , 2.25 (1H, dd) , 1.79 (2H, m) , 1.67-1.39 (4H, m) , 1.38 (6H, m) .
Example 7
Synthesis of ( 3R, 5R) -3- ( (R) -2 , 3-bis (nitrooxy) propoxy) propyl 7- (2- (4-fluorophenyl) -5-isopropyl-3-phenyl-4- (phenyl carbamoyl) -lH-pyrrol-l-yl) -3, 5-dihydroxyheptanoate
(Compound (5)
Figure imgf000043_0001
(5)
T) 3- ( (S) -2, 3-dinitrooxypropoxy) propyl 2- ( ( 6R, 8R) -2 , 2 , 4 , 4- tetraisopropyl-8- (2- (2-isopropyl-4 , 5-diphenyl-3- (phenyl carbamoyl) -lH-pyrrol-l-yl) ethyl) -1,3,5,2, 4-trioxadisilocan- 6-yl ) acetate
Figure imgf000044_0001
ON02
To a solution of compound G) (1.99 g, 2.57 mmol) in CH2CI2 (20 mL) , (S) -1, 2-dinitrooxyhexan-6-ol (0.55 g, 2.48 mmol) was added. The solution was cooled at 0°C and DMAP (91 mg, 0.74 mmol) and EDAC (0.71 g, 3.72 mmol) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with CH2CI2 (100 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x50 mL) . The organic layer was washed with ¾0 (4x30 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, gradient from n- hexane/AcOEt 95/5 to n-hexane/EtOAc 60/40 during 1.4 L) affording the title compound (2.09 g, yield 79%) .
U) ( 3R, 5R) -3- ( (R) -2 , 3-bis (nitrooxy) propoxy) propyl 7- (2- (4- fluorophenyl ) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -1H- pyrrol-l-yl) -3, 5-dihydroxyheptanoate (Compound (5))
To a solution of compound T) (1.97 g, 1.91 mmol) in dry THF (40 mL) , cooled at 0°C, acetic acid (0.78 mL, 12.64 mmol) and a 1M THF solution of TBAF (11.5 mL, 11.5 mmol) were added. The reaction was stirred at room temperature for 24 hours. The solution was diluted with AcOEt (250 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x100 mL) . The organic layer was washed with ¾0 (4x100 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, eluent: gradient from n-hexane/EtOAc 7/3 to EtOAc during 1.4 L) affording the compound (5) (0.95 g, yield 64%).
1H-NMR (DMSO-de) : 9.75 (1H, s), 7.50 (2H, d) , 7.19 (6H, m) , 6.99 (6H, m) , 5.56 (1H, m) , 4.93 (1H, dd) , 4.76 (2H, m) , 4.62 (1H, d) , 4.04 (2H, t) , 3.96-3.70 (5H, m) , 3.51 (3H, m) , 3.23 (1H, m) , 2.37 (1H, dd) , 2.25 (1H, dd) , 1.79 (2H, m) , 1.67-1.39 (4H, m) , 1.38 (6H, m) .
Example 8
Synthesis of (3R, 5R) -( (S) -5, 6-bis (nitrooxy) hexyl) -7- (2- (4- fluorophenyl ) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -1H- pyrrol-l-yl) -3, 5-dihydroxyheptanoate (Compound (4))
Figure imgf000045_0001
(4)
V) (S) 5 , 6-dinitrooxyhexyl-2- ( (6R,8R)-2,2,4,4-tetraiso propyl-8- (2 (2-isopropyl-4 , 5-diphenyl-3- (phenylcarbamoyl) - lH-pyrrol-1 yl) ethyl) -1,3,5,2, 4-trioxadisilocan-6-yl) acetate
Figure imgf000045_0002
02
To a solution of compound G) (2.09 g, 2.62 mmol) in CH2C I 2 (50 mL) , (S) -1, 2-dinitrooxyhexan-6-ol (0.70 g, 3.15 mmol) was added. The solution was cooled at 0°C and DMAP (128 mg, 1.04 mmol) and EDAC (1.0 g, 5.24 mmol) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with CH2C I 2 (100 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x50 mL) . The organic layer was washed with ¾0 (4x30 mL) and brine, then dried over Na2SC>4 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, gradient from n-hexane to n-hexane/EtOAc 60/40 during 1.7 L) affording the title compound (2.38 g, yield 90%).
W) (3R, 5R) - ( (S) -5, 6-bis (nitrooxy) hexyl) -7- (2- (4-fluoro phenyl) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -lH-pyrrol- 1-yl) -3, 5-dihydroxyheptanoate (Compound (4))
To a solution of compound V) (2.38 g, 2.36 mmol) in dry THF (40 mL) , cooled at 0°C, acetic acid (0.96 mL, 15.57 mmol) and a 1M THF solution of TBAF (11.1 mL, 11.1 mmol) were added. The reaction was stirred at room temperature for 24 hours. The solution was diluted with AcOEt (250 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x100 mL) . The organic layer was washed with ¾0 (4x100 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, eluent: gradient from n-hexane/EtOAc 9/1 to n-hexane/EtOAc 2/8 during 1.4 L) affording the compound (4) (1.5 g, yield 83%) .
1H-NMR (DMSO-de) : 9.73 (1H, s), 7.50 (2H, d) , 7.19 (6H, m) , 6.99 (6H, m) , 5.41 (1H, m) , 4.92 (1H, dd) , 4.70 (2H, m) , 4.59 (1H, d) , 4.01 (2H, t) , 3.98-3.66 (3H, m) , 3.51 (1H, m) , 3.23 (1H, m) , 2.31 (2H, m) , 1.80-1.39 (6H, m) , 1.38 (6H, m) . Example 9
Synthesis of (3R, 5R) -( (S) -1- ( (R) -2, 3-bis (nitrooxy) propyl) pyrrolidin-2-yl) methyl 7- (2- (4-fluorophenyl) -5-isopropyl-3- phenyl-4- (phenylcarbamoyl ) -lH-pyrrol-l-yl) -3, 5-dihydroxy heptanoate (Com ound (43) )
Figure imgf000047_0001
(43)
X) ( (S) -1- ( (R) -2, 3-dinitrooxypropyl) pyrrolidin-2-yl) methyl 2- ( (6R, 8R) -2, 2, 4, 4-tetraisopropyl- 8- (2- (2-isopropyl-4 , 5- diphenyl-3- (phenylcarbamoyl) -lH-pyrrol-l-yl) ethyl) -1, 3, 5, 2, 4-trioxadisilocan-6- l) acetate
Figure imgf000047_0002
To a solution of compound G) (0.72 g, 0.9 mmol) in CH2CI2 (14.5 mL) , (R) -3- ( (S) -2- (hydroxymethyl) pyrrolidin-l-yl) - 1 , 2-dinitrooxypropane (0.25 g, 0.96 mmol) was added. The solution was cooled at 0°C and DMAP (22 mg, 0.18 mmol) and EDAC (259 mg, 1.35 mmol)) were added. The reaction was stirred at room temperature for 24 hours. The solution was diluted with CH2CI2 (15 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x30 mL) . The organic layer was washed with ¾0 (4x15 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 50 g, gradient n-hexane/AcOEt 8/2 (75mL), from n-hexane/AcOEt 8/2 to n-hexane/EtOAc 1/1 during 520 mL, n-hexane/EtOAc 1/1 (75 mL) ) affording the title compound (0.66 g, yield 70%). Y) (3R, 5R) - ( (S) -1- ( (R) -2, 3-bis (nitrooxy)propyl)pyrrolidin- 2 -yl) methyl 7- (2- (4-fluorophenyl) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -lH-pyrrol-l-yl) -3, 5-dihydroxyheptanoate (Compound (43) )
To a solution of compound X) (0.64 g, 0.61 mmol) in dry THF (6 mL) , cooled at 0°C, acetic acid (0.24 mL, 3.99 mmol) and a 1M THF solution of TBAF (3.37 mL, 3.37 mmol) were added. The reaction was stirred at room temperature for 17 hours. The solution was diluted with AcOEt (50 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x30 mL) . The organic layer was washed with ¾0 (4x30 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 50 g, eluent: gradient n- hexane/AcOEt 1/1 (75mL) , from n-hexane/AcOEt 1/1 to n- hexane/EtOAc 1/9 during 230 mL, n-hexane/EtOAc 1/9 (230 mL) ) affording the compound (43) (0.59 g, yield 11%).
1H-NMR (DMSO-de) : 9.74 (1H, s), 7.50 (2H, d) , 7.19 (6H, m) , 6.99 (6H, m) , 5.51 (1H, m) , 4.97-4.59 (4H, m) , 4.04-3.63 (5H, m) , 3.51 (1H, m) , 3.33-2.98 (3H, m) , 2.75 (2H, m) , 2.43-2.20 (3H, m) , 1.88-1.33 (8H, m) , 1.38 (6H, m) .
Example 10
Synthesis of (3R, 5R) -( (S) -2- (nitrooxy) -3- (3- (nitrooxy) propoxy) propyl ) 7- (2- (4-fluorophenyl) -5-isopropyl-3-phenyl- 4- (phenylcarbamoyl ) -lH-pyrrol-l-yl) -3, 5-dihydroxyheptanoate (Compound ( 42 ) )
Figure imgf000048_0001
ON02
(42) AB) (S) -2-nitrooxy-3- (3-nitrooxypropoxy) propyl 2-((6R,8R)- 2,2,4,4-tetraisopropyl-8- (2- (2-isopropyl-4, 5-diphenyl-3- (phenylcarbamoyl ) -lH-pyrrol-l-yl) ethyl) -1,3,5,2, 4-trioxa disilocan- 6-yl ) acetate
Figure imgf000049_0001
To a solution of compound G) (0.89 g, 1.11 mmol) in CH2CI2 (20 mL) , (S) -2-nitrooxy-3- (3-nitrooxypropoxy) propan-l-ol (0.26 g, 1.11 mmol) was added. The solution was cooled at 0°C and DMAP (54 mg, 0.4 mmol) and EDAC (425 mg, 2.2 mmol)) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with CH2CI2 (30 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x30 mL) . The organic layer was washed with ¾0 (4x30 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 50 g, gradient from n-hexane/AcOEt 9/1 to n-hexane/EtOAc 1/1 during 750 mL) affording the title compound (1.0 g, yield 88%) .
AC) (3R, 5R) - ( (S) -2- (nitrooxy) -3- (3- (nitrooxy) propoxy) propyl) -7- (2- (4-fluorophenyl) -5-isopropyl-3-phenyl-4-
(phenylcarbamoyl ) -lH-pyrrol-l-yl) -3, 5-dihydroxyheptanoate
(Compound ( 42 ) )
To a solution of compound AB) (1.13 g, 1.1 mmol) in dry THF (20 mL) , cooled at 0°C, acetic acid (0.41 mL, 6.6 mmol) and a 1M THF solution of TBAF (6.6 mL, 6.6 mmol) were added. The reaction was stirred at room temperature for 2 hours. The solution was diluted with AcOEt (50 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x30 mL) . The organic layer was washed with ¾0 (4x30 mL) and brine, then dried over Na2SC>4 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 50 g, eluent: gradient from n-hexane/AcOEt 9/1 to EtOAc during 1.7 L) affording the compound (42) (0.24 g, yield 28%).
1H-NMR (DMSO-de) : 9.75 (1H, s), 7.50 (2H, d) , 7.19 (6H, m) , 6.99 (6H, m) , 5.47 (1H, m) , 4.74 (1H, dd) , 4.60 (3H, m) , 4.42-4.21 (2H, m) , 4.08-3.42 (8H, m) , 3.23 (1H, m) , 2.43 (1H, dd) , 2.30 (1H, dd) , 1.91 (2H, m) , 1.73-1.39 (6H, m) , 1.38 (6H, m) .
Example 11
Synthesis of ( 3R, 5R) -(( S ) -2 , 6-bis (nitrooxy) hexyl ) 7- (2- (4- fluorophenyl ) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -1H- pyrrol-l-yl) -3 5-dihydroxyheptanoate (Compound (9))
Figure imgf000050_0001
(9)
AD) (S) -2, 6-dinitrooxyhexyl-2- ( (6R, 8R) -2, 2, 4, 4-tetra isopropyl-8- (2- (2-isopropyl-4 , 5-diphenyl-3- (phenyl
carbamoyl) -lH-pyrrol-l-yl) ethyl) -1,3,5,2, 4-trioxadisilocan- 6-yl ) acetate
Figure imgf000050_0002
To a solution of compound G) (0.46 g, 0.58 mmol) in CH2CI2 (10 mL) , ( S ) -2 , 6-dinitrooxyhexanol (0.46 g, 0.58 mmol) was added. The solution was cooled at 0°C and DMAP (28 mg, 0.23 mmol) and EDAC (222 mg, 1.1 mmol) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with CH2CI2 (30 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x30 mL) . The organic layer was washed with ¾0 (4x30 mL) and brine, then dried over Na2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 50 g, gradient from n-hexane/AcOEt 9/1 to n-hexane/EtOAc 1/1 during 750 mL) affording the title compound (0.51 g, yield 87%).
AE) (3R, 5R) - ( (S) -2, 6-bis (nitrooxy) hexyl) 7- (2- (4-fluoro phenyl) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -lH-pyrrol- 1-yl) -3, 5-dihydroxyheptanoate (Compound (9))
To a solution of compound AD) (0.51 g, 0.5 mmol) in dry THF (10 mL) , cooled at 0°C, acetic acid (0.19 mL, 3.05 mmol) and a 1M THF solution of TBAF (3.05 mL, 3.05 mmol) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with AcOEt (15 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x10 mL) . The organic layer was washed with ¾0 (4x10 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, eluent: gradient from n-hexane/AcOEt 9/1 to EtOAc during 1.7 L) affording the compound (9) (0.14 g, yield 36%).
1H-NMR (DMSO-de) : 9.74 (1H, s), 7.50 (2H, d) , 7.19 (6H, m) , 6.99 (6H, m) , 5.28 (1H, m) , 4.72 (1H, d) , 4.61 (1H, d) , 4.51 (2H, t), 4.34 (1H, dd) , 4.17(1H, dd) , 4.03-3.66 (3H, m) , 3.51 (1H, m) , 3.23 (3H, m) , 2.34 (2H, m) , 1.78-1.21 (8H, m) , 1.36 (6H, m) . Example 12
Synthesis of ( 3R, 5R) -( (R) -2 , 6-bis (nitrooxy) hexyl ) 7- (2- (4- fluorophenyl ) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -1H- pyrrol-l-yl) -3 5-dihydroxyheptanoate (Compound (9))
Figure imgf000052_0001
(9)
AJ) (R) -2, 6-dinitrooxyhexyl-2- ( ( 6R, 8R) -2 , 2 , 4 , 4-tetraiso propyl-8- (2- (2-isopropyl-4 , 5-diphenyl-3- (phenylcarbamoyl) - lH-pyrrol-l-yl) ethyl) -1,3,5,2, 4-trioxadisilocan-6-yl) acetate
Figure imgf000052_0002
To a solution of compound G) (0.4 g, 0.5 mmol) in CH2CI2 (10 mL) , (R) -2-6-dinitrooxyhexanol (0.11 g, 0.5 mmol) was added. The solution was cooled at 0°C and DMAP (24 mg, 0.2 mmol) and EDAC (192 mg, 1 mmol)) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with CH2CI2 (60 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x30 mL) . The organic layer was washed with ¾0 (4x30 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 50 g, gradient from n-hexane/AcOEt 9/1 to EtOAc during 750 mL) affording the title compound (0.38 g, yield 76%) . AK) (3R, 5R) - ( (R) -2, 6-bis (nitrooxy) hexyl) -7- (2- (4-fluoro phenyl) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -lH-pyrrol- 1-yl) -3, 5-dihydroxyheptanoate (Compound (9)
To a solution of compound AH) (0.38 g, 0.37 mmol) in dry THF (6 mL) , cooled at 0°C, acetic acid (0.15 mL, 2.49 mmol) and a 1M THF solution of TBAF (2.3 mL, 2.3 mmol) were added. The reaction was stirred at room temperature for 24 hours. The solution was diluted with AcOEt (35 mL) and washed with an aqueous solution of 5 % aH2 P04 (2x20 mL) . The organic layer was washed with ¾0 (4x20 mL) and brine, then dried over a2 S 04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 50 g, eluent: gradient from n-hexane/AcOEt 9/1 to AcOEt during 750 mL) affording the compound (9) (0.27 g, yield 72%) .
NMR (DMSO-de) : 9.74 (1H, s), 7.50 (2H, d) , 7.19 (6H, m) , 6.99 (6H, m) , 5.29 (1H, m) , 4.73 (1H, d) , 4.61 (1H, d) , 4.51(2H, t), 4.39 (1H, dd) , 4.15(1H, dd) , 4.03-3.66 (3H, m) , 3.51 (1H, m) , 3.23 (3H, m) , 2.41 (1H, dd) , 2.28 (1H, m) , 1.78-1.21 (8H, m) , 1.36 (6H, m) .
Example 13
Synthesis of (3R, 5R) - ( (S) -3, 4-bis (nitrooxy) butyl) -7- (2- (4- fluorophenyl ) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl) -1H- pyrrol-l-yl) -3 5-dihydroxyheptanoate (Compound (6))
Figure imgf000053_0001
(6)
AL) (S) -3, 4-dinitrooxybutyl-2- ( (6R, 8R) -2 , 2 , 4 , 4-tetraiso propyl-8- (2- (2-isopropyl-4 , 5-diphenyl-3- (phenylcarbamoyl) - lH-pyrrol-l-yl) ethyl) -1,3,5,2, 4-trioxadisilocan-6-yl) acetate
Figure imgf000054_0001
To a solution of compound G) (1.63 g, 2.04 mmol) in CH2CI2 (30 mL) , (S) -3-4-dinitrooxybutanol (0.4 g, 2.04 mmol) was added. The solution was cooled at 0°C and DMAP (24 mg, 0.2 mmol) and EDAC (410 mg, 2.14 mmol)) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with CH2CI2 (50 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x20 mL) . The organic layer was washed with ¾0 (4x20 mL) and brine, then dried over Na2SC>4 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, gradient from n- hexane/AcOEt 84/16 to n-hexane/AcOEt 75/25 during 1750 mL) affording the title compound (1.15 g, yield 75%) .
AM) (3R, 5R) - ( (S) -3, 4-bis (nitrooxy) butyl ) -7- (2- (4-fluoro phenyl) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -lH-pyrrol- 1-yl) -3, 5-dihydroxyheptanoate (Compound (6))
To a solution of compound AL) (1.5 g, 1.5 mmol) in dry THF (30 mL) , cooled at 0°C, triethylamine trihydrofluoride (1 mL, 6.17 mmol) was added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with AcOEt (100 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x50 mL) . The organic layer was washed with ¾0 (4x50 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, eluent: gradient from n- hexane/AcOEt 55/45 to n-hexane/AcOEt 3/7 during 1680 mL) affording the compound (6) (0.38 g, yield 37%).
NMR (CDC13) : 7.26-6.94 (14H, m) , 6.87 (1H, s), 5.41 (1H, m) , 4.79 (1H, dd) , 4.53 (1H, dd) , 4.28 (2H, t) , 4.15 (2H, m) , 3.97(1H, m) , 3.75 (1H, m) , 3.59 (1H, m) , 2.43 (2H, d) ,
2.11- (2H, m) , 1.79-1.40 (4H, m) , 1.55 (6H, m) , 1.28 (1H, m) .
Example 14
Synthesis of ( 3R, 5R) -(( S ) -2 , 3-bis (nitrooxy) propyl ) 7- (2 fluorophenyl) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) - pyrrol-l-yl) -3, 5-dih droxyheptanoate (Compound (6))
Figure imgf000055_0001
(6)
AN) (S) -3, 4-dinitrooxybutyl 2- ( ( 6R, 8R) -2 , 2 , 4 , 4-tetraiso propyl-8- (2- (2-isopropyl-4 , 5-diphenyl-3- (phenylcarbamoyl) - lH-pyrrol-l-yl) ethyl) -1,3,5,2, 4-trioxadisilocan-6-yl) acetate
Figure imgf000055_0002
To a solution of compound G) (1.63 g, 2.04 mmol) in CH2CI2 (30 mL) , (S) -3-4-dinitrooxybutanol (0.4 g, 2.04 mmol) was added. The solution was cooled at 0°C and DMAP (24 mg, 0.2 mmol) and EDAC (410 mg, 2.14 mmol) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with CH2CI2 (50 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x20 mL) . The organic layer was washed with ¾0 (4x20 mL) and brine, then dried over Na2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, gradient from n-hexane/AcOEt 84/16 to n-hexane/AcOEt 75/25 during 1750 mL) affording the title compound (1.15 g, yield 75%). AO) (3R, 5R) - ( (S) -3, 4-bis (nitrooxy) butyl ) -7- (2- (4-fluoro phenyl) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -lH-pyrrol- 1-yl) -3, 5-dihydroxyheptanoate (Compound (6))
To a solution of compound AN) (1.5 g, 1.5 mmol) in dry THF (30 mL) , cooled at 0°C, triethylamine trihydrofluoride (1 mL, 6.17 mmol) was added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with AcOEt (100 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x50 mL) . The organic layer was washed with ¾0 (4x50 mL) and brine, then dried over Na2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, eluent: gradient from n- hexane/AcOEt 55/45 to n-hexane/AcOEt 3/7 during 1680 mL) affording the compound (6) (0.38 g, yield 37%).
NMR (CDCI3) : 7.26-6.94 (14H, m) , 6.87 (1H, s), 5.47 (1H, m) , 4.78 (1H, dd) , 4.66 (1H, dd) , 4.55 (1H, dd) , 4.30 (1H, dd) , 4.23-4.04 (2H, m) , 3.97 (1H, m) , 3.75 (1H, m) , 3.59 (1H, m) , 2.47 (2H, d) , 1.79-1.40 (4H, m) , 1.55 (6H, m) . Example 15
Synthesis of (3R, 5R) - ( (R) -2, 3-bis (nitrooxy) propyl) -7- (2- (4- fluorophenyl ) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl) -1H- pyrrol-l-yl) -3, 5-dihydroxyheptanoate (Compound (7))
Figure imgf000057_0001
(7)
AP) (R) -2, 3-dinitrooxypropyl 2- ( ( 6R, 8R) -2 , 2 , 4 , 4-tetraiso propyl-8- (2- (2-isopropyl-4 , 5-diphenyl-3- (phenylcarbamoyl ) - lH-pyrrol-l-yl) ethyl) -1,3,5,2, 4-trioxadisilocan-6-yl) acetate
Figure imgf000057_0002
To a solution of compound G) (1.72 g, 2.14 mmol) in CH2CI2 (20 mL) , (R) -2 , 3-dinitrooxypropanol (0.39 g, 2.14 mmol) was added. The solution was cooled at 0°C and DMAP (24 mg, 0.2 mmol) and EDAC (430 mg, 2.25 mmol)) were added. The reaction was stirred at room temperature for 6 hours. The solution was diluted with CH2CI2 (50 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x20 mL) . The organic layer was washed with ¾0 (4x20 mL) and brine, then dried over Na2SC>4 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, gradient from n- hexane/AcOEt 85/15 to n-hexane/AcOEt 7/3 during 1750 mL) affording the title compound (1.69 g, yield 82%) .
AQ) (3R, 5R) - ( (S) -2, 3-bis (nitrooxy) propyl ) -7- (2- (4-fluoro phenyl) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl) -lH-pyrrol- 1-yl) -3, 5-dihydroxyheptanoate (Compound (7)) To a solution of compound AP) (1.8 g, 1.86 mmol) in dry THF (15 mL) , cooled at 0°C, triethylamine trihydrofluoride (1.2 mL, 7.3 mmol) was added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with AcOEt (100 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x50 mL) . The organic layer was washed with ¾0 (4x50 mL) and brine, then dried over Na2SC>4 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, eluent: gradient from n- hexane/AcOEt 55/45 to n-hexane/AcOEt 3/7 during 1740 mL) affording the compound (7) (0.31 g, yield 23%).
NMR (CDC13) : 7.26-6.94 (14H, m) , 6.87 (1H, s), 5.47 (1H, m) , 4.78 (1H, dd) , 4.66 (1H, dd) , 4.55 (1H, dd) , 4.30 (1H, dd) , 4.23-4.04 (2H, m) , 3.97 (1H, m) , 3.75 (1H, m) , 3.59 (1H, m) , 2.47 (2H, d) , 1.79-1.40 (4H, m) , 1.55 (6H, m) .
Example 16
(3R, 5R) - ( (R) -3, 4-bis (nitrooxy) butyl ) 7- (2- (4-fluorophenyl) 5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -lH-pyrrol-l-yl) - 3 , 5-dihydrox heptanoate (Compound (7))
Figure imgf000058_0001
(7)
AR) (R) -3, 4-dinitrooxybutyl-2- ( ( 6R, 8R) -2 , 2 , 4 , 4-tetraiso propyl-8- (2- (2-isopropyl-4 , 5-diphenyl-3- (phenylcarbamoyl) - lH-pyrrol-l-yl) ethyl) -1,3,5,2, 4-trioxadisilocan-6-yl) acetate
Figure imgf000059_0001
To a solution of compound G) (1.63 g, 2.04 mmol) in CH2CI2 (20 mL) , (R) -3, 4-dinitrooxybutanol (0.40 g, 2.04 mmol) was added. The solution was cooled at 0°C and DMAP (24 mg, 0.2 mmol) and EDAC (410 mg, 2.1 mmol)) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with CH2CI2 (50 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x20 mL) . The organic layer was washed with ¾0 (4x20 mL) and brine, then dried over Na2 S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, gradient from n-hexane/AcOEt 8/2 to n-hexane/AcOEt 7/3 during 1750 mL) affording the title compound (1.32 g, yield 66%).
AS) (3R, 5R) - ( (R) -3, 4 -bis (nitrooxy) butyl) -7- (2- (4-fluoro phenyl) -5-isopropyl-3-phenyl-4- (phenylcarbamoyl ) -lH-pyrrol- 1-yl) -3, 5-dihydroxyheptanoate (Compound (7))
To a solution of compound AR) (1.32 g, 1.34 mmol) in dry THF (30 mL) , cooled at 0°C, triethylamine trihydrofluoride (0.87 mL, 5.3 mmol) was added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with AcOEt (100 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x50 mL) . The organic layer was washed with ¾0 (4x50 mL) and brine, then dried over a2 S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, eluent: gradient from n- hexane/AcOEt 55/45 to n-hexane/AcOEt 35/65 during 1740 mL) affording the compound (7) (0.52 g, yield 53%). NMR (CDCI3) : 7.26-6.94 (14H, m) , 6.87 (1H, s), 5.41 (1H, m) , 4.79 (1H, dd) , 4.53 (1H, dd) , 4.28 (2H, t) , 4.15 (2H, m) , 3.97 (1H, m) , 3.75 (1H, m) , 3.59 (1H, m) , 2.43 (2H, d) , 2.11 (2H, m) , 1.79-1.40 (4H, m) , 1.55 (6H, m) , 1.28 (1H, m) .
Example 17
Synthesis of (3R, 5S, E) - ( (R) -5, 6-bis (nitrooxy) hexyl) -7- (4- (4-fluorophenyl) -6-isopropyl-2- (N-methylmethylsulfonamido) pyrimidin-5-yl ) -3 5-dihydroxyhept- 6-enoate (Compound (47))
Figure imgf000060_0001
(47)
AT) (3R, 5S, E) -methyl 7- (4- (4-fluorophenyl) -6-isopropyl-2- N-methylmethylsulfonamido) pyrimidin-5-yl ) -3, 5-dihydroxy
Figure imgf000060_0002
To a solution of rosuvastatin calcium (1.0 g, 0.2 mmol) in MeOH (10 mL) and DMF (5 mL) , trimethylchlorosilane (0.41 g, 4 mmol) was added. The reaction was stirred at room temperature for 3 hours. The solution was diluted with AcOEt (60 mL) and washed with ¾0 (4x30 mL) and brine, then dried over Na2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, gradient from n- hexane/AcOEt 1/1 to n-hexane/AcOEt 3/7 during 1750 mL) affording the title compound (0.78 g, yield 78%) . AU) (3R, 5S, E) -methyl 3, 5-bis (tert-butyldimethylsilyloxy) -7- (4- (4-fluorophenyl) -6-isopropyl-2- (N-methylmethylsulfon amido) pyrimidin-5-yl hept- 6-enoate
Figure imgf000061_0001
To a solution of compound AT) (0.9 g, 2 mmol) in DMF (10 mL) , tertbutyldimethylchlorosilane (0.64 g, 4.1 mmol) and imidazole (0.28 g, 4.2 mmol) were added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with AcOEt (100 mL) and washed with H20 (4x40 mL) and brine, then dried over Na2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, gradient from n-hexane/AcOEt 9/1 to n-hexane/AcOEt 75/25 during 1750 mL) affording the title compound (0.98 g, yield 68%) .
AV) (3R, 5S,E) -3, 5-bis (tert-butyldimethylsilyloxy) -7- (4- (4- fluorophenyl ) -6-isopropyl-2- (N-methylmethylsulfonamido) pyrimidin-5-yl ) hept- 6-enoic acid
Figure imgf000061_0002
To a solution of compound AU) (0.4 g, 0.55 mmol) in THF (3 mL) and MeOH (2 mL) , LiOH (0.04 g, 1.65 mmol) was added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with AcOEt (50 mL) and washed with ¾0 (4x30 mL) and brine, then dried over Na2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 50 g, gradient from n-hexane/AcOEt 8/2 to n-hexane/AcOEt 7/3 during 900 mL) affording the title compound (0.21 g, yield 53%). AW) (3R, 5S,E) - ( (R) -5, 6-dinitrooxyhexyl ) -3, 5-bis (tert- butyldimethylsilyloxy) -7- (4- (4-fluorophenyl) -6-isopropyl-2- (N-methylmethylsulfonamido pyrimidin-5-yl ) hept- 6-enoate
Figure imgf000062_0001
To a solution of compound AV) (0.8 g, 1.13 mmol) in CH2CI2 (20 mL) , (R) -5 , 6-dinitrooxyhexanol (0.25 g, 1.13 mmol) was added. The solution was cooled at 0°C and DMAP (24 mg, 0.2 mmol) and EDAC (227 mg, 1.18 mmol)) were added. The reaction was stirred at room temperature for 5 hours. The solution was diluted with CH2CI2 (50 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x20 mL) . The organic layer was washed with ¾0 (4x20 mL) and brine, then dried over a2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, gradient from n- hexane/AcOEt 8/2 to n-hexane/AcOEt 7/3 during 1450 mL) affording the title compound (0.63 g, yield 61%) .
AX) (3R, 5S,E) - ( (R) -5, 6-bis (nitrooxy) hexyl ) -7- (4- (4-fluoro phenyl) -6-isopropyl-2- (N-methylmethylsulfonamido) pyrimidin- 5-yl ) -3 , 5-dihydroxyhept- 6-enoate (Compound (47))
To a solution of compound AW) (0.63 g, 0.69 mmol) in dry THF (10 mL) , cooled at 0°C, tetrabutylammonium fluoride Solution 1M in THF (2.08 mL, 2.08 mmol) was added. The reaction was stirred at room temperature for 24 hours. The solution was diluted with AcOEt (50 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x30 mL) . The organic layer was washed with ¾0 (4x30 mL) and brine, then dried over Na2SC>4 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 50 g, eluent: n-hexane/AcOEt 1/1 (900 mL) ) affording the compound (47) (0.21 g, yield 44%) .
NMR (CDC13) : 7.66 (2H, m) , 7.09 (2H, m) , 6.66 (1H, d) , 5.47 (1H, dd) , 5.29 (1H, m) , 4.75 (1H, dd) , 4.49 (2H, m) , 4.27- 4.08 (4H, m) , 3.58 (3H, s) , 3.52 (3H, s) , 3.36 (1H, m) , 2.49 (2H, m) , 1.75 (4H, m) , 1.55 (4H, m) , 1.28 (6H, m) .
Example 18
Synthesis of (3R, 5S, E) -( (S) -5, 6-bis (nitrooxy) hexyl) -7- (4- (4-fluorophenyl) -6-isopropyl-2- (N-methylmethylsulfonamido) pyrimidin-5-yl ) -3 5-dihydroxyhept- 6-enoate (Compound (47))
Figure imgf000063_0001
(47)
AY) (3R, 5S, E) -allyl 7- (4- (4-fluorophenyl) -6-isopropyl-2- (N- methylmethylsulfonamido) pyrimidin-5-yl ) -3, 5-dihydroxyhept- 6-enoate
Figure imgf000063_0002
To a solution of rosuvastatin calcium salt (7.0 g, 7 mmol) in DMF (100 mL) , was added allyl bromine (1.7 g, 14 mmol) . The solution was stirred at room temperature for 12 hours. The solution was diluted with ¾0 (200 mL) and extracted with EtOAc (3 x 200 mL) . The combined organic layers were washed with brine (2 x 100 mL) , dried over Na?S04 and concentrated under reduced pressure affording the title compound (4.81 g, yield 66%).
AZ) Allyl 2- ( (6R, 8S) -8- ( (E) -2- (4- (4-fluorophenyl) -6- isopropyl-2- (N-methylmethylsulfonamido) pyrimidin-5-yl ) vinyl) -2, 2, 4, 4-tetraisopropyl-l, 3, 5, 2, 4-trioxadisilocan-6- yl ) acetate
Figure imgf000064_0001
To a solution of compound AY) (3.3 g; 5.83 mmol) in DMF (15 mL) , was added imidazole (0.87 g; 12.83 mmol). A solution of 1, 3-dichloro-l, 1, 3, 3-tetraisopropyldisiloxane (1.84 g; 5.83 mmol) in DMF (5mL) was then added dropwise. The reaction was stirred at room temperature for 4 hours. The solution was diluted with an aqueous solution of 5 % NaH2P04 (50 mL) and extracted with AcOEt (3 x 25 mL) . The combined organic layers were washed with ¾0 (4 X 20 mL) and brine, then dried over Na2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 340 g, eluent: gradient from n-hexane/EtOAc 9/1 to n- hexane/EtOAc 8/2 in 10 L) affording the title compound (1.86 g, 42%) .
BA) 2- ( (6R, 8S) -8- ( (E) -2- (4- (4-fluorophenyl) -6-isopropyl-2- (N-methylmethylsulfonamido) pyrimidin-5-yl ) vinyl )-2, 2,4,4- tetraisopropyl-1 , 3,5,2, 4-trioxadisilocan-6-yl) acetic acid
Figure imgf000065_0001
To a solution of compound AZ) (0.29 g; 0.39 mmol) in dry THF (20 mL) , was added palladium tetrakis (0.27 g; 0.23 mmol) followed by morpholine (2.05 mL, 23.55 mmol) . The solution was stirred for 12 hours. The solution was diluted with AcOEt (100 mL) and washed with H20 (4 X 40 mL) and brine, then dried over Na2S04 and concentrated under reduced pressure then concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 100 g, eluent: gradient n- hexane/EtOAc 7/3 to n-hexane/EtOAc 55/45 in 870 mL) affording the title compound (1.51 g, 89%) .
NMR (CDC13) : 7.61 (2H, d) , 7.09 (2H, d) , 6.65 (1H, d) , 5.43 (1H, d) , 4.47 (2H, dd) , 4.13 (1H, q) , 3.58 (3H, s), 3.52 (3H, s), 3.33 (1H, m) , 2.59 (2H, m) , 1.77-1.42 (5H, m) , 1.27 (6H, d) , 1.06 (24H, m) .
BB) (S) -5, 6-dinitrooxyhexyl-2- ( (6R, 8S) -8- ( (E) -2- (4- (4- fluorophenyl ) -6-isopropyl-2- (N-methylmethylsulfonamido) pyrimidin-5-yl ) inyl) -2, 2, 4, 4-tetraisopropyl-l, 3, 5, 2, 4- trioxadisilocan- 6- l ) acetate
Figure imgf000065_0002
To a solution of compound BA) (0.28 g, 0.39 mmol) in CH2CI2 (10 mL) , ( S ) -5 , 6-dinitrooxyhexanol (0.08 g, 0.39 mmol) was added. The solution was cooled at 0°C and DMAP (12 mg, 0.1 mmol) and EDAC (80 mg, 0.41 mmol)) were added. The reaction was stirred at room temperature for 5 hours. The solution was diluted with CH2CI2 (50 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x20 mL) . The organic layer was washed with ¾0 (4x20 mL) and brine, then dried over Na2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 50 g, gradient from n-hexane/AcOEt 8/2 to n-hexane/AcOEt 7/3 during 900 mL) affording the title compound (0.32 g, yield 86%). BC) (3R, 5S,E) -( (S) -5, 6-bis (nitrooxy) hexyl) -7- (4- (4-fluoro phenyl) -6-isopropyl-2- (N-methylmethylsulfonamido) pyrimidin -5-yl ) -3 , 5-dihydroxyhept- 6-enoate (Compound (47))
To a solution of compound BB) (0.32 g, 0.34 mmol) in dry THF (30 mL) , cooled at 0°C, triethylamine trihydrofluoride (0.85 mL, 5.2 mmol) was added. The reaction was stirred at room temperature for 12 hours. The solution was diluted with AcOEt (90 mL) and washed with an aqueous solution of 5 % Na¾P04 (2x40 mL) . The organic layer was washed with ¾0 (4x40 mL) and brine, then dried over Na2S04 and concentrated under reduced pressure. The residue was purified by flash chromatography (Biotage System, SNAP Cartridge silica 50 g, eluent: n-hexane/AcOEt 1/1 (900 mL) ) affording the title compound (0.21 g, yield 44%) . affording the compound (47) (0.18 g, yield 80%).
NMR (CDCI3) : 7.66 (2H, m) , 7.09 (2H, m) , 6.66 (1H, d) , 5.47 (1H, dd) , 5.29 (1H, m) , 4.75 (1H, dd) , 4.49 (2H, m) , 4.27- 4.08 (4H, m) , 3.58 (3H, s) , 3.52 (3H, s) , 3.36 (1H, m) , 2.49 (2H, m) , 1.75 (4H, m) , 1.55 (4H, m) , 1.28 (6H, m) . Example Fl : Evaluation of the effect of the compounds on vascular tone
The ability of statins dinitrate derivatives disclosed in examples 1-3, 5-8, 10, 11 and 13-16 to induce vasodilation in comparison to native statin (atovarstatin) and statins mononitrate derivatives (Compounds A and B) , was tested in vitro in isolated rabbit thoracic aorta preparations (Wanstall J.C. et al . , Br. J. Pharmacol., 134:463-472, 2001) .
Com ound A
Figure imgf000067_0001
Com ound B
Figure imgf000067_0002
Male New Zealand rabbits were anaesthetized with thiopental-Na (50 mg/kg, iv) , sacrificed by exsanguinations and then the thorax was opened and the aorta dissected. Single ring preparations (4 mm in length) of thoracic aorta were set up in physiological salt solution (PSS) at 37°C in small organ chambers (5 ml) . The composition of PSS was (mM) : NaCl 130, NaHC03 14.9, KH2P04 1.2, MgS04 1.2, HEPES 10, CaCl2 , ascorbic acid 170 and glucose 1.1 (95% O2 /5% CO2 ; pH 7.4) . Each ring was mounted under 2 g passive tension in 5 ml organ bath. Isometric tension was recorded with a Grass transducer (Grass FT03) attached to a BIOPAC MP150 System. Preparations were allowed to equilibrate for 1 h, then contracted submaximally with noradrenaline (NA, 1 μΜ) and, when the contraction was stable, acetylcholine (ACh, 10 μΜ) was added. A relaxant response to ACh indicated the presence of a functional endothelium. When a stable precontraction was reached, a cumulative concentration- response curve to each vasorelaxant agent was obtained in the presence of a functional endothelium. Time intervals between different concentrations were based on the time needed to reach a full response. Moreover, the effect of the soluble guanylyl cyclase inhibitor ODQ (1-H- (1, 2, 4) - oxadiazol (4, 3-a) quinoxalin-l-one) on the dilator responses elicited by the compounds was examined preincubating the aortic rings with ODQ (10 μΜ) for 20 min. Responses to relaxing agents are expressed as a percentage of residual contraction and plotted against concentration of test compound. EC50 values (EC50 is the concentration producing 50% of the maximum relaxation to the test compound) were interpolated from these plots. During the experimental period, the plateau obtained with NA was stable without significant spontaneous loss of contraction in the aortic rings. Under these experimental conditions, native statin did not produce relaxation up to 100 μΜ, the curve being not different from that obtained in presence of vehicle alone .
Table 1 reports the EC50 of the tested compounds, the results shown that the dinitrates of the invention showed improved EC50 values in vessel relaxation assay compared to mononitrate derivatives (Compound A and B) which are greater than two fold. Furthermore, in experiments performed in presence of ODQ (10 μΜ) , the vasorelaxant responses to all the tested drugs were inhibited showing that the vasodilative effects are mediated by NO.
Figure imgf000069_0001
Example F2 : Evaluation of pharmacokinetic parameters
This study investigated the bioavailability following oral administration of the compounds of the present invention in comparison to atorvastatin. Tested compounds:
Compounds of examples 10, 11, 13 and 14;
Reference compound: Atorvastatin .
The formulations of the tested compounds were prepared on the day of dosing, immediately before of administration, by dissolving an appropriate amount of the test compound in PEG400/DMA 70/30, in order to obtain the following target concentrations :
Figure imgf000070_0001
Male Sprague Dawley Rats, age 7-9 weeks, bodyweight in the range 216-279 g at the dosing, were supplied by Charles River Italia, Calco (Italy) .
Approximately three days before dosing the rats, under anesthesia (an aqueous mixture of ketamine, xylazine and acepromazine, by subcutaneous administration) , were surgically prepared for serial blood samples collection from superior vena cava (SVC) . Rats were fitted with a flexible plastic cannula implanted in the superior vena cava via the jugular vein. Each animal was allowed approximately five days postoperative recovery period prior to dosing.
Test compounds were administered orally to each animal by gavage . Animals were fasted overnight before dosing. On the day of dosing the food was offered about 4 h post dosing. Serial blood samples (about 0.3 mL) were collected from the catheter of each SVC catheterized rat, using syringes containing Na-heparin as anticoagulant, at pre-dose, 15 min, 30 min, 1, 2, 4, 6 and 8 h post-dosing. The samples were then transferred into pre-cooled tubes (containing 7.5 L of a solution of sodium fluorure, 40 mg/mL and DICHLORVOS, 10 mg/mL, in order to inhibit plasma esterase activity, in water for injection), protected from light. The blood was centrifuged at 10000 rpm for 3 min at +4 °C to collect plasma (within 30 min from blood collection) , and plasma samples were stored at -80°C.
Plasma samples were thawed in ice bath. Plasma aliquots of 50 yL were protein precipitated in triplicate using 150 yL of acetonitrile to which were also added 10 yL of DMSO, vortex-mixed and centrifuged 10 min at 4°C at 4000 rpm. The supernatant was transferred to a clean plate and injected onto a LC-MS system. Atorvastatin, ortho and para hydroxy metabolites levels were quantified using a calibration curve (range 0.01-10 μΜ ) prepared in rat plasma, spiking an appropriate volume of working solution in 50 yL of plasma and then precipitating the proteins (see below for standard curve preparation) using 150 yL of acetonitrile, vortex-mixed and centrifuged 10 min at 4°C at 4000 rpm. The supernatant was transferred to a clean plate and injected onto a LC-MS system. Analytical conditions were as follows: Column: ACQUITY BEH Phenyl 50 x 2.1 mm (1.7y) @ 40°C
Mobile Phase: A. water + 0.1% formic acid
B. methanol + 0.1% formic acid
Flow Rate: 0.5 mL/min Gradient profile:
Time (min) : 0.0 1.0 1.5
A (%) : 60 0.0 0.0
B (%) : 40 100 100
Injection Volume 5 μΐ
Detection was performed using a Waters Quattro Micro API mass spectrometer operated in ESI+ mode. The multiple reaction monitoring (MRM) pair monitored was m/z 559.26-^ 440.2 for atorvastatin and m/z 575.16 -> 440.2 for ortho and para hydroxy metabolites. The autosampler cooler was maintained at 4°C.
The results reported in tables 2 and 3 show that the compounds of the invention have a better oral bioavailability in comparison to the native statin.
Table 2 reports a summary of pharmacokinetic parameters of the compounds of the invention and of atorvastatin following single oral administration to SD rats (dose equimolar to atorvastatin 20 mg/kg)
Figure imgf000072_0001
Table 3 reports a summary of pharmacokinetic parameters of 2-hydroxy metabolite of atorvastatin formed from the compounds of the invention following single oral administration to SD rats (dose equimolar to atorvastatin 20 mg/kg)
Figure imgf000073_0001

Claims

1. A compound of general formula (I) or a pharmaceutically acceptable salt or stereoisomer thereof
Figure imgf000074_0001
(I) wherein R is selected from the group consisting of (Ila)- (Ilf) :
Figure imgf000074_0002
Ila) (lib)
Figure imgf000074_0003
(He) (Ilf) w is an integer selected between 0 and 1 ;
R1 and R2 can be equal or different and are independently selected from the group consisting of hydrogen and C1-C4 alkyl ;
Y is selected from the group consisting of:
A) [R3-0] P-R4-CH (ON02) - (R5) n-CH (ON02) - [O-R6] PL-R7
B) R4-CH (ON02) -R3-0-R5-CH (ON02) -R7
C) (R8) n-R9- [R3-0] P-R4-CH (ON02) - (R5) ni-CH (ON02) - [O-R6] PL-R7 wherein
R3, R4, R5, R6 and R8 are independently a straight or branched C1-C10 alkylene;
n is an integer from 0 to 1 ;
nl is an integer from 0 to 1 ;
p is an integer from 0 to 4 ;
pi is an integer from 0 to 4 ;
R7 is selected from the group consisting of hydrogen and
C^-Cg alkyl;
R9 is a saturated, unsaturated or aromatic heterocyclic 5 or 6 members ring, containing one or more heteroatoms selected from nitrogen, oxygen or sulphur.
2. A compound of general formula (I) or a pharmaceutically acceptable salt or stereoisomer thereof according to claim 1 wherein R is the group (lid) or (lie)
3. A compound of general formula (I) or a pharmaceutically acceptable salt or stereoisomer thereof according to claim 1 or 2 wherein w is 1.
4. A compound of general formula (I) or a pharmaceutically acceptable salt or stereoisomer thereof according to claim 3 wherein R1 and R2 can be equal or different and are independently selected from the group consisting of hydrogen and C¾.
5. A compound of general formula (I) according to claim 1 or 2 wherein w is 0.
6. A compound of formula (I) according to anyone of the claims 1-5 or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Y of the group A) is selected from the roup consisting of:
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000077_0002
Figure imgf000077_0003
Figure imgf000077_0004
Figure imgf000078_0001
Figure imgf000078_0002
Figure imgf000078_0003
7. A compound of formula (I) according to anyone of the claims 1-5 or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Y of the group B) is selected from the roup consisting of:
Figure imgf000078_0004
Figure imgf000079_0001
Figure imgf000079_0002
8. A compound of formula (I) according to anyone of the claims 1-5 or a pharmaceutically acceptable salt or stereoisomer thereof, wherein Y of the group C) is selected from the group consisting of:
Figure imgf000080_0001
9. A compound of formula (I) according to anyone of the claims 1-8 or a pharmaceutically acceptable salt or stereoisomer thereof selected from the roup consisting of:
Figure imgf000080_0002
Figure imgf000081_0001
Figure imgf000081_0002
Figure imgf000081_0003
Figure imgf000081_0004
Figure imgf000081_0005
Figure imgf000081_0006
Figure imgf000081_0007
Figure imgf000082_0001
Figure imgf000082_0002
Figure imgf000082_0003
Figure imgf000082_0004
Figure imgf000082_0005
Figure imgf000082_0006
Figure imgf000082_0007
Figure imgf000083_0001
Figure imgf000083_0002
Figure imgf000083_0003
Figure imgf000083_0004
Figure imgf000083_0005
Figure imgf000083_0006
Figure imgf000083_0007
82
Figure imgf000084_0001
Figure imgf000084_0002
Figure imgf000084_0003
Figure imgf000084_0004
Figure imgf000084_0005
Figure imgf000084_0006
83
Figure imgf000085_0001
Figure imgf000085_0002
Figure imgf000085_0004
Figure imgf000085_0005
Figure imgf000085_0006
Figure imgf000086_0001
Figure imgf000086_0002
Figure imgf000086_0003
Figure imgf000086_0004
Figure imgf000086_0005
Figure imgf000086_0006
Figure imgf000087_0001
Figure imgf000087_0002
Figure imgf000087_0003
Figure imgf000087_0004
Figure imgf000087_0005
(87) (88)
10. A compound of formula (I) according to claims 1-9 for use as a medicament.
11. A compound according to claims 1-9 for use as a drug having anti-inflammatory, antithrombotic and antiplatelet activity .
12. A compound according to claims 1-9 for reducing cholesterol and triglycerides levels and/or for raising HDL-C levels.
13. A compound according to claims 1-9 for use in the treatment of acute coronary syndromes, stroke, peripheral vascular diseases and all disorders associated with endothelial dysfunctions.
14. A compound according to claim 13 for use in the treatment of peripheral ischemia, vascular complications in diabetic patients and atherosclerosis.
15. A compound according to claims 1-9 for use in the treatment of neurodegenerative and autoimmune disorders.
16. A compound according to claim 15 for use in the treatment of Alzheimer's disease, Parkinson's disease and multiple sclerosis.
17. A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a pharmaceutically effective amount of a compound of general formula (I) and/or a salt or stereoisomer thereof according to claims 1-9.
18. A pharmaceutical composition according to claim 17 in a suitable form for the oral, parenteral, rectal, topic and transdermic administration, by inhalation spray or aerosol or iontophoresis devices.
19. A pharmaceutical composition comprising a compound of formula (I) according to claims 1-9, and at least a compound used to treat cardiovascular disease and a pharmaceutically acceptable carrier.
20. A pharmaceutical composition according to claim 19, wherein the compound used to treat cardiovascular disease is selected from the group consisting of: ACE inhibitors, renin inhibitors, angiotensin II receptor antagonists, beta-adrenergic blockers, calcium channel blockers, antithrombotics, aspirin, nitrosated ACE inhibitors, nitrosated renin inhibitors nitrosated angiotensin II receptor antagonists, nitrosated beta-adrenergic blockers and nitrosated aspirin.
21. A pharmaceutical kit comprising the composition of claim 17 and a compound used to treat cardiovascular disease .
22. A process for the preparation of compounds of formula (I) as defined in claim 1,
wherein w is 0,
Figure imgf000089_0001
ined in claim 1, comprising the following steps: 1) reacting compounds of formula (III)
Figure imgf000090_0001
(III)
wherein M is an alkali metal or alkaline earth metal such as sodium or calcium and R is as defined in claim 1 ;
with a compound of formula X-Hal wherein X is selected from the formulas (IXa)-(IXb)
Figure imgf000090_0002
(IXa) (IXb)
and Hal is selected from Br or CI,
in an aprotic polar/non polar solvent such as THF, DMF or CH2CI2, at temperature ranging from -80°C to 60°C;
2) reacting a compound of formula (IV) obtained in step 1)
Figure imgf000090_0003
(IV)
wherein R and X are as above defined, with 1 , 3-dichloro- 1 , 1 , 3 , 3-tetraisopropyldisiloxane in presence of imidazole in an aprotic polar/non polar solvent such as THF, DMF or CH2CI2, at temperature ranging from -80°C to 60°C;
3) deprotecting of the carboxylic function of compound of formula (V) obtained in step 2)
Figure imgf000090_0004
(V)
wherein R and X are as above defined:
when X is (IXa) by catalytic hydrogenation in presence of Pd/C in an aprotic polar/non polar solvent such as THF at room temperature and pressure; when X is (IXb) with palladium tetrakis, in presence of dimedone and triphenylphosphine or in presence of morpholine, in an aprotic polar/non polar solvent such as THF, DMF or CH2CI2, at temperature ranging from -10 °C to 30°C;
4) reacting a compound of formula (VI) obtained in step 3)
Figure imgf000091_0001
(VI)
wherein R is as above defined with a compound of formula (X)
HO-Y
(X)
wherein Y is as above defined in the presence of a condensing agent such as dicyclohexylcarbodiimide (DCC) , N' - ( 3-dimethylaminopropyl ) -N-ethylcarbodiimide
hydrochloride (EDAC) , N, N' -carbonyldiimidazole (CDI), optionally in the presence of a base, for example DMAP . The reaction is carried out in an inert organic solvent dry such as N, N' -dimethylformamide, tetrahydrofuran, benzene, toluene, dioxane, a polyhalogenated aliphatic hydrocarbon at a temperature from -20°C and 50°C. The reaction is completed within a time range from 30 minutes to 36 hours; 5) deprotecting of the diol function of compound of formula (VII) obtained in step 4
Figure imgf000091_0002
(VII)
wherein R is as above defined in presence of fluoride salt of alkylamine such as triethylamine trihydrofluoride or tetrabutylamonium fluoride and acetic acid, in an aprotic polar/non polar solvent such as THF, at temperature ranging from -10°C to 30°C.
23. A compound of general formula (VI) or a pharmaceutically acceptable salt or stereoisomer thereof
Figure imgf000092_0001
(VI)
wherein R is selected from the group consisting of (Ila)- (Ilf) .
24. A compound of general formula (VI) according to claim 23 wherein R is (lid) .
25. A compound of general formula (VI) according to claim 23 wherein R is (He) .
PCT/EP2011/059778 2010-06-21 2011-06-14 Statin derivatives WO2011160974A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP10166595.8 2010-06-21
EP10166595 2010-06-21

Publications (2)

Publication Number Publication Date
WO2011160974A2 true WO2011160974A2 (en) 2011-12-29
WO2011160974A3 WO2011160974A3 (en) 2012-04-05

Family

ID=44533472

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/059778 WO2011160974A2 (en) 2010-06-21 2011-06-14 Statin derivatives

Country Status (1)

Country Link
WO (1) WO2011160974A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103724278A (en) * 2013-12-12 2014-04-16 江苏阿尔法药业有限公司 Preparation method for tatin intermediate and derivatives thereof
WO2020069065A1 (en) * 2018-09-28 2020-04-02 The Regents Of The University Of California Compounds and methods for treating gastrointestinal disease
EP3737676A4 (en) * 2018-01-09 2021-09-15 Ligand Pharmaceuticals, Inc. Acetal compounds and therapeutic uses thereof
US11278559B2 (en) 2014-02-13 2022-03-22 Ligand Pharmaceuticals Incorporated Prodrug compounds and their uses
EP4005633A2 (en) 2022-02-08 2022-06-01 Sawant, Mohit Manikrao Crystalline form of rosuvastatin allyl ester
WO2023152754A1 (en) * 2022-02-08 2023-08-17 Sawant, Mohit Manikrao Crystalline form of rosuvastatin allyl ester

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997016405A1 (en) 1995-10-31 1997-05-09 Nicox S.A. New compounds and their compositions having anti-inflammatory and anti-thrombotic activities
WO1998021193A1 (en) 1996-11-14 1998-05-22 Nicox S.A. Antithrombotic organic nitrates
WO2004106300A1 (en) 2003-05-28 2004-12-09 Nicox S.A. Captopril derivatives
WO2004105754A1 (en) 2003-05-27 2004-12-09 Nicox S.A. Nitrooxyderivatives of fluvastatin, pravastatin, cerivastatin, atorvastatin and rosuvastatin as cholesterol-reducing agents with improved anti-inflammatory, antithrombotic and antiplatelet activity
WO2004110432A1 (en) 2003-06-19 2004-12-23 Nicox S.A. Enalapril-nitroxyderivatives derivatives and related compounds as ace inhibitors for the treatment of cardiovascular diseases
WO2005011646A2 (en) 2003-07-31 2005-02-10 Nicox S.A. Nitrooxy derivatives of losartan, valsatan, candesartan, telmisartan, eprosartan and olmesartan as angiotensin-ii receptor blockers for the treatment of cardiovascular diseases
WO2007045551A2 (en) 2005-10-18 2007-04-26 Nicox S.A. Renin inhibitors nitroderivatives

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007132482A2 (en) * 2006-05-17 2007-11-22 Manne Satyanarayana Reddy Novel process for the preparation of pitavastatin and its pharmaceutically acceptable salts
WO2009000592A1 (en) * 2007-06-25 2008-12-31 Nicox S.A. Use of nitric oxide-releasing statins in the treatment of pulmonary arterial hypertension

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997016405A1 (en) 1995-10-31 1997-05-09 Nicox S.A. New compounds and their compositions having anti-inflammatory and anti-thrombotic activities
WO1998021193A1 (en) 1996-11-14 1998-05-22 Nicox S.A. Antithrombotic organic nitrates
WO2004105754A1 (en) 2003-05-27 2004-12-09 Nicox S.A. Nitrooxyderivatives of fluvastatin, pravastatin, cerivastatin, atorvastatin and rosuvastatin as cholesterol-reducing agents with improved anti-inflammatory, antithrombotic and antiplatelet activity
WO2004106300A1 (en) 2003-05-28 2004-12-09 Nicox S.A. Captopril derivatives
WO2004110432A1 (en) 2003-06-19 2004-12-23 Nicox S.A. Enalapril-nitroxyderivatives derivatives and related compounds as ace inhibitors for the treatment of cardiovascular diseases
WO2005011646A2 (en) 2003-07-31 2005-02-10 Nicox S.A. Nitrooxy derivatives of losartan, valsatan, candesartan, telmisartan, eprosartan and olmesartan as angiotensin-ii receptor blockers for the treatment of cardiovascular diseases
WO2007045551A2 (en) 2005-10-18 2007-04-26 Nicox S.A. Renin inhibitors nitroderivatives

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
"The Merck Index"
P.O. BONETTI ET AL., EUROPEAN HEART JOURNAL, vol. 24, 2003, pages 225 - 248
WANSTALL J.C. ET AL., BR. J. PHARMACOL., vol. 134, 2001, pages 463 - 472

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103724278A (en) * 2013-12-12 2014-04-16 江苏阿尔法药业有限公司 Preparation method for tatin intermediate and derivatives thereof
US11278559B2 (en) 2014-02-13 2022-03-22 Ligand Pharmaceuticals Incorporated Prodrug compounds and their uses
EP3737676A4 (en) * 2018-01-09 2021-09-15 Ligand Pharmaceuticals, Inc. Acetal compounds and therapeutic uses thereof
US11970482B2 (en) 2018-01-09 2024-04-30 Ligand Pharmaceuticals Inc. Acetal compounds and therapeutic uses thereof
WO2020069065A1 (en) * 2018-09-28 2020-04-02 The Regents Of The University Of California Compounds and methods for treating gastrointestinal disease
CN113166067A (en) * 2018-09-28 2021-07-23 加利福尼亚大学董事会 Compounds and methods for treating gastrointestinal disorders
EP4005633A2 (en) 2022-02-08 2022-06-01 Sawant, Mohit Manikrao Crystalline form of rosuvastatin allyl ester
EP4005633A3 (en) * 2022-02-08 2022-06-08 Sawant, Mohit Manikrao Crystalline form of rosuvastatin allyl ester
WO2023152754A1 (en) * 2022-02-08 2023-08-17 Sawant, Mohit Manikrao Crystalline form of rosuvastatin allyl ester

Also Published As

Publication number Publication date
WO2011160974A3 (en) 2012-04-05

Similar Documents

Publication Publication Date Title
WO2011160974A2 (en) Statin derivatives
EP2125695B1 (en) Nitric oxide donor compounds
US7297808B2 (en) Statin derivatives
EP1653950B1 (en) Nitrooxy derivatives of losartan, valsatan, candesartan, telmisartan, eprosartan and olmesartan as angiotensin-ii receptor blockers for the treatment of cardiovascular diseases
US20020091105A1 (en) Phosphate derivatives as immunoregulatory agents
EP1780197A1 (en) 5-substituted-2-phenylamino-benzamide as mek inhibitor
EP1560826B1 (en) Pyridopyrimidinone compounds, method for production thereof and medicaments comprising the same
EP2021324B1 (en) Nitrated heterocyclic compounds as endothelin receptor antagonist
EP1558612B1 (en) Imidazopyridine derivatives, preparation method and pharmaceutical compositions containing same
WO2006079610A1 (en) Nitrooxy sartan derivatives as angiotensin ii receptor blockers for the treatment of cardiovascular and inflammatory diseases
JP2011513269A (en) Novel angiotensin II receptor blocker derivatives
EP1934186A2 (en) Renin inhibitors nitroderivatives
JP3116256B2 (en) (Thio) urea derivatives
EP1748994B1 (en) Nitrooxyderivatives of antihypertensive drugs
SK43499A3 (en) New pentaerythritol derivatives, their production and use and intermediates for their synthesis
SK169599A3 (en) Novel pentaerythrite derivatives, the production and use thereof and intermediate products for the synthesis of the same
EP0638572B1 (en) Bicyclic derivatives of imidazole, process for their preparation, intermediates; their application as medicaments and pharmaceutical compostions containing them
KR20060013413A (en) Captopril derivatives
WO2007104652A2 (en) Non-peptidic renin inhibitors nitroderivatives
CZ20004071A3 (en) Benzofuroxan derivatives and their use when treating angina pectoris

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: 11725085

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11725085

Country of ref document: EP

Kind code of ref document: A2