WO2000037423A1 - Substituted 2-phenyl-1-(3,4-dihydroxy-5-nitrophenyl)-1-ethanones, their use in the treatment of some central and peripheral nervous system disorders and pharmaceutical compositions containing them - Google Patents

Substituted 2-phenyl-1-(3,4-dihydroxy-5-nitrophenyl)-1-ethanones, their use in the treatment of some central and peripheral nervous system disorders and pharmaceutical compositions containing them Download PDF

Info

Publication number
WO2000037423A1
WO2000037423A1 PCT/GB1999/004310 GB9904310W WO0037423A1 WO 2000037423 A1 WO2000037423 A1 WO 2000037423A1 GB 9904310 W GB9904310 W GB 9904310W WO 0037423 A1 WO0037423 A1 WO 0037423A1
Authority
WO
WIPO (PCT)
Prior art keywords
dihydroxy
optionally substituted
group
comt
ethanone
Prior art date
Application number
PCT/GB1999/004310
Other languages
French (fr)
Inventor
Jan BENEŠ
Patrício Manuel Vieira Araújo SOARES DA SILVA
David Alexander Learmonth
Original Assignee
Portela & C.A., 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 Portela & C.A., S.A. filed Critical Portela & C.A., S.A.
Priority to BRPI9908084-2A priority Critical patent/BR9908084B1/en
Priority to AU18731/00A priority patent/AU754113B2/en
Priority to HU0100786A priority patent/HU226396B1/en
Priority to PL99342542A priority patent/PL193998B1/en
Publication of WO2000037423A1 publication Critical patent/WO2000037423A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D317/00Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D317/08Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3
    • C07D317/44Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D317/46Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • C07D317/48Methylenedioxybenzenes or hydrogenated methylenedioxybenzenes, unsubstituted on the hetero ring
    • C07D317/50Methylenedioxybenzenes or hydrogenated methylenedioxybenzenes, unsubstituted on the hetero ring with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to atoms of the carbocyclic ring
    • C07D317/54Radicals substituted by oxygen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/10Antioedematous agents; Diuretics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C205/00Compounds containing nitro groups bound to a carbon skeleton
    • C07C205/39Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by esterified hydroxy groups
    • C07C205/42Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by esterified hydroxy groups having nitro groups or esterified hydroxy groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton
    • C07C205/43Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by esterified hydroxy groups having nitro groups or esterified hydroxy groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton to carbon atoms of the same non-condensed six-membered aromatic ring or to carbon atoms of six-membered aromatic rings being part of the same condensed ring system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C205/00Compounds containing nitro groups bound to a carbon skeleton
    • C07C205/45Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by at least one doubly—bound oxygen atom, not being part of a —CHO group
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C205/00Compounds containing nitro groups bound to a carbon skeleton
    • C07C205/49Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by carboxyl groups
    • C07C205/57Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by carboxyl groups having nitro groups and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton
    • C07C205/59Compounds containing nitro groups bound to a carbon skeleton the carbon skeleton being further substituted by carboxyl groups having nitro groups and carboxyl groups bound to carbon atoms of six-membered aromatic rings of the carbon skeleton the carbon skeleton being further substituted by singly-bound oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C255/00Carboxylic acid nitriles
    • C07C255/49Carboxylic acid nitriles having cyano groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton
    • C07C255/56Carboxylic acid nitriles having cyano groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton containing cyano groups and doubly-bound oxygen atoms bound to the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C309/00Sulfonic acids; Halides, esters, or anhydrides thereof
    • C07C309/63Esters of sulfonic acids
    • C07C309/72Esters of sulfonic acids having sulfur atoms of esterified sulfo groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton
    • C07C309/73Esters of sulfonic acids having sulfur atoms of esterified sulfo groups bound to carbon atoms of six-membered aromatic rings of a carbon skeleton to carbon atoms of non-condensed six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • C07C45/27Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by oxidation
    • C07C45/29Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by oxidation of hydroxy groups
    • C07C45/292Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by oxidation of hydroxy groups with chromium derivatives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • C07C45/45Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by condensation
    • C07C45/46Friedel-Crafts reactions
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C45/00Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds
    • C07C45/61Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups
    • C07C45/67Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by isomerisation; by change of size of the carbon skeleton
    • C07C45/673Preparation of compounds having >C = O groups bound only to carbon or hydrogen atoms; Preparation of chelates of such compounds by reactions not involving the formation of >C = O groups by isomerisation; by change of size of the carbon skeleton by change of size of the carbon skeleton

Definitions

  • L-DOPA L- ⁇ -3,4-dihydroxyphenylala ⁇ i ⁇ e
  • AADC aromatic L-amino acid decarboxylase
  • peripheral AADC inhibitor be ⁇ serazide or carbidopa
  • peripheral AADC inhibitor be ⁇ serazide or carbidopa
  • L-DOPA-i ⁇ duced clinical improvement wanes at the end of each dose cycle, producing the "end-of-dose” or "wearing-off” pattern of motor fluctuations.
  • a close relationship has been described between accumulation of 3-O-methyl-L-DOPA and development of the "weari ⁇ g- off' phenomenon (Tohgi, H., et al., Neurosci. Letters, 132:19-22, 1992).
  • pharmacokinetic alterations may be an advantage over increasing the dose of L-DOPA, which also increases AUC, but additionally raises peak concentrations.
  • raising peak concentrations relates to adverse effects such as dyski ⁇ esia, which occurs immediately when COMT inhibitors are given but can be anticipated by either reducing the dose of L-DOPA or increasing the time intervals between doses.
  • the effects of COMT inhibition also differ from those of co ⁇ trolled-release L-DOPA formulation which slow down absorption and reduce bioavailability.
  • the pharmacokinetic changes induced by COMT inhibition reduce the daily L- DOPA dose by enabling a reduction of each dose or an increase in dose intervals.
  • tolcapone and entacapone are recommended to be administered as frequently as 3 times a day; because the half-life of entacapone is considerably shorter than that of tolcapone, the recommended dose for entacapone is twice that for tolcapone.
  • the molecule generally enhances inhibition of the COMT catalysed
  • the invention relates to substituted 2-phenyl-1-(3,4-dihydroxy-5-
  • R ⁇ and R are hydrogens or groups hydrolysable under
  • R 3 are the same or different and signify hydrogen, optionally substituted
  • alkanoylamino group lower dialkanoylamino group, carboxyi, optionally
  • dialkylami ⁇ o or cyano group or taken together signify aliphatic or
  • halogen denotes
  • aryl denotes a
  • carb ⁇ cyciic aromatic group preferably mono- or bicyclic groups.
  • inert pharmaceutically acceptable carriers are admixed with the active compounds.
  • the pharmaceutically acceptable carriers may be
  • Solid form preparations include powders, tablets,
  • a solid carrier can be one or more
  • flavouring agents substances which may also act as diluents, flavouring agents,
  • solubilizers lubricants, suspending agents, binders or tablet
  • disintegrating agents it may also be an encapsulating material.
  • the pharmaceutical preparation is in unit dosage form, e.g.
  • the dosages may be varied depending on the requirement of the
  • the total daily dosage may be divided and
  • Fig. 1 is a graph showing brain COMT activity at different times after oral
  • Fig. 2. is a graph showing liver COMT activity at different times after oral
  • Fig. 3 is a graph showing concentration dependent inhibition of brain
  • Fig. 4 is a graph showing concentration dependent inhibition of liver
  • Fig. 5 is a graph showing concentration-dependent amphetamine-
  • Fig. 6 is a graph showing concentration-dependent amphetamine-
  • each column represents the mean of eight experiments per group and vertical lines the respective SEM.
  • reaction mixture was preincubated for 20 min with increasing concentrations of test compounds (0.5 to 1 ,000 nM); the incubation was performed in the presence of a concentration of adrenaline five times the corresponding K m value as determined in saturation experiments.
  • test compounds were given by gastric tube to overnight fasted rats. Therafter, at defined intervals, animals were killed by decapitation and livers and brains removed and used to determine COMT activity as described above. At the end of the incubation period (brain, 15 min; liver, 5 min) the tubes were transferred to ice and the reaction was stopped by the addition of 200 ⁇ l of 2 M perchloric acid. The samples were then centrifuged (200xo ⁇ 4 min, 4°C), and 500 ⁇ l aliquots of the supernatant, filtered on 0.22 ⁇ m pore size Spin-X filter tubes (Costar) were used for the assay of meta ⁇ ephrine.
  • the assay of metanephrine was carried out by means of high pressure liquid chromatography with electrochemical detection.
  • the lower limits for detection of metanephrine ranged from 350 to 500 fmol (0.5 to 1.0 pmol/mg protei ⁇ /h).
  • K m and V ma ⁇ values for COMT activity were calculated from non-linear regression analysis using the GraphPad Prism statistics software package (Motulsky, H.G., et al., GraphPad Prisms, GraphPad Prism
  • Spontaneous locomotor activity was measured using a San Diego I ⁇ struments rodent activity monitor (model Flex Field, San Diego Instruments, San Diego, CA) with 48 infrared motion sensors.
  • the lower frame was 50.5 x 50.5 cm, with 32 photocells (separated by 2.5 cm) located lengthwise 5 cm above the floor.
  • the upper frame was 50.5 x 50.5 cm, with 16 photocells (separated by 2.5 cm) located lengthwise 15 cm above the floor.
  • the test field was an acrylic chamber with internal dimensions 40 x 40 x 37 cm. Ten-minute activity recording began immediately after placing the test subject at the centre of the chamber. Activity was measured automatically with a personal computer using
  • Flex Field software (San Diego Instruments) which provides user-defined intervals of total interruptions. Three parameters of normal spontaneous ' locomotion were recorded: horizontal activity, vertical activity and centre time. Stereotypical behaviour (intense sniffing, repetitive head and limb movements and licking and biting, as defined by Feldma ⁇ , R.S., Meyer, J.S., Quenzer, L.F., Principles of Neuropharmacology, 1997, Sinauer Associates, Inc. Publishers, Su ⁇ deriand, MA) were quantified by an independent observer after being recorded on tape by means of video tracking system (VP200, HVS Image, Ltd) placed 70 cm above the test field. Animals were habituated to the test field environment for one hour prior to behavioural testing.
  • video tracking system VP200, HVS Image, Ltd
  • Compou ⁇ ds of formulae A - E were found to be potent inn: both brain and liver COMT, the maximal inhibitory effect being a within 30 min after their oral administration (table 2).
  • Compc A presented a similar inhibitory profile in brain and liver COMT, whereas compound E was much more potent upon liver COMT than brain COMT.
  • compound B was also much more potent as a peripheral COMT inhibitor than in brain.
  • Compounds with longer carbon chains were less potent at inhibiting brain COMT in comparison with their effects upon liver COMT. This difference may have to do with difficulties in access to brain.
  • Compounds with short carbon chains (A, B and C) were not equally potent at inhibiting peripheral and central COMT, but this difference was not so striking as observed with compounds with long carbon chains.
  • compound B was less potent than tolcapone at inhibiting brain COMT with ED 5 o's of 5.3 ⁇ 1.1 and 1.6 ⁇ 0.1 mg/kg, respectively. At the highest dose tested (30 mg/kg), entacapone failed to reach the 50% inhibition level.
  • Amphetamine is a potent psychostimulant that depending on the dose administered produces increased locomotor behaviour and various stereotypical activities.
  • a single low dose of amphetamine administered to rats leads to a characteristic response pattern consisting of increased locomotor activity, rearing, mild sniffing and head bobbing.
  • Gradually increasing the dose of amphetamine results in a decrease in locomotion and rearing, which are replaced by focused stereotypies (repetitive, seemingly aimless behaviours performed in a relatively invariant manner) confined to a small area of the cage floor (Feldman, R.S., Meyer, J.S., Quenzer, L.F., Principles of Neuropharmacology, 1997, Sinauer Associates, inc. Publishers, Sunderla ⁇ d, MA).
  • the cerebral dopami ⁇ ergic system has traditionally been crucial to the ability of amphetamine to stimulate locomotor activity and stereotypical behaviours. With respect to the anatomic substrates of amphetamine action, there is evidence that stimulation of dopaminergic activity in the nucleus accumbens is responsible for amphetamine-induced locomotor activity, whereas stimulation of dopami ⁇ ergic activity in the caudate- putamen is linked with focused stereotypies produced by high doses of
  • Parkinson' disease may be of therapeutical benefit, such as Parkinson' disease and
  • inhibitor with limited access to the brain such as compound B
  • peripheral AADC inhibitor Due to the possibility that COMT inhibitors
  • DOPA treated patients the use of a substance such as compound B is
  • the crystalline residue was recrystallised from a mixture of dichloromethane and diethylether, yielding 1-(4-be ⁇ zyloxy-3- methoxyphenyl)-2-phenyl-1 -etha ⁇ o ⁇ e as white crystals, m.p 134 to 135° C.
  • Example 13 1 -(3,4-dihydroxy-5-nitrophenyl)-2-(2-methylphe ⁇ yl)-1 -
  • the emulsion formed was extracted by ethyl acetate, and the organic layer was washed with brine, dried, and the solvent was evaporated under diminished pressure.
  • the residue was chromatographed on a column of silica gel with a mixture of petroleum ether and ethyl acetate to give 1-(4-hydroxy-3- methoxyphenyl)-2-(2-methylphe ⁇ yl)-1 -ethanone as off white crystals, m.p. 79 to 81° C.
  • nitrophe ⁇ yl)-2-phenyl-1 -ethanone in 90 mL of dichloromethane was treated with 7.85 g (lOOmmol) of acetylchloride, 7.51 g (95 mmol) of pyridine, and a catalytic amount of 4-dimethylami ⁇ opyridine.
  • the solution formed was washed successively with ice-cold 0.2N hydrochloric acid, 1 % aqueous solution of sodium bicarbonate and brine.
  • the dried (Na 2 S0 4 ) solution was evaporated under reduced pressure, and the residue recrystallised from a mixture of diethylether and petroleum ether giving the desired product as yellow crystals, m.p. 94 to 95° C.

Abstract

New compounds of formula (I) are described. The compounds have potentially valuable pharmaceutical properties in the treatment of some central and peripheral nervous system disorders, where a reduction in the O-methylation of catecholamines may be of therapeutical benefit, such as Parkinson's disease and parkinsonian disorders, gastrointestinal disturbances, edema formation states and hypertension.

Description

Description
Substituted 2-pheπyl-1-(3,4-dihydroxy-5-πitrophenyI)-1-ethaπoπes,
their use in the treatment of some central and peripheral nervous
system disorders and pharmaceutical compositions containing
them.
The most effective symptomatic treatment of Parkinson's disease involves the administration of L-β-3,4-dihydroxyphenylalaπiπe (L-DOPA), the immediate precursor of dopamine. Orally administered L-DOPA is predominantly metabolised in the periphery by aromatic L-amino acid decarboxylase (AADC) to dopamine, which can cause serious adverse effects such as emesis, orthostatic hypotension and cardiac aσhvthmia. Therefore, L-DOPA is usually administered in combination with a
peripheral AADC inhibitor (beπserazide or carbidopa). When administered together with such inhibitors, very little dopamiπe is formed in the periphery, but only a small amount of an oral dose of L-DOPA reaches the brain because a considerable amount of the drug undergoes methylation to, 3-O-methyl-L-DOPA ( annistδ, P.A., et al., Progress Drug Research, 39: 291-350, 1992). The duration of the L-DOPA-induced
clinical improvement is brief as a result of the short half-life of L-DOPA, which contrasts with the long half-life of 3-O-methyl-L-DOPA. Within a few years after starting L-DOPA therapy with the customary 2 to 4 doses per day, L-DOPA-iπduced clinical improvement wanes at the end of each dose cycle, producing the "end-of-dose" or "wearing-off" pattern of motor fluctuations. A close relationship has been described between accumulation of 3-O-methyl-L-DOPA and development of the "weariπg- off' phenomenon (Tohgi, H., et al., Neurosci. Letters, 132:19-22, 1992). It has been anticipated that this might result from inhibition of L-DOPA transport at the level of the blood-brain barrier by its O-methylated metabolite (Reches, A., et al., Neurology, 32:887-888, 1982) or simply because there is less L-DOPA available to reach the brain (Nutt, J.G., Fellman, J.H., Clin. Neuropharmaco!., 7:35-49, 1984).
In recent years, the development of new inhibitors of the enzyme catechol-O-methyl traπsferase (COMT) has been accelerated by the hypothesis that inhibition of this enzyme may provide significant clinical improvements in patients afflicted by Parkinson's disease undergoing treatment with L-DOPA plus a peripheral AADC inhibitor. The rationale for the use of COMT inhibitors is based on their capacity to inhibit the O- methylation of L-DOPA to 3-O-methyl-L-Dopa. COMT inhibition slows elimination of L-DOPA from the plasma by increasing plasma half-life (increases area under the curve [AUC] without altering the time L-DOPA plasma to peak or the maximum concentration). Thus pharmacokinetic alterations may be an advantage over increasing the dose of L-DOPA, which also increases AUC, but additionally raises peak concentrations. In turn, raising peak concentrations relates to adverse effects such as dyskiπesia, which occurs immediately when COMT inhibitors are given but can be anticipated by either reducing the dose of L-DOPA or increasing the time intervals between doses. The effects of COMT inhibition also differ from those of coπtrolled-release L-DOPA formulation which slow down absorption and reduce bioavailability. The pharmacokinetic changes induced by COMT inhibition reduce the daily L- DOPA dose by enabling a reduction of each dose or an increase in dose intervals. With repeated doses of L-DOPA every 2-6 h in the presence of COMT inhibition, the mean plasma L-DOPA concentration is raised and the through concentrations are increased proportionally more than the peak concentrations despite a reduction in L-DOPA dose. As would be predicted by the slowed elimination of L-DOPA, the duration of antiparkinsonian action with single doses of L-DOPA is prolonged by COMT inhibition (Nutt, J.G., Lancet, 351 :1221-1222, 1998). The most potent and selective COMT inhibitors found so far are very active and do not interact with other enzymes, receptors, ionic channels or transporters up to very high doses. Some of them were demonstrated to have beneficial effects both in experimental models of parkinsonism and in Parkinson's disease patients. Other therapeutic applications of these COMT inhibitors have also been put forward, namely in the treatment of depression or anxiety, as gastroprotective drugs and as πatriuretic and antihypertensive agents.
The most potent COMT inhibitors thusfar reported, 3,4-dihydroxy-4'- methyl-5-nitrobeπzophenoπe (tolcapone, Australian Pat. AU-B-69764/87),
and (E)-2-cyaπo-N,N-diethyl-3-(3,4-dihydroxy-5-nitropheπyl)acryiamide (eπtacapone, German Pat. DE 3740383 A 1 ) have inhibition constants in the low πM range. Tolcapone differs from eπtacapoπe in being a more potent inhibitor of COMT in the periphery and furthermore at penetrating into the brain to inhibit brain COMT as well. It has not been established which of these two inhibitors is more useful in the treatment of Parkinson's disease. Compounds penetrating the blood-brain barrier may be assumed to be more effective as theoretically they might have additional benefits of decreasing dopamine methylation to 3- methoxytyramine and homovaπillic acid. Conversely, central inhibition may be unimportant if the more significant action is to protect L-DOPA from breakdown in the periphery. This distinction may have practical importance, as the use of COMT inhibitors which are excluded from the brain may avoid potential uπdesired CNS side effects of these agents.
In this respect, it is interesting to underline the lack of antiparkinsoπian action of tolcapone when given alone (Hauser, R.A., et al., Mov Disord, 1998, 13, 643-647), and the relatively frequent observations of increased central dopamiπergic stimulation, primarily dyskiπesia and confusion, in patients taking L-DOPA plus tolcapone (Nutt, J.G., Lancet, 351 :1221-
1222, 1998). This suggests that the central effects of COMT inhibition are very small when given alone, but when given with L-DOPA the risk of inhibition of brain COMT may be associated with the appearance of symptoms related to increased dopamiπergic stimulation which may require cessation of therapy.
Another potential problem with COMT inhibitors concerns their
relatively short half-life (tolcapone, 2 h [Dingemanse, J., et al., Gin.
Pharmacol. Ther., 57:508-517, 1995]; entacapone, 0.3 h [Keranen, T., et
al., Eur. J. Clin. Pharmacol., 46:151-157, 1994]). To circumvent this
problem both tolcapone and entacapone are recommended to be administered as frequently as 3 times a day; because the half-life of entacapone is considerably shorter than that of tolcapone, the recommended dose for entacapone is twice that for tolcapone.
As previously mentioned, the 3,4-dihydroxy-5-nitrophenyl group was
identified as an active pharmacophore and it was simultaneously
discovered that the presence of a carboπyl group (e.g. in tolcapone ) or
enone group (e.g. in entacapone) conjugated to the pharmacophore of
the molecule generally enhances inhibition of the COMT catalysed
transfer of the methyl group from the S-adeπosyl-L-methioniπe
coeπzyme to a substrate containing a catechol functional group. Among
many tested compounds bearing a 3,4-dihydroxy-5-nitrobenzoyl group,
the corresponding beπzophenones were recognized as the most potent
COMT inhibitors with ED50 < 1 mg/kg (rat, p.o.) (Borguiya J. et al.,
Helvetica Chimica Acta 72, 952-968, 1989).
Formation of homologues of known biologically active compounds as potentially improved drugs is a well known principle and is used mainiy
for optimisation of activity of structurally nonspecific drugs or for
achieving changes in predominant biological action in structurally
specific drugs (Korolkovas A. Essentials of Medicinal Chemistry, p. 76,
1988 by J.Wiley & Sons, Inc.). On the other hand, homologation is not
generally used nor expected to influence predictably the half-life of a
compound.
We have surprisingly proven that the next higher homologue of 3,4-
dihydroxy-5-nitrobenzophenoπe i.e the compound with one more
methylene group between the substituted benzoyl group and pheπyl
group is endowed with selective COMT inhibition of long duration and
that this effect is unique in a series of the higher homologues.
The invention relates to substituted 2-phenyl-1-(3,4-dihydroxy-5-
πitropheπyl)-1-ethanones of formula I
Figure imgf000008_0001
where R^ and R are hydrogens or groups hydrolysable under
physiological conditions, the same or different, and signify optionally
substituted lower alkaπoyl or aroyl, optionally substituted lower alkyl or
arylsulphoπyl or optionally substituted lower alkylcarbamoyi, or taken together signify a lower alkylidene or cycloalkylideπe group; R3, R and
R3 are the same or different and signify hydrogen, optionally substituted
saturated or partially unsaturated lower hydrocarbon residue, hydroxyl,
optionally substituted lower aikoxy or aryioxy group, optionally
substituted aryl, optionally substituted alkaπoyl or aroyl group, lower
alkanoylamino group, lower dialkanoylamino group, carboxyi, optionally
substituted lower alkyloxycarboπyl or aryloxycarbonyl group, optionally
substituted carbamoyl, halogen, nitro, amino, lower alkylamiπo or lower
dialkylamiπo or cyano group, or taken together signify aliphatic or
heteroaliphatic rings or aromatic or heteroaromatic rings, and
pharmaceutical acceptable salts thereof; to the use of the compounds for
prevention or treatment of certain pathological states in humans and to
the preparation of pharmaceutical compositions containing them.
The term "lower" denotes residues with a maximum of 8, preferentially
a maximum of 4 carbon atoms. The term "alkyl" taken alone or in
combination with terms such as "alkanoyl, alkoxycarboπyl, alkylidene,
cycloalkylidene, alkoxycarbonyloxy, alkylamiπo " denotes straight-chain
or branched saturated hydrocarbon residues. The term halogen denotes
fluorine, chlorine, bromine, and iodine. The term "aryl" denotes a
carbσcyciic aromatic group, preferably mono- or bicyclic groups.
For the preparation of pharmaceutical compositions of compounds of
formula I, inert pharmaceutically acceptable carriers are admixed with the active compounds. The pharmaceutically acceptable carriers may be
either solid or liquid. Solid form preparations include powders, tablets,
dispersibie granules and capsules. A solid carrier can be one or more
substances which may also act as diluents, flavouring agents,
solubilizers, lubricants, suspending agents, binders or tablet
disintegrating agents; it may also be an encapsulating material.
Preferably, the pharmaceutical preparation is in unit dosage form, e.g.
packaged preparation, the package containing discrete quantities of
preparation such as packeted tablets, capsules and powders in vials or
ampules.
The dosages may be varied depending on the requirement of the
patient, the severity of the disease and the particular compound being
employed. For convenience, the total daily dosage may be divided and
administered in portions throughout the day. Determination of the proper
dosage for a particular situation is within the skill of those in the medical
art.
Reference is now made to the accompanying drawings in which:
Fig. 1 is a graph showing brain COMT activity at different times after oral
administration of compound B (closed squares), entacapone (open
circles) or tolcapone (open squares).
Fig. 2. is a graph showing liver COMT activity at different times after oral
administration of compound B (closed squares), entacapone (open circles) or tolcapone (open squares).
Fig. 3 is a graph showing concentration dependent inhibition of brain
COMT activity at one hour after oral administration of compound B
(closed squares), entacapone (open circles) or tolcapone (open
squares).
Fig. 4 is a graph showing concentration dependent inhibition of liver
COMT activity at one hour after oral administration of compound B
(closed squares), entacapone (open circles) or tolcapone (open
squares).
Fig. 5 is a graph showing concentration-dependent amphetamine-
induced horizontal activity after oral administration of vehicle (open
columns), tolcapone (closed columns), entacapone (hatched columns)
and compound B (cross hatched columns).
Fig. 6 is a graph showing concentration-dependent amphetamine-
induced stereotypies after oral administration of vehicle (open columns),
tolcapone (closed columns), entacapone (hatched columns) and
compound B (cross hatched columns).
In Figs. 1 and 2 each point represents the mean of four to eight
experiments per group and vertical lines the respective SEM.
In Figs. 3 and 4 each point represents the mean of eight experiments per
group and vertical lines the respective SEM.
In Figs. 5 and 6 each column represents the mean of eight experiments per group and vertical lines the respective SEM.
Materials and Methods
Assay of COMT activity
Livers and brains from 60 day old male Wistar rats weighing 240-260 g
(Harlaπ-lnterfauπa Iberica, Barcelona, Spain), kept two per cage under controlled environmental conditions (12 h light/dark cycle and room temperature 24 °C) were used in all experiments. After decapitation, the organs were immediately removed and homogenised in 5 mM phosphate buffer of pH 7.8. COMT activity was evaluated by the ability to methylate adrenaline to metanephrine. Aliquots of 0.5 ml of liver and whole brain homogenates were preincubated for 20 min with 0.4 ml of phosphate buffer (5 mM); thereafter, the reaction mixture was incubated for 15 min with increasing concentrations of epinephriπe (0.1 to 2000 μM; 0.1 ml) in the presence of a saturating concentration of S-adeπosyl-L-methioniπe, the methyl donor (brain, 100 μM; liver, 500 μM); the incubation medium contained also pargyline (100 μM), MgCI2 (100 μM) and EGTA (1 mM). The preincubatioπ and incubation were carried out at 37°C under conditions of light protection with continuous shaking and without oxygenation.
In experiments conducted with the aim of studying the inhibitory effect of COMT inhibitors on enzyme activity, the reaction mixture was preincubated for 20 min with increasing concentrations of test compounds (0.5 to 1 ,000 nM); the incubation was performed in the presence of a concentration of adrenaline five times the corresponding Km value as determined in saturation experiments.
In experiments designed to evaluate the oral bioavailabiiity, half-life and brain access, test compounds were given by gastric tube to overnight fasted rats. Therafter, at defined intervals, animals were killed by decapitation and livers and brains removed and used to determine COMT activity as described above. At the end of the incubation period (brain, 15 min; liver, 5 min) the tubes were transferred to ice and the reaction was stopped by the addition of 200 μl of 2 M perchloric acid. The samples were then centrifuged (200xo\ 4 min, 4°C), and 500 μl aliquots of the supernatant, filtered on 0.22 μm pore size Spin-X filter tubes (Costar) were used for the assay of metaπephrine. The assay of metanephrine was carried out by means of high pressure liquid chromatography with electrochemical detection. The lower limits for detection of metanephrine ranged from 350 to 500 fmol (0.5 to 1.0 pmol/mg proteiπ/h).
Km and Vmaχ values for COMT activity were calculated from non-linear regression analysis using the GraphPad Prism statistics software package (Motulsky, H.G., et al., GraphPad Prisms, GraphPad Prism
Software Inc., San Diego, 1994). For the calculation of the !CS0 values, the parameters of the equation for one site inhibition were fitted to the experimental data. Geometric means are given with 95% confidence
limits and arithmetic means are given with S.E.M.. Statistical analysis
was performed by one-way analysis of variance (ANOVA) using Newman-Keuls multiple comparison test to compare values.
5 The protein content in the homogeπates was determined by the
method of Bradford (Bradford, M.M., Anal. Biochem., 72: 248-254, 1976)
with human serum albumin as standard. The protein content was similar
in all samples (approximately 5 mg/500 μl homogenate).
l o Beha vioural testing
The experimental design used in the present study was aimed at
determining the potentiatioπ of amphetamine-induced hyperactivity of
brain dopaminergic systems by COMT inhibitors. For this purpose 128
5 rats were divided into 16 groups, and were given the vehicle or one of
the three COMT inhibitors tested 6 hours prior to behavioural evaluation.
In all groups of rats, behavioural testing started 15 min after the s.c.
injection of vehicle or increasing doses of amphetamine (0.5, 2.0 or 4.0
mg/kg).
On the test day, 7 h before the experiment began, animals were
transferred to a dimly illuminated and sound attenuating room separate
from the animal colony room where the test cages were kept;
temperature and humidity were the same as in the colony room.
Spontaneous locomotor activity was measured using a San Diego Iπstruments rodent activity monitor (model Flex Field, San Diego Instruments, San Diego, CA) with 48 infrared motion sensors. The lower frame was 50.5 x 50.5 cm, with 32 photocells (separated by 2.5 cm) located lengthwise 5 cm above the floor. The upper frame was 50.5 x 50.5 cm, with 16 photocells (separated by 2.5 cm) located lengthwise 15 cm above the floor. The test field was an acrylic chamber with internal dimensions 40 x 40 x 37 cm. Ten-minute activity recording began immediately after placing the test subject at the centre of the chamber. Activity was measured automatically with a personal computer using
Flex Field software (San Diego Instruments) which provides user-defined intervals of total interruptions. Three parameters of normal spontaneous ' locomotion were recorded: horizontal activity, vertical activity and centre time. Stereotypical behaviour (intense sniffing, repetitive head and limb movements and licking and biting, as defined by Feldmaπ, R.S., Meyer, J.S., Quenzer, L.F., Principles of Neuropharmacology, 1997, Sinauer Associates, Inc. Publishers, Suπderiand, MA) were quantified by an independent observer after being recorded on tape by means of video tracking system (VP200, HVS Image, Ltd) placed 70 cm above the test field. Animals were habituated to the test field environment for one hour prior to behavioural testing.
Results
In vitro COMT inhibition studies
Incubation of liver and whole brain homogenates in the presence of increasing concentrations of adrenaline resulted in a concentration- dependent formation of metanephrine, yielding Km (in μM) and Vmaχ (in nmol mg protein*1 h'1) values of 0.7 (0.5, 0.9; 95% confidence intervals) and 1.31 ±0.02 for brain and 238.5 (128.5; 348.5) and 61.6+3.8 for liver, respectively. From these kinetic parameters, a saturating concentration of adrenaline was chosen to use in inhibition studies (liver, adrenaline = 1000 μM; brain, adrenaline = 100 μM). Compounds of formulae A - E,
Figure imgf000016_0001
plus entacapone and tolcapone (the reference compounds) produced a concentration-dependent decrease in the O-methylatioπ of adrenaline with ICso values in the low nM range for the brain and in the μM range for the liver (see table 1).
Table 1. ICso values (in nM) for inhibition of rat brain and liver COMT.
Figure imgf000016_0002
Compouπds of formulae A - E were found to be potent inn: both brain and liver COMT, the maximal inhibitory effect being a within 30 min after their oral administration (table 2). Compc A presented a similar inhibitory profile in brain and liver COMT, whereas compound E was much more potent upon liver COMT than brain COMT. Similarly, compound B was also much more potent as a peripheral COMT inhibitor than in brain. Compounds with longer carbon chains were less potent at inhibiting brain COMT in comparison with their effects upon liver COMT. This difference may have to do with difficulties in access to brain. Compounds with short carbon chains (A, B and C) were not equally potent at inhibiting peripheral and central COMT, but this difference was not so striking as observed with compounds with long carbon chains. When looking at the duration of inhibitory effect upon liver COMT it became evident that compound B (2 carbon chain) was a particularly long acting compound. Notably, inhibition of liver COMT by this compound at 9 h after oral administration almost achieved 70% inhibition, whereas compounds with shorter and longer carbon chains were not endowed of such a long acting effect. Tolcapone at 6 h and 9 h after administration produced marked inhibition in brain and liver COMT. As shown in figures 1 and 2, nine hours after administration, compound
B and tolcapone were equally potent at inhibiting liver COMT, whereas entacapone was almost devoid of COMT inhibitory properties. On the other hand, compound B and entacapone were much less potent than tolcapone at inhibiting brain COMT.
Table 2. Percent inhibition of COMT activity by compounds A - E, entacapone (Enta) and tolcapone (Tolc) in homogenates of rat brain and liver, determined at 0.5, 1 , 3, 6 and 9 h after their administration by gastric tube. Results are means±S.E.M. of 4 experiments per group.
Brain % inhibition
Time course
0.5 h 1 h 3 h 6 h 9 h
A 96.3±0.4 96.8±0.3 97.0±0.3 85.8±7.5 34.9±6.0
B 83.6+1.3 80.9±2.7 65.0±3.9 31.5±3.2 21.9+2.7
C 89.9±0.7 86.2±0.5 59.8±5.8 33.4±7.0 0.4±5.3
D 85.1+1.7 69.3±5.1 33.5±4.4 26.7±4.0 12.2±5.8
E 87.4±1.3 74.2±4.2 25.0±3.1 -5.6+7.5 -6.7±5.0
Enta 71.7±7.0 44.8±7.0 30.1±6.4 19.9+7.1 22.8±3.4 Tolc 98.9+0.1 98.7±0.2 97.0±0.5 85.8±8.2 77.5±1.8
Liver % inhibition
Time course
0.5 h 1 h 3 h 6 h 9 h
A I 99.0±0.2 98.7±0.2 96.9±2.5 80.5±7.3 31.7±5.5
B 98.6±0.4 96.7±1.7 96.2±0.8 75.9±4.2 69.8±3.6
C 98.4±0.3 97.8±0.2 95.0±0.7 70.8±12.8 39.9±11.1
D 97.5±0.1 95.3±0.8 67.5±7.8 52.0±9.5 39.0±13.2
E 99.2+0.1 98.9±0.3 88.1±3.9 36.0±5.3 -4.0+8.0
Enta 98.2±0.3 96.2±1.1 85.9±2.2 73.6±5.4 24.7±7.9
Tolc 100.0+0.0 99.9±0.1 98. O±O.7 94.1±0.3 67.0±4.0 Compounds F - J (see below) were also tested at 6 h and 9 h after administration and found to produce an inhibitory profile similar to that described for compound B. (Table 3).
Figure imgf000019_0001
Figure imgf000019_0002
The potency of compound B, tolcapone and entacapone at inhibiting brain and liver COMT was evaluated in experiments in which rats were given increasing doses of the compounds under test (0.3 to 30 mg/kg). In these experiments rats were killed 1 h after the administration of the compounds (at twax) and COMT activity determined as described above. The results obtained are shown in figures 3 and 4 and indicate that compound B and tolcapone were equally potent at inhibiting liver COMT with ED50's of 0.7±1.1 and 0.7±0.1 mg/kg, respectively; entacapone was slightly less potent with a ED50 value of 1.9±0.2 mg/kg,. However, compound B was less potent than tolcapone at inhibiting brain COMT with ED5o's of 5.3±1.1 and 1.6±0.1 mg/kg, respectively. At the highest dose tested (30 mg/kg), entacapone failed to reach the 50% inhibition level.
Table 3. Percent inhibition of COMT activity by compounds F - J in homogenates of rat brain and liver, determined at 6 and 9 h after their administration by gastric tube. Results are means±S.E.M. of 4 experiments per group.
Liver I Brain
6 h 9 h 6 h 9 h
F I 70.2±3.3 37.7±4.2 I 10.3+5.1 0.4+7.1
G 77.8±4.5 51.2±3.7 28.2±5.1 27.1±5.0
H 82.8±2.2 45.8±10.3 17.1±4.0 6.8±2.7
I 74.0±4.6 46.3±10.6 33.0±2.0 24.6±6.8
J 68.6±4.2 57.1±8.1 12.1±3.2 25.8±2.1 Behavioural testing
Amphetamine is a potent psychostimulant that depending on the dose administered produces increased locomotor behaviour and various stereotypical activities. A single low dose of amphetamine administered to rats leads to a characteristic response pattern consisting of increased locomotor activity, rearing, mild sniffing and head bobbing. Gradually increasing the dose of amphetamine results in a decrease in locomotion and rearing, which are replaced by focused stereotypies (repetitive, seemingly aimless behaviours performed in a relatively invariant manner) confined to a small area of the cage floor (Feldman, R.S., Meyer, J.S., Quenzer, L.F., Principles of Neuropharmacology, 1997, Sinauer Associates, inc. Publishers, Sunderlaπd, MA). The cerebral dopamiπergic system has traditionally been crucial to the ability of amphetamine to stimulate locomotor activity and stereotypical behaviours. With respect to the anatomic substrates of amphetamine action, there is evidence that stimulation of dopaminergic activity in the nucleus accumbens is responsible for amphetamine-induced locomotor activity, whereas stimulation of dopamiπergic activity in the caudate- putamen is linked with focused stereotypies produced by high doses of
amphetamine.
As predicted, low doses of amphetamine (0.5 and 2.0 mg/kg, s.c.) were found to produce dose-dependent increases in horizontal activity and rearing, with no evidence of stereotyped behavior (Figures 5 and 6). By contrast, a high dose of amphetamine (4.0 mg/kg, s.c.) was found to produce no further increase in locomotor activity, but resulted in the appearance of stereotypies which lasted for 250 s during the 600 s observation period. Tolcapone (30 mg/kg, p.o.) administered 6 h before amphetamine challenge was found to significantly increase locomotor activity in rats treated with 0.5 and 2.0 mg/kg amphetamine. By contrast, in rats given 4.0 mg/kg amphetamine, tolcapone produced a marked decrease in locomotor activity and increased two-fold the duration of stereotyped behavior. Rats treated with entacapone (30 mg/kg, p.o.) or compound B six hours before amphetamine challenge presented the same pattern of locomotor activitiy and stereotyped behaviour as their corresponding controls.
Conclusion
Compounds of formula I are very potent catechol-O-methyltraπsferase
(COMT) inhibitors and have potentially valuable pharmaceutical
properties in the treatment of some central and peripheral nervous
system disorders where inhibition of O-methylatioπ of catεcholamines
may be of therapeutical benefit, such as Parkinson' disease and
parkiπsoπiaπ disorders, gastrointestinal disturbances, edema formation
states and hypertension. The possibility to use a long acting COMT
inhibitor with limited access to the brain, such as compound B, opens new perspectives in said therapies by improving selectivity and prolong
COMT inhibition. This is particularly important when thinking of treating
patients afflicted by Parkinson's disease and taking L-DOPA plus a
peripheral AADC inhibitor. Due to the possibility that COMT inhibitors
which have easy access to the brain may cause excessive dopamiπergic
stimulation, namely by inducing dyskiπesia and mental confusion in L-
DOPA treated patients, the use of a substance such as compound B is
expected to be devoid of such effects yet possessing the benefits of a
long acting substance.
The invention disclosed herein is exemplified by the following
examples of preparation, which should not be construed to limit the
scope of the disclosure. Alternative pathways and analogous structures
may be apparent to those skilled in the art.
Example 1 1 -(3,4-dihydroxy-5-nitropheπyl)-2-phenyl-1 -ethanoπe
A solution of 20 g (82.64 mmol) of O-benzylvaπillin in 200 mL of dry tetrahydrofuraπ was slowly added to a stirred solution of benzyl magnesium chloride (103.30 mmol) in 150 mL of diethylether at 10° C over 20 min, and the reaction mixture was then boiled for 10 min, cooled, quenched with a mixture of ice and dilute hydrochloric acid and evaporated at reduced pressure. The residue was dissolved in dichloromethane, the solution washed with brine, dried with sodium sulphate and the solvent was evaporated under reduced pressure leaving
a crystalline residue that was recrystallised from diethylether and petroleum ether. 1 -(4-Beπzyloxy-3-methoxypheπyl)-2-pheπyl-1 -ethanol was obtained as white crystals, m.p. 97 to 98° C.
A solution of 10g (30 mmol) of the above secondary alcohol in 90 mL of dichloromethane and 30 mL of diethylether was cooled to 0° C and 7.5 g of CeliteR was added at once with stirring, followed by 9 g (90 mmol) of chromium trioxide. The reaction mixture was stirred overnight at room temperature, filtered, and the filtrate was evaporated at reduced pressure.
The crystalline residue was recrystallised from a mixture of dichloromethane and diethylether, yielding 1-(4-beπzyloxy-3- methoxyphenyl)-2-phenyl-1 -ethaπoπe as white crystals, m.p 134 to 135° C.
A solution of 5.9 g, (17.8 mmol) of the above ketone in a mixture of dichloromethane (60 mL) and 30% hydrobromic acid in acetic acid (27 mL) was stirrred for 1.5 h at room temperature and then the dichloromethane was evaporated at reduced pressure and the reaction mixture was poured onto 200 mL of an ice/water mixture. The precipitate formed was filtered off, and dried under vacuum to provide 1-(4-hydroxy- 3-methoxypheπyl)-2-phenyl-1-ethaπone as beige crystals, m.p. 107 to 108° C.
To a solution of 3.87 g (16 mmol) of the above intermediate in 40 mL of acetic acid there was added 1.4 mL (17.6 mmol) of 12.6 M nitric acid under cooling to 10° C and the reaction mixture was stirred for 30 min at room temperature and then poured over an ice/water mixture. The precipitate formed was filtered off, washed with water and dried giving 1- (4-hydroxy-3-methoxy-5-nitrophenyl)-2-phenyl-1-ethanoπe as a yellow powder m.p.129 to 130° C.
The above πitroderivative (3.76g, 13 mmol) was boiled with a mixture of azeotropic hydrobromic acid (37 mL) and 30% HBr in acetic acid (18 mL) for 16 hours and the cooled reaction mixture was poured onto a mixture of ice/water. The precipitate formed was filtered off, washed thoroughly with water and recrystallised from acetic acid to give the desired product as yellow crystals m.p. 181 to 182° C.
Examples 2-12
By the application of the above described technique and related procedures known to those skilled in the art, and using appropriate metalorgaπic reagents the following compounds were prepared:
1-(3,4-dihydroxy-5-nitropheπyl)-2-(4-hydroxyphenyl)-1-ethanone 1-(3,4-dihydroxy-5-πitropheπy!)-2-(2-methylpheπyl)-1 -ethanone' 1 -(3,4-dihydroxy-5-nitrophenyl)-2-(3-methylpheπyf)-1 -ethaπone 1 -(3,4-dihydroxy-5-nitrophenyl)-2-(4-methylphenyl)-1 -ethanone 1 -(3,4-dihydroxy-5-nitrophenyl)-2-(4-butylphenyl)-1 -ethaπone
1-(3,4-dihydroxy-5-πitropheπyl)-2-(3,4-dimethylphenyl)-1 -ethanone 1-(3,4-dihydroxy-5-nitropheπyl)-2-(3,4-dimethoxypheπyl)-1 -ethanone 1 -(3,4-dihydroxy-5-πitrophenyl)-2-(4-butyloxypheπyl)-1 -ethanone 1 -(3,4-dihydroxy-5-nitropheπyl)-2-(1 -methyl-5-indolyl)-1 -ethaπone
1-(3,4-dihydroxy-5-nitrophenyl)-2-(3,4-methylenedioxyphenyl)-1 -ethanone 1-(3,4-dihydroxy-5-nitrophenyl)-2-(2,4f6-trimethylpheπyl)-1 -ethaπone
Example 13 1 -(3,4-dihydroxy-5-nitrophenyl)-2-(2-methylpheπyl)-1 -
ethanone
To a mixture of guaiacol (1.24 g, 10 mmol), o-tolylacetic acid (1.50 g, 10 mmol), and ZπCI2 (5 g,36.7 mmol) there was added POCI3 (15 mL, 161 mmol) and the resulting suspension was stirred and heated to 80° C for 1.5 h. The reaction mixture was cooled and poured onto ice/water and the resulting suspension was stirred at room temperature for 1 h and then extracted with ethyl acetate. The organic layer was separated, washed with brine and dried with sodium sulphate. Volatiles were evaporated under reduced pressure and the residue was dissolved in diethylether. The solution was extracted twice with 50 mL of 2N aqueous solution of NaOH and the combined aqueous layers were combined and acidified with hydrochloric acid to pH=2. The emulsion formed was extracted by ethyl acetate, and the organic layer was washed with brine, dried, and the solvent was evaporated under diminished pressure. The residue was chromatographed on a column of silica gel with a mixture of petroleum ether and ethyl acetate to give 1-(4-hydroxy-3- methoxyphenyl)-2-(2-methylpheπyl)-1 -ethanone as off white crystals, m.p. 79 to 81° C. To a solution of 4.01 g (16 mmol) of the above intermediate in 40 mL of acetic acid, there was added 1.4 mL (17.6 mmol) of 12.6 M nitric acid under cooling to 10° C and the reaction mixture was stirred for 30 min at room temperature and then poured over an ice/water mixture. The precipitate formed was filtered off, washed with water and dried giving 1- (4-hydroxy-3-methoxy-5-nitropheπyl)-2-(2-methylphenyl)-1 -ethaπone as a
yellow powder m.p.150 to 151° C.
The above nitroderivative (3.91 g, 13 mmol) was boiled with a mixture of azeotropic hydrobromic acid (37 L) and 30% HEr in acetic acid (18 mL) for 16 hours and the cooled reaction mixture was poured onto a mixture of ice/water. The precipitate formed was filtered off, washed thoroughly with water and recrystallized from acetic acid to give the desired product as yellow crystals m.p. 128 to 129° C.
Examples 14-21
By the application of the above described technique and related procedures known to those skilled in the art, and using appropriately substituted phenylacetic acids the following compounds were prepared:
1-(3,4-dihydroxy-5-πitropheπyl)-2-(4-carboxyphenyl)-1 -ethanone 1-(3,4-dihydroxy-5-nitrophenyl)-2-(2-nitrophenyl)-1 -ethanone 1 -(3,4-dihydroxy-5-πitrophenyl)-2-(4-bipheπyl)-1 -ethanone 1-(3,4-dihydroxy-5-πitropheπyl)-2-(3-cyaπophenyl)-1 -ethanone 1 -(3,4-dihydroxy-5-nitrophenyI)-2-(1 -naphthyl)-1 -ethanone 1 -(3,4-dihydroxy-5-nitrophenyl)-2-(2-πaphthyl)-1 -ethanone 1 -(3,4-dihydroxy-5-nitrophenyl)-2-(2-chlorophenyl)-1 -ethanone 1 -(3,4-dihydroxy-5-πitrophenyl)-2-(4-chlorophenyl)-1 -ethanone
Example 22 1 -(3,4-diacetoxy-5-nitrophenyl)-2-pheπyl-1 -ethanone
A suspension of 9.20 g (33.6 rπmol) of 1-(3,4-dihydroxy-5-
nitropheπyl)-2-phenyl-1 -ethanone in 90 mL of dichloromethane was treated with 7.85 g (lOOmmol) of acetylchloride, 7.51 g (95 mmol) of pyridine, and a catalytic amount of 4-dimethylamiπopyridine. After 1 h of stirring at room temperature the solution formed was washed successively with ice-cold 0.2N hydrochloric acid, 1 % aqueous solution of sodium bicarbonate and brine. The dried (Na2S04) solution was evaporated under reduced pressure, and the residue recrystallised from a mixture of diethylether and petroleum ether giving the desired product as yellow crystals, m.p. 94 to 95° C.
Examples 23-27
By the application of the above described technique and related procedures known to those skilled in the art, and using appropriately substituted 1-(3,4-dihydroxy-5-nitrophenyl)-2-phenyl-1-ethanoπes and halogenides or anhydrides of acids, the following compounds were prepared:
1 -(3,4-dimethoxymethyloxy-5-nitrophenyl)-2-phenyl-1 -ethaπone 1-(3,4-dibutyryloxy-5-nitrophenyl)-2-phenyl-1 -ethaπone -(3,4-di(4-tolylsulphonyloxy)-5-nitrophenyl)-2-phenyl-1 -ethaπone -(3,4-dibutyryloxycarbonyloxy-5-nitrophenyl)-2-phenyl-1-ethaπone -(3,4-diacetoxy-5-πitrophenyl)-2-(4-acetoxyphenyl)-1-ethaπσπe

Claims

Claims
1. A compound of formula
Figure imgf000030_0001
where Ri and R2 are the same or different and signify hydrogens or
groups hydrolysable under physiological conditions, optionally
substituted lower alkanoyl or aroyl, optionally substituted lower alkyl or
arylsulphonyl or optionally substituted lower alkylcarbamoyl, or taken
together signify a lower alkylidene or cycloalkylidene group; R3, R4 and
R5 are the same or different and signify hydrogen, optionally substituted
saturated or partially unsaturated lower hydrocarbon residue, hydroxyl,
optionally substituted lower alkoxy or aryloxy group, optionally
substituted aryl, optionally substituted alkanoyl or aroyl group, lower
alkaπoylamino group, lower dialkanoyfamiπo group, carboxyl, optionally
substituted lower alkyloxycarbonyl or aryloxycarbonyl group, optionally
substituted carbamoyl, halogen, nitro, amino, lower alkylamiπo or lower
dialkylamino or cyaπo group, or taken together signify aliphatic or
heteroaliphatic rings or aromatic or heteroaromatic rings, and pharmaceutically acceptable salts thereof.
2. A compound according to claim 1 , comprising: 1-(3,4-dihydroxy-5- nitropheπyl)-2-phenyl-1 -ethanone; 1 -(3,4-dihydroxy-5-nitrophenyl)-2-(2- methylpheπyl)-1 -ethanone; 1-(3,4-dihydroxy-5-πitrophenyl)-2-(4- chloropheπyl)-1 -ethanone; 1-(3,4-dihydroxy-5-πitropheπyl)-2-(1- naphthyl)-1-ethanoπe; 1-(3,4-dihydroxy-5-nitrophenyl)-2-(2-naphthyl)- 1 -ethanone or 1-(3,4-dihydroxy-5-nitropheπyl)-2-(4-biphenyl)-1- ethaπone.
3. A method of treating a subject afflicted by some central and peripheral nervous system disorders, where a reduction in the O-methylation of catecholamines may be of therapeutical benefit, such as Parkinson's disease and parkinsonian disorders, gastrointestinal disturbances, edema formation states and hypertension, which comprises administering to the subject an amount of a compound according to claim 1 or 2 effective to treat said diseases in the subject.
4. A pharmaceutical composition comprising a therapeutically effective amount of a compound according to claim 1 or 2 in combination with a
pharmaceutically acceptable carrier.
5. The use of a compound according to claim 1 or 2 in the manufacture of a medication for treating a subject afflicted by central or peripheral nervous system disorders.
6. The use of a compound according to claim 1 or 2 in the manufacture of a medication for treating Parkinson's disease and parkinsonian disorders, gastrointestinal disturbances, edema formation states and
hypertension.
7. The use of a compound according to claim 1 or 2 in therapy.
PCT/GB1999/004310 1998-12-18 1999-12-17 Substituted 2-phenyl-1-(3,4-dihydroxy-5-nitrophenyl)-1-ethanones, their use in the treatment of some central and peripheral nervous system disorders and pharmaceutical compositions containing them WO2000037423A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
BRPI9908084-2A BR9908084B1 (en) 1998-12-18 1999-12-17 2-phenyl-1- (3,4-dihydroxy-5-nitrophenyl) -1-ethanone compound, pharmaceutical composition, and use of a compound.
AU18731/00A AU754113B2 (en) 1998-12-18 1999-12-17 Substituted 2-phenyl-1-(3,4-dihydroxy-5-nitrophenyl)-1-ethanones, their use in the treatment of some central and peripheral nervous system disorders and pharmaceutical compositions containing them
HU0100786A HU226396B1 (en) 1998-12-18 1999-12-17 Substituted 2-phenyl-1-(3,4-dihydroxy-5-nitrophenyl)-1-ethanones, and pharmaceutical compositions therof for use in the treatment of some central and peripheral nervous system disorders
PL99342542A PL193998B1 (en) 1998-12-18 1999-12-17 Substituted 2-phenyl-1-(3,4-diphydroxy-5-nitrophenyl)-ethanones

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB9827996.1 1998-12-18
GB9827996A GB2344819A (en) 1998-12-18 1998-12-18 2-Phenyl-1-(3,4-dihydroxy-5-nitrophenyl)-1-ethanones

Publications (1)

Publication Number Publication Date
WO2000037423A1 true WO2000037423A1 (en) 2000-06-29

Family

ID=10844561

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB1999/004310 WO2000037423A1 (en) 1998-12-18 1999-12-17 Substituted 2-phenyl-1-(3,4-dihydroxy-5-nitrophenyl)-1-ethanones, their use in the treatment of some central and peripheral nervous system disorders and pharmaceutical compositions containing them

Country Status (20)

Country Link
US (1) US6512136B1 (en)
EP (1) EP1010688B1 (en)
CN (1) CN1173926C (en)
AR (1) AR021893A1 (en)
AT (1) ATE236870T1 (en)
AU (1) AU754113B2 (en)
BR (1) BR9908084B1 (en)
CA (1) CA2292968C (en)
CZ (1) CZ297919B6 (en)
DE (1) DE69906671T2 (en)
DK (1) DK1010688T3 (en)
ES (1) ES2197583T3 (en)
GB (1) GB2344819A (en)
HU (1) HU226396B1 (en)
PL (1) PL193998B1 (en)
PT (1) PT1010688E (en)
RU (1) RU2232748C2 (en)
SI (1) SI1010688T1 (en)
TR (1) TR200003019T1 (en)
WO (1) WO2000037423A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011107653A2 (en) 2010-03-04 2011-09-09 Orion Corporation Method for treating parkinson's disease
EP2543368A1 (en) 2007-12-11 2013-01-09 Viamet Pharmaceuticals, Inc. Metalloenzyme inhibitors using metal binding moieties in combination with targeting moieties
US8536203B2 (en) 2006-04-10 2013-09-17 Bial-Portela & Ca, S.A. Pharmaceutical compounds
US9132094B2 (en) 2009-04-01 2015-09-15 Bial—Portela & Ca, S.A. Pharmaceutical formulations comprising nitrocatechol derivatives and methods of making thereof
US9458128B2 (en) 2012-05-24 2016-10-04 Orion Corporation Catechol O-methyltransferase activity inhibiting compounds
US9550759B2 (en) 2005-07-26 2017-01-24 Bial—Portela & Ca, S.A. Nitrocatechol derivatives as COMT inhibitors
US9630955B2 (en) 2011-12-13 2017-04-25 BIAL—Portela & Cª., S.A Chemical compound useful as intermediate for preparing a catechol-O-methyltransferase inhibitor
US9745290B2 (en) 2007-01-31 2017-08-29 Bial—Portela & Ca, S.A. Dosage regimen for COMT inhibitors
US9845316B2 (en) 2008-03-17 2017-12-19 BIAL—Portela & CA., S.A. Crystal forms of 5-[3-(2,5-dichloro-4, 6-dimethyl-1-oxy-pyridine-3-yl)[1,2,4]oxadiazol-5-yl]-3-nitrobenzene-1,2-diol
US10065944B2 (en) 2011-02-11 2018-09-04 Bial-Portela & Ca, S.A. Administration regime for nitrocatechols
WO2019129121A1 (en) * 2017-12-28 2019-07-04 Rpxds Co., Ltd Derivatives of phenylmethanone as fto inhibitors
US10357468B2 (en) 2014-11-28 2019-07-23 Bial—Portela & Ca, S.A. Medicaments for slowing Parkinson's disease

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0015228D0 (en) * 2000-06-21 2000-08-16 Portela & Ca Sa Substituted nitrated catechols, their use in the treatment of some central and peripheral nervous system disorders
GB2377934B (en) * 2001-07-25 2005-01-12 Portela & Ca Sa Method for the nitration of phenolic compounds
FI20012242A0 (en) * 2001-11-19 2001-11-19 Orion Corp New pharmaceutical compounds
WO2004100929A1 (en) 2003-05-12 2004-11-25 Synergia Pharma, Inc. Threo-dops controlled release formulation
US8158149B2 (en) * 2004-05-12 2012-04-17 Chelsea Therapeutics, Inc. Threo-DOPS controlled release formulation
US20070010584A1 (en) * 2003-09-04 2007-01-11 Peroutka Stephen J Compositions and methods for orthostatic intolerance
US20060241183A1 (en) * 2004-09-28 2006-10-26 Farouk Karoum Compositions and methods of using D-DOPA to treat Parkinson's disease
MY148644A (en) * 2005-07-18 2013-05-15 Orion Corp New pharmaceutical compounds
EP1764095A1 (en) * 2005-09-20 2007-03-21 Revotar Biopharmaceuticals AG Novel nitrocatechol derivatives having selectin ligand activity
US20090012177A1 (en) * 2006-03-23 2009-01-08 Rahim Shafa Treatment of psychiatric disorders using entacapone, tolcapone and other COMT inhibitor or MB-COMT inhibitor drugs
EP2363123A1 (en) * 2006-06-28 2011-09-07 Chelsea Therapeutics, Inc. Pharmaceutical compositions comprising droxidopa
ES2480966T3 (en) * 2007-03-09 2014-07-29 Chelsea Therapeutics, Inc. Droxidopa and its pharmaceutical composition for the treatment of fibromyalgia
CA2686723A1 (en) * 2007-05-07 2008-11-13 Chelsea Therapeutics, Inc. Droxidopa and pharmaceutical composition thereof for the treatment of mood disorders, sleep disorders, or attention deficit disorders
WO2009108077A2 (en) * 2008-02-28 2009-09-03 Bial - Portela & Ca., S.A. Pharmaceutical composition for poorly soluble drugs
AU2010231962B2 (en) * 2009-04-01 2015-05-21 Bial - Portela & Ca., S.A. Pharmaceutical formulations comprising nitrocatechol derivatives and methods of making the same
UY32656A (en) * 2009-05-27 2010-12-31 Sanofi Aventis PROCEDURE TO PRODUCE BENZOFURANS
JP5880913B2 (en) 2011-05-17 2016-03-09 三郎 佐古田 Treatment for trunk symptoms (postural reflex abnormalities) in Parkinson's disease
EP2809315A1 (en) 2012-01-31 2014-12-10 Lundbeck Na Ltd Improving postural stability administering droxidopa
WO2014147464A2 (en) * 2013-03-20 2014-09-25 Ra Chem Pharma Limited Novel process for the preparation of tolcapone
EP3972971A1 (en) * 2019-05-21 2022-03-30 H. Lundbeck A/S New catecholamine prodrugs for use in the treatment of parkinson's diseases

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0237929A1 (en) * 1986-03-11 1987-09-23 F. Hoffmann-La Roche Ag 3,5-Disubstituted pyrocatechol derivatives
FR2607493A1 (en) * 1986-11-28 1988-06-03 Orion Yhtymae Oy NOVEL PHARMACOLOGICALLY ACTIVE COMPOUNDS, PROCESSES FOR PREPARING THEM AND COMPOSITIONS CONTAINING SAME

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0237929A1 (en) * 1986-03-11 1987-09-23 F. Hoffmann-La Roche Ag 3,5-Disubstituted pyrocatechol derivatives
FR2607493A1 (en) * 1986-11-28 1988-06-03 Orion Yhtymae Oy NOVEL PHARMACOLOGICALLY ACTIVE COMPOUNDS, PROCESSES FOR PREPARING THEM AND COMPOSITIONS CONTAINING SAME

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BAECKSTROEM R ET AL: "SYNTHESIS OF SOME NOVEL POTENT AND SELECTIVE CATECHOL O-METHYLTRANSFERASE INHIBITORS", JOURNAL OF MEDICINAL CHEMISTRY,US,AMERICAN CHEMICAL SOCIETY. WASHINGTON, vol. 32, no. 4, 1 January 1989 (1989-01-01), pages 841 - 846, XP002012888, ISSN: 0022-2623 *

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9550759B2 (en) 2005-07-26 2017-01-24 Bial—Portela & Ca, S.A. Nitrocatechol derivatives as COMT inhibitors
US10336740B2 (en) 2005-07-26 2019-07-02 Bial—Portela & Ca, S.A. Nitrocatechol derivatives as COMT inhibitors
US9446012B2 (en) 2006-04-10 2016-09-20 Bial—Portela & Ca, S.A. Pharmaceutical compounds
US8536203B2 (en) 2006-04-10 2013-09-17 Bial-Portela & Ca, S.A. Pharmaceutical compounds
US9745290B2 (en) 2007-01-31 2017-08-29 Bial—Portela & Ca, S.A. Dosage regimen for COMT inhibitors
EP2543368A1 (en) 2007-12-11 2013-01-09 Viamet Pharmaceuticals, Inc. Metalloenzyme inhibitors using metal binding moieties in combination with targeting moieties
US9845316B2 (en) 2008-03-17 2017-12-19 BIAL—Portela & CA., S.A. Crystal forms of 5-[3-(2,5-dichloro-4, 6-dimethyl-1-oxy-pyridine-3-yl)[1,2,4]oxadiazol-5-yl]-3-nitrobenzene-1,2-diol
US10071085B2 (en) 2009-04-01 2018-09-11 Bial—Portela & Ca, S.A. Pharmaceutical formulations comprising nitrocatechol derivatives and methods of making thereof
US9132094B2 (en) 2009-04-01 2015-09-15 Bial—Portela & Ca, S.A. Pharmaceutical formulations comprising nitrocatechol derivatives and methods of making thereof
US10583130B2 (en) 2009-04-01 2020-03-10 Bial-Portela & Ca, S.A. Pharmaceutical formulations compromising nitrocatechol derivatives and methods of making thereof
WO2011107653A2 (en) 2010-03-04 2011-09-09 Orion Corporation Method for treating parkinson's disease
US10857120B2 (en) 2010-03-04 2020-12-08 Orion Corporation Use of levodopa, carbidopa and entacapone for treating Parkinson's disease
US11771675B2 (en) 2010-03-04 2023-10-03 Orion Corporation Use of levodopa, carbidopa and entacapone for treating Parkinson's disease
US10065944B2 (en) 2011-02-11 2018-09-04 Bial-Portela & Ca, S.A. Administration regime for nitrocatechols
US9630955B2 (en) 2011-12-13 2017-04-25 BIAL—Portela & Cª., S.A Chemical compound useful as intermediate for preparing a catechol-O-methyltransferase inhibitor
US9458128B2 (en) 2012-05-24 2016-10-04 Orion Corporation Catechol O-methyltransferase activity inhibiting compounds
US10357468B2 (en) 2014-11-28 2019-07-23 Bial—Portela & Ca, S.A. Medicaments for slowing Parkinson's disease
WO2019129121A1 (en) * 2017-12-28 2019-07-04 Rpxds Co., Ltd Derivatives of phenylmethanone as fto inhibitors

Also Published As

Publication number Publication date
ATE236870T1 (en) 2003-04-15
GB2344819A (en) 2000-06-21
PL193998B1 (en) 2007-04-30
CA2292968C (en) 2009-05-19
AU754113B2 (en) 2002-11-07
HUP0100786A2 (en) 2001-08-28
CZ20003018A3 (en) 2000-12-13
SI1010688T1 (en) 2003-10-31
PT1010688E (en) 2003-08-29
BR9908084A (en) 2000-10-24
DE69906671D1 (en) 2003-05-15
ES2197583T3 (en) 2004-01-01
RU2232748C2 (en) 2004-07-20
DK1010688T3 (en) 2003-08-04
AR021893A1 (en) 2002-08-07
CA2292968A1 (en) 2000-06-18
DE69906671T2 (en) 2004-03-04
TR200003019T1 (en) 2001-02-21
CN1296471A (en) 2001-05-23
HUP0100786A3 (en) 2002-12-28
GB9827996D0 (en) 1999-02-10
US6512136B1 (en) 2003-01-28
AU1873100A (en) 2000-07-12
EP1010688A1 (en) 2000-06-21
HU226396B1 (en) 2008-11-28
CZ297919B6 (en) 2007-04-25
CN1173926C (en) 2004-11-03
EP1010688B1 (en) 2003-04-09
PL342542A1 (en) 2001-06-18
BR9908084B1 (en) 2009-12-01

Similar Documents

Publication Publication Date Title
AU754113B2 (en) Substituted 2-phenyl-1-(3,4-dihydroxy-5-nitrophenyl)-1-ethanones, their use in the treatment of some central and peripheral nervous system disorders and pharmaceutical compositions containing them
DE60215919T2 (en) 4-FLUORO-N-INDAN-2-YL BENZAMIDE AND ITS USE AS A MEDICAMENT
US4329495A (en) Enkephalinase enzyme inhibiting compounds
PL154186B1 (en) Method for manufacturing arylic derivatives of the hydroxamic acid
US11701370B2 (en) Phosphonium ion channel blockers and methods for use
EP0858443A1 (en) 4-phenyl-4-oxo-butanoic acid derivatives with kynurenine-3-hydroxylase inhibiting activity
JPS6033823B2 (en) Salicylanilide derivative and method for producing the same
US5110831A (en) Vinylogous hydroxamic acids and derivatives thereof as 5-lipoxygenase inhibitors
US4124725A (en) Anti-inflammatory method
CS267551B1 (en) Pharmaceutical agents for asthma treatment and method of effective substance production
EP0646578B1 (en) Phenyl carboxamide-isoxazole-derivatives and salts, process for their preparation, their use as pharmaceuticals, and pharmaceutical compositions containing them
JP2003055214A (en) 2-phenyl-1-(3,4-dihydroxy-5-nitrophenyl)-1-ethanone- substituted compound, method for using the same in treating central and peripheral nervous disorder, and pharmaceutical composition including the same
KR20060088885A (en) Treating neuropathic pain with neuropeptide ff receptor 2 agonists
JPS627182B2 (en)
JPH0730024B2 (en) Novel aminoalkyl pyridine amide compound
US4898887A (en) Compounds and compositions having anti-inflammatory and analgesic activity
US4244970A (en) Method of treating inflammation
MXPA00008048A (en) Substituted 2-phenyl-1-(3,4-dihydroxy-5-nitrophenyl)-1-ethanones, their use in the treatment of some central and peripheral nervous system disorders and pharmaceutical compositions containing them
Creveling et al. The Depletion of Norepinephrine-3H from Heart by α-Methyl-m-tyrosine. A Novel and Convenient Method for Assaying the Inhibition of Aromatic Amino Acid Decarboxylase in Vivo
IE60234B1 (en) Therapeutic compositions containing benzhydrylthiomethane derivatives
US4165383A (en) Anti-inflammatory method
Rooks et al. The anti-inflammatory and analgesic profile of 6, 11-dihydrodibenzo-[be]-thiepin-11-one-3-acetic acid (Tiopinac)
EP0061406A1 (en) Benzamido-alkyl-hydroxamic-acid derivatives, preparation thereof and therapeutical composition
US4435591A (en) Compound with analgesic, antiinflammatory and antipyretic activity, and pharmaceutical compositions therefrom
SU1499901A1 (en) N-quinoline-6-ylamide of pentaacetylglycyrrisinic acid displaying antiinelammatory activity

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 99805000.8

Country of ref document: CN

AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

WWE Wipo information: entry into national phase

Ref document number: 18731/00

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: PA/a/2000/008048

Country of ref document: MX

Ref document number: PV2000-3018

Country of ref document: CZ

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2000/03019

Country of ref document: TR

WWP Wipo information: published in national office

Ref document number: PV2000-3018

Country of ref document: CZ

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
WWG Wipo information: grant in national office

Ref document number: 18731/00

Country of ref document: AU

WWG Wipo information: grant in national office

Ref document number: PV2000-3018

Country of ref document: CZ