WO1993001196A1 - Ketoalkylglycerophospholipids having antitumor and anti-platelet aggregation activities, processes for the preparation thereof and pharmaceutical compositions therefrom - Google Patents

Ketoalkylglycerophospholipids having antitumor and anti-platelet aggregation activities, processes for the preparation thereof and pharmaceutical compositions therefrom Download PDF

Info

Publication number
WO1993001196A1
WO1993001196A1 PCT/EP1992/001502 EP9201502W WO9301196A1 WO 1993001196 A1 WO1993001196 A1 WO 1993001196A1 EP 9201502 W EP9201502 W EP 9201502W WO 9301196 A1 WO9301196 A1 WO 9301196A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
oxooctadecyl
methylglycero
group
Prior art date
Application number
PCT/EP1992/001502
Other languages
French (fr)
Inventor
Germano Carganico
David Mauleon Casellas
Maria De Los Desemparados Fos Torro
Original Assignee
Laboratorios Menarini 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 Laboratorios Menarini S.A. filed Critical Laboratorios Menarini S.A.
Publication of WO1993001196A1 publication Critical patent/WO1993001196A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/06Phosphorus compounds without P—C bonds
    • C07F9/08Esters of oxyacids of phosphorus
    • C07F9/09Esters of phosphoric acids
    • C07F9/10Phosphatides, e.g. lecithin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/553Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having one nitrogen atom as the only ring hetero atom
    • C07F9/576Six-membered rings
    • C07F9/58Pyridine rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/645Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having two nitrogen atoms as the only ring hetero atoms
    • C07F9/6503Five-membered rings
    • C07F9/6506Five-membered rings having the nitrogen atoms in positions 1 and 3
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6536Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom having nitrogen and sulfur atoms with or without oxygen atoms, as the only ring hetero atoms
    • C07F9/6539Five-membered rings

Definitions

  • the present invention relates to novel glycerophospholipids having antitumor and anti-platelet aggregating activities, a process for the preparation thereof and pharmaceutical compositions containing them.
  • PAF Platelet activating factor
  • 1-O-alkyl-2-acetyl-sn-glycero-3-phosphocholine of formula A
  • PAF-antagonists can be interesting in therapy for the treatment of those pathological processes in which PAF is involved.
  • ALPs alkyl-lyso-phospholipids
  • PAF phosphatidylcholine
  • An example of phospholipids of this kind is ET-18-OCH 3 , of formula B
  • Said compounds show an antitumor activity in various pharmacological models [W.E. Berdel and P.G. Munder in "PAF and related lipid mediators", Ed. F. Snyder, Plenum Press, New York, 1987, p. 449] and no interactions with cell DNA.
  • ET-18-OCH 3 and other analogues show some of PAF actions, such as serotonine release, bronchoconstriction, platelet aggregation and hypotension. Said actions are contra-indicated in case of antitumor treatment and they can cause remarkable circulatory disorders.
  • a compound having the cytostatic action of ET-18-OCH 3 (formula B) and lacking the PAF-like action and capable of blocking the PAF action on the specific cell receptors thereof, could advantageously be used in human therapy as an antitumor agent, without suffering from the drawbacks of compound B.
  • the compounds of the present invention (formula I), containing a keto group in the chain 1, show an antitumor action which can be compared to that of ET- 18-OCH 3 , without having PAF-agonist action. Moreover, said compounds cause no hemolysis and have a PAF-antagonist activity.
  • the present invention relates to compounds of general formula (I)
  • R 1 is a C 1 -C 16 straight or branched alkyl group, a phenyl group or a phenyl-C 1 -C 16 alkyl group ; n is an integer from 3 to 18, inclusive;
  • R 2 is a C 1 -C 12 straight or branched alkyl group
  • R 3 is hydrogen, a carboxy, alkoxycarbonyl, arylalkoxycarbonyl or aryloxycarbonyl group of less than 12 carbon atoms in the last three cases;
  • R 4 , R 5 and R 6 which can be the same or different, are hydrogen, a C 1 -C 6 alkyl group or N +
  • R 4 R 5 R 6 is a cyclic ammonium group, which can be aromatic or non-aromatic, wherein two of the R 4 , R 5 or R 6 groups form a ring together with the nitrogen atom, and the other group is hydrogen or C 1 -C 6 alkyl.
  • X- is a pharmaceutically acceptable anion, such as chloride, bromide or iodide anions, and the other symbols have the above mentioned meanings,
  • M + is an alkali metal cation, (for example Na + ,
  • the present invention also relates to all the possible stereoisomers of compounds (I) and the mixtures thereof.
  • R 2 when R 2 is an alkyl group, this can be methyl, ethyl, propyl, butyl, pentyl or hexyl; when R 3 is an alkoxycarbonyl or arylalkoxycarbonyl group, this can be methoxycarbonyl, ethoxycarbonyl, butoxycarbonyl, pentoxycarbonyl or benzyloxycarbonyl; when R 3 is an aryloxycarbonyl group, this can be phenoxycarbonyl; when R 4 , R 5 and R 6 are an alkyl group, this can be methyl, ethyl or propyl; when N + R 4 R-R 6 is a cyclic ammonium group, this can be an aromatic group, such as 1-pyridinium, 1-quinolinium, 1-imidazolium, 1-pyrazolium, 3-thiazolium, 3-oxazolium, 1-benzimidazolium, 3-benzothiazolium or 3-benzoxa
  • Preferred compounds of the invention are those in which R 1 and R 3 have the above mentioned meanings, R 2 is a C 1 -C 3 alkyl group, n is an integer form 3 to 18, R 4 , R 5 and R 6 are hydrogen, methyl or ethyl, or N + R 4 R 5 R 6 is preferably 1-pyridinium, 1-imidazolium, 3- thiazolium, 1-pyrrolidinium, 1-piperidinium or 4- morpholinium.
  • Particularly preferred compounds of the invention are the following ones:
  • the present invention also relates to pharmaceutical compositions containing a compound of formula (I), or a pharmaceutically acceptable salt thereof, in admixture with a suitable carrier or excipient, as well as the use of compounds (I) for the preparation of a medicament.
  • compounds (I) can be prepared starting from a racemic or enantiomerically pure compound of formula (XI)
  • n, R 1 and R 2 have the above mentioned meanings, and Z is oxygen or a keto-protecting group (such as ethylenedioxy or dimethyl acetal).
  • X and Y which can be the same or different, are halogens (such as Cl, Br, I), in the presence of an amine such as triethylamine or pyridine, in an inert organic solvent, such as ethyl ether, tetrahydrofuran (THF) or chloroform, at a temperature from 0° to 40 oC, to give a compound of formula (XIII)
  • R 4 , R 5 and R 6 have the above mentioned meanings, in a suitable organic solvent, such as chloroform, acetonitrile, dimethylformamide (DMF) or toluene, at a temperature from 40°C to the solvent's reflux temperature, to give a compound of formula (I), wherein n, R 1 , R 2 , R 4 , R 5 and R 6 have the above mentioned meanings and R 3 is hydrogen.
  • a suitable organic solvent such as chloroform, acetonitrile, dimethylformamide (DMF) or toluene
  • X is halogen (such as Cl, Br, I) in a suitable organic solvent, such as ethyl ether or benzene, in the presence of an amine such as triethylamine or pyridine, at a temperature from 0° to 40 °C, to give a compound of formula (XVII)
  • compound (XVII) is treated with a compound of formula (XV), in suitable organic solvents, such as dichloromethane, acetonitrile or benzene, at a temperature from 40oC to the solvent's reflux temperature, to give a compound of formula (I) wherein n , R 1 , R 2 , R 4 R 5 and R 6 have the above mentioned meanings and R 3 is hydrogen .
  • suitable organic solvents such as dichloromethane, acetonitrile or benzene
  • R 1 and R 2 have the above mentioned meanings, is reacted with PCI 3 in the presence of a slightly nucleophilic base, such as imidazol, and an acid-binding agent, such as triethylamine or pyridine, in suitable organic solvents, such as acetonitrile or toluene, at a temperature from 0o to 40°C, to give a compound of formula (XVIII)
  • R 7 is an amino-protecting group, such as benzyloxycarbonyl or t-butoxycarbonyl
  • R 3 can be an alkoxycarbonyl group having less than 12 carbon atoms, or it can be a carboxy group protected in form of the benzyl or t-butyl esters, in the presence of a base such as pyridine and of a condensing agent, such aspivaloyl chloride or 5,5-dimethyl-2-oxo-2-chloro-1,3,2-dioxophospholane, at a temperature from 0° to 40 °C, to give a compound of formula (XX)
  • n, Z, R 1 , R 2 , R 3 and R 7 have the above mentioned meanings, which is oxidized with an oxidizing agent, such as iodine, to give a compound of formula (XXI)
  • n , Z , R 1 , R 2 R 3 and R 7 have the above mentioned meanings.
  • This compound is transformed into a compound of formula (I) by removing the protecting groups R 7 and Y, if necessary, when R 3 is a carboxy group protected in form of a benzyl or t-butyl ester.
  • both the protecting groups, as well as Z can be removed in an acid medium (such as HCl).
  • an acid medium such as HCl
  • R 7 is a benzyloxycarbonyl group and R 3 is a benzyl ester
  • they can be deprotected simultaneously with the elimination of Z, by means of catalytic hydrogenation with 10% Pd/C or Pd(OH) 2 /c in solvents such as methanol, water or mixtures thereof, in the presence of an acid, such as HCl, under pressures ranging from the atmospheric one to 50 psi.
  • a compound of formula (XI), wherein n, R 1 and R 2 have the above mentioned meanings and Z is oxygen, can be obtained according to step f) by reacting a compound of formula (VII) with a Grignard reactive R 1 MgX, such as alkylmagnesium bromide or phenylalkylmagnesium bromide, in a suitable solvent (such as ethyl ether, THF, benzene or mixtures thereof) at a temperature from 25oC to the solvent's reflux one, followed by hydrolysis and detritylation in an acid medium (by HCl or H 2 SO 4 ), in the presence of a suitable solvent, such as dioxane, at a temperature from the room one to the solvent's reflux temperature.
  • a suitable solvent such as ethyl ether, THF, benzene or mixtures thereof
  • a compound of formula (VII) can be prepared, for example, following the synthetic sequence of scheme 1, by reacting the compound of formula (II), which is commercially available, with a compound Q(CH 2 ) n Br (XXII), wherein Q is halogen (for example Cl, Br) and n has the above mentioned meanings, in a suitable organic solvent, such as DMF or THF, in the presence of an alkali hydride, such as NaH, at a temperature from 0° to 50°C, to obtain a compound of formula (III).
  • a suitable organic solvent such as DMF or THF
  • an alkali hydride such as NaH
  • Compound (III) is subjected to a substitution reaction with NaCN or KCN in a suitable organic solvent, such as DMSO or an ethanol-water mixture, at a temperature from the room's one to 100°C, to obtain compound (IV).
  • a compound of formula (V) can be obtained starting from a compound of formula (IV), by means of acid hydrolysis with HCl or H 2 SO 4 , for example, at a temperature from room temperature to the solvent's reflux temperature, in a suitable solvent such as THF.
  • a compound of formula (VI) can be prepared by reacting a compound of formula (V) with trityl chloride, or in the presence of bases, such as pyridine or triethylamine, with or without DMAP, at at temperature from the room's one to 100 °C.
  • Step e) can be carried out by reacting a compound of formula (VI) with a compound R 2 X, wherein X is halogen (such as Br, Cl, I) and R 2 can have the above mentioned meanings, in a suitable solvent, such as DMF, THF or benzene, in the presence of sodium hydride or potassium hydride, at a temperature from the room's one to the reflux temperature of the solvent.
  • a suitable solvent such as DMF, THF or benzene
  • a compound of formula (XI), wherein n, Z, R 1 and R 2 have the above mentioned meanings, can be prepared, according to steps g) and h), by reacting an alcohol of formula (VIII), whose preparation is well disclosed in literature [M.Takatani et al., J. Med.
  • a compound of formula (IX) can be prepared, for example, according to the reaction scheme 3: Scheme 3
  • Compounds of formula (XXIV) can be prepared by reacting compounds of formula (XXIII) with sodium or potassium cyanides, in DMSO or in an ethanol-water mixture, at a temperature ranging from 50 °C to the solvent's reflux temperature.
  • XXIV with a Grignard reactive R 1 MgX, for example alkylmagnesium bromide or phenylalkylmagnesium bromide, in a suitable solvent, such as ethyl ether, THF, benzene or mixtures thereof, at a temperature ranging from 20 °C to the solvent's reflux temperature, followed by acid hydrolysis, for example with HCl or H 2 SO 4 , in the presence of a suitable solvent, such as ethyl ether, THF or dioxane and at a temperature ranging from room temperature to the solvent's reflux temperature.
  • a suitable solvent such as ethyl ether, THF, benzene or mixtures thereof
  • keto function of compounds of formula (XXV) can be protected by reaction with a suitable alcohol, such as ethylene glycol or methanol in an acid medium, such as p-toluenesulfonic acid, in a suitable solvent, such as benzene or toluene, at a temperature ranging from 25 °C to the solvent's reflux temperature.
  • a suitable alcohol such as ethylene glycol or methanol
  • an acid medium such as p-toluenesulfonic acid
  • a suitable solvent such as benzene or toluene
  • Compounds of formula (XXVII) can be prepared starting from compounds of formula (XXVI), or, when z is oxygen, starting from compounds of formula (XXV), by catalytic hydrogenation under atmospheric pressure, for example with 10% Pd/C or Pd(OH) 2 /C, in solvents such as methanol, ethanol, water or mixtures thereof.
  • the compounds of the present invention can be used in human therapy as antitumor, antiasthmatic, antithrombotic, anti-inflammatory, anti-allergic agents as well as in the prophylaxis of anaphylactic shock. Moreover, they can be used as hypotensive or antianginal agents.
  • the compounds of the invention can be formulated according to conventional techniques and excipients, such as those described in "Remington's Pharmaceutical Science Handbook", Mack Pub. Co., New York, U.S.A. Examples of said forms include capsules, tablets, syrups and the like, containing 1 to 1000 mg of the active ingredient per unitary dose.
  • a suspension of 0.188 g (6.2 mmoles) of 80% NaH in anhydrous DMF is added with a solution of 2.73 g (11.2 mmoles) of dibromohexane in DMF.
  • a solution of 0.74 g (5.6 mmoles) of isopropylidene glycerol in DMF is dropped therein.
  • the reaction mixture is left under stirring at room temperature for 18 hours, after that ethanol and water are added.
  • the solution is extracted with ether, washed with a sodium chloride saturated solution, dried and solvent is evaporated off.
  • the obtained crude product is purified by distillation under high vacuum, the excess 1, 6-dibromohexane distils at 42-44 °C/0.0 torr and 1-O-(6-bromohexyl)-2,3-O-isopropylideneglycerol distils at 70-72 °C/0.3 torr.
  • the distillation residue is purified by flash chromatography on silica gel, eluting with dichloromethane. 0.680 g of 1-O-(6-bromohexyl)-2,3-O-isopropylideneglycerol are obtained (41% yield).
  • IR (NaCl) cm -1 2960, 2900, 2840, 1430, 1360, 1240,
  • a solution of 1.19 g (24 mmoles) of sodium cyanide in 25 ml of DMSO is heated to 90°C and added with a solution of 6 g (20 mmoles) of 1-O-(6-bromohexyl)-2,3- O-isopropylideneglycerol in DMSO.
  • the mixture is left at 90 °C for 1 hour, then water and ethyl ether are added, the two phases are separated and the organic phase is washed with a sodium chloride saturated solution, dried, and solvent is evaporated off to obtain 4.55 g of 1-O-(6-cyanohexyl)-2,3-O-isopropylideneglycerol (95% yield).
  • IR (NaCl) cm -1 3500, 3100, 3060, 3040, 2950, 2850, 2250, 1590, 1385
  • the mixture is refluxed for 6 hours and left for 3 days at room temperature, after that solvent is evaporated off and 100 ml of dioxane and 30 ml of HCl IN are added thereto.
  • the reaction mixture is refluxed for two hours, neutralized with 5% NaHCO 3 and extracted with ethyl ether. After drying and evaporating the solvent, a crude product is obtained which is purified by flash chromatography on silica gel. By eluting with mixtures of petroleum ether:ethyl ether of increasing polarity, 0.575 g of 1-O-(7-oxooctadecyl)-2-O-methylglycerol are obtained (35% yield).
  • reaction mixture is extracted with ethyl ether, dried and solvent is evaporated off to obtain 0.150 g of 1-O-(7-oxooctadecyl)-2-O-methylglycero-3-(2-bromoethyl)phosphate (quantitative yield).
  • a solution of 0.576 g (11.7 mmoles) of sodium cyanide in DMSO is heated to 90°C and added with a solution of 3 g (11.7 mmoles) of 1-benzyloxy-6-bromohexane in DMSO.
  • the reaction mixture is left at 90°C for 16 hours, then water and ethyl ether are added, the two phases are separated and the organic phase is washed with a NaCl saturated solution, dried and solvent is evaporated off to obtain 2.053 g of 7-benzyloxyheptanonitrile (87% yield).
  • a solution of 0.580 g (2.9 mmoles) of 3-O-benzyl-2-O-methylglycerol in toluene is added to a suspension of 0.684 g (12.2 mmoles) of powder KOH in 24 ml of anhydrous toluene.
  • the reaction mixture is refluxed for 1 hour in a Dean-Stark apparatus.
  • 1 g (2.4 mmoles) of 4-ethylenedioxyoctadecyl methanesulfonate dissolved in 16 ml of toluene is added. Reflux in the Dean-Stark apparatus is maintained for 5 hours, then toluene is evaporated off, some water is added and the mixture is extracted with ethyl ether.
  • the ether phase is washed with water to neutral pH.
  • the organic phase is dried and evaporated, to obtain a crude product which is purified by silica gel chromatography. Eluting with mixtures of petroleum ether: ethyl ether of increasing polarity, 0.569 g of 1-O-(4- ethylenedioxyoctadecyl)-2-O-methyl-3-O-benzylglycerol are obtained (46% yield).
  • 0.061 ml (0.5 mmole) of pivaloyl chloride is added to a mixture of 0.120 g (0.25 mmole) of 1-O-(7-ethylenedioxyoctadecyl)-2-O-methylglycerol phosphonate, 0.132 g (0.4 mmole) of N-benzyloxycarbonylserine benzyl ester and 3.5 ml of pyridine.
  • the reaction mixture is stirred for 1.5 hours at room temperature, 0.2 ml of water and 0.127 g (0.5 mmole) of iodine are added.
  • HL-60 line cells obtained starting from human promyelocyte leukemia, are incubated in 6- well culture plates for 24 hours at 37°C, at a concentration of 1 ⁇ 10 6 cells/ml, in the presence of the test products, which have previously been dissolved in the culture medium. Cytotoxicity is measured by means of the Trypan Blue exclusion technique (Practical Immunology, pp 29-32, Oxford, England : Blackwell Scientific Publications, 1976). Cytotoxic activities of the products, expressed as IC 50 , are reported in Tables IA and IB.
  • Platelets are prepared starting from blood samples obtained by means of a carotid cannula from male albino New Zealand rabbits (2-2.5 kg), subjected to anticoagulant treatment with sodium citrate dihydrate (3.2%) 1:10. Blood is centrifuged (150xg, 10 min, 22°C) to separate the platelet-rich plasma fraction (PRP) from erythrocytes. Platelets are separated from plasmatic components by serial washings and centrifugations (Blood, 1986, 62:337), thereafter they are suspended in a physiological buffer at a concentration of 5 ⁇ 10 5 /ml.
  • a 400 ⁇ l volume of the above mentioned suspension is placed into the cuvette of a multicanal aggregometer (Aggrecoder HU) where it is kept at constant temperature and stirring.
  • platelets are added with 50 ⁇ l of physiological buffer containing 100 ⁇ M acetylsalicylic acid and apirase 5 mg/ml, in order to block any non PAF-related platelet activation metabolic pathways.
  • the platelet suspensions are added with 50 ⁇ l of the different solutions to be tested. Changes in light transmission, due to platelet aggregation in the suspension, are recorded and expressed as percent platelet aggregation, according to the procedure by Born and Cross (J. Physiol., 1962, 162:67). Standard curve is performed with PAF within a concentration range of 0.1-1 nM.
  • Capability to inhibit PAF-induced platelet aggregation is evaluated by means of the aggregometry technique in suspensions of rabbit washed platelets, obtained by means of the same procedure as described in Example 6.
  • the inhibition potency of the products under test expressed as the corresponding IC 50 , is evaluated on the maximum aggregation values obtained in PAF standard curve.
  • the results obtained from the tests, in which PAF-antagonist activity was measured, are reported in Tables IIA, IIB.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Compounds of general formula (I) wherein R1 is a C¿1?-C16 straight or branched alkyl group, a phenyl group or a phenyl-C6-C16alkyl group; n is an integer from 3 to 18, inclusive; R?2¿ is a C¿1?-C12 straight or branched alkyl group; R?3¿ is hydrogen, a carboxy, alkoxycarbonyl, arylalkoxycarbonyl or aryloxycarbonyl group of less than 12 carbon atoms in the last three cases; R?4, R5 and R6¿, which can be the same or different, are hydrogen, a C¿1?-C6 alkyl group or N?+R4R5R6¿ is a cyclic ammonium group, which can be aromatic or non-aromatic, wherein two of the R?4, R5 or R6¿ groups form a ring together with the nitrogen atom, and the other group is hydrogen or C¿1?-C6 alkyl, have antitumor and anti-platelet aggregating activities.

Description

KETOALKYLGLYCEROPHOSPHOLIPIDS HAVING ANTITUMOR AND ANTI-PLATELET AGGREGATION ACTIVITIES, PROCESSES FOR THE PREPARATION THEREOF AND PHARMACEUTICAL COMPOSITIONS THEREFROM
The present invention relates to novel glycerophospholipids having antitumor and anti-platelet aggregating activities, a process for the preparation thereof and pharmaceutical compositions containing them.
TECHNOLOGICAL BACKGROUND
Platelet activating factor (PAF), or 1-O-alkyl-2-acetyl-sn-glycero-3-phosphocholine, of formula A
Figure imgf000003_0001
is a potent cell mediator of phospholipid nature, which is ascribed to play a paramount role in various pathological processes, such as asthma, anaphylaxis, inflammation and ischemia [P. Braguet et al., Pharmacol. Rev., 39(2), 97 (1987)]. As a consequence, a great number of searches have been carried out in order to find PAF-antagonist analogues, which can block PAF action on PAF receptors at the level of the cell membrane [K. Cooper and M.J. Parry, Ann. Rep. Med. Chem., 24, 81 (1989)].
Therefore, specific PAF-antagonists can be interesting in therapy for the treatment of those pathological processes in which PAF is involved.
Another class of synthetic phospholipids is represented by those named alkyl-lyso-phospholipids (ALPs), which are structurally related to lyso- phosphatidylcholine and PAF, from which compound they differ mainly in that they contain a second ether function, which is difficult to metabolize, at the 2- position of glycerol. An example of phospholipids of this kind is ET-18-OCH3, of formula B
Figure imgf000004_0001
Said compounds show an antitumor activity in various pharmacological models [W.E. Berdel and P.G. Munder in "PAF and related lipid mediators", Ed. F. Snyder, Plenum Press, New York, 1987, p. 449] and no interactions with cell DNA. Undoubtedly, ET-18-OCH3 and other analogues show some of PAF actions, such as serotonine release, bronchoconstriction, platelet aggregation and hypotension. Said actions are contra-indicated in case of antitumor treatment and they can cause remarkable circulatory disorders.
As a consequence, a compound having the cytostatic action of ET-18-OCH3 (formula B) and lacking the PAF-like action and capable of blocking the PAF action on the specific cell receptors thereof, could advantageously be used in human therapy as an antitumor agent, without suffering from the drawbacks of compound B.
The compounds of the present invention (formula I), containing a keto group in the chain 1, show an antitumor action which can be compared to that of ET- 18-OCH3, without having PAF-agonist action. Moreover, said compounds cause no hemolysis and have a PAF-antagonist activity.
DISCLOSURE OF THE INVENTION
The present invention relates to compounds of general formula (I)
Figure imgf000005_0001
wherein R1 is a C1-C16 straight or branched alkyl group, a phenyl group or a phenyl-C1-C16alkyl group ; n is an integer from 3 to 18, inclusive;
R2 is a C1-C12 straight or branched alkyl group;
R3 is hydrogen, a carboxy, alkoxycarbonyl, arylalkoxycarbonyl or aryloxycarbonyl group of less than 12 carbon atoms in the last three cases;
R4, R5 and R6, which can be the same or different, are hydrogen, a C1-C6 alkyl group or N+R4R5R6 is a cyclic ammonium group, which can be aromatic or non-aromatic, wherein two of the R4, R5 or R6 groups form a ring together with the nitrogen atom, and the other group is hydrogen or C1-C6 alkyl.
The compounds of general formula (I) can be obtained in the form of salts, which can be represented either by formula (la)
Figure imgf000006_0001
wherein X-is a pharmaceutically acceptable anion, such as chloride, bromide or iodide anions, and the other symbols have the above mentioned meanings,
or by formula (Ib)
Figure imgf000006_0002
wherein M+ is an alkali metal cation, (for example Na+,
K+), or the equivalent of an alkaline-earth metal (for example 1/2 Ca++, 1/2 Mg++) and the other symbols have the above mentioned meanings. When R3 is a carboxy group, the compounds of general formula (I) can also be obtained in form of salts, which can be represented by formula (Ic)
Figure imgf000007_0001
wherein all the symbols have the above mentioned meanings.
Since the compounds of the present invention have one or more asymmetric carbon atoms, the present invention also relates to all the possible stereoisomers of compounds (I) and the mixtures thereof.
In compounds of general formula I, when R2 is an alkyl group, this can be methyl, ethyl, propyl, butyl, pentyl or hexyl; when R3 is an alkoxycarbonyl or arylalkoxycarbonyl group, this can be methoxycarbonyl, ethoxycarbonyl, butoxycarbonyl, pentoxycarbonyl or benzyloxycarbonyl; when R3 is an aryloxycarbonyl group, this can be phenoxycarbonyl; when R4 , R5 and R6 are an alkyl group, this can be methyl, ethyl or propyl; when N+R4R-R6 is a cyclic ammonium group, this can be an aromatic group, such as 1-pyridinium, 1-quinolinium, 1-imidazolium, 1-pyrazolium, 3-thiazolium, 3-oxazolium, 1-benzimidazolium, 3-benzothiazolium or 3-benzoxazolium, or it can be non aromatic, in which case two of R4, R5 and R6 form, together with the nitrogen atom they are linked to, a 1-pyrrolidino, 1-piperidino, 4-morpholino or 1-piperazino ring, and the remaining group is hydrogen or a C1-C6 alkyl group.
Preferred compounds of the invention are those in which R1 and R3 have the above mentioned meanings, R2 is a C1-C3 alkyl group, n is an integer form 3 to 18, R4, R5 and R6 are hydrogen, methyl or ethyl, or N+R4R5R6 is preferably 1-pyridinium, 1-imidazolium, 3- thiazolium, 1-pyrrolidinium, 1-piperidinium or 4- morpholinium.
Particularly preferred compounds of the invention are the following ones:
1-O-(4-oxooctadecyl)-2-O-methylglycero-3-phosphocholine 1-O-(4-oxooctadecyl)-2-O-methylglycero-3-phosphoserine
1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phosphocholine 1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phosphoserine 1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phosphoserine methyl ester
1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phospho(N,N,N-trimethyl)serine
2-O-methyl-3-O-(7-oxooctadecyl)-sn-glycero-1-phosphocholine
2-O-methyl-1-O-(7-oxooctadecyl)-sn-glycero-3-phospho- choline
1-O-(7-oxooctadecyl)-2-O-ethylglycero-3-phosphocholine 1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phospho[2-(3-thiazolium)ethyl]
1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phospho[2-(1-imidazolium)ethyl]
1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phospho[2-(N-methylpiperidinium)ethyl] 1-O-(12-oxooctadecyl)-2-O-methylglycero-3-phosphocholine
1-O-(12-oxooctadecyl)-2-O-methylglycero-3-phosphoserine
1-O-(11-phenyl-11-oxoundecyl)-2-O-methylglycero-3-phosphocholine
1-O-(14-phenyl-11-oxotetradecyl)-2-O-methylglycero-3-phosphocholine
1-O-(14-phenyl-11-oxotetradecyl)-2-O-methylglycero-3-phosphoserine.
The present invention also relates to pharmaceutical compositions containing a compound of formula (I), or a pharmaceutically acceptable salt thereof, in admixture with a suitable carrier or excipient, as well as the use of compounds (I) for the preparation of a medicament.
According to the present invention, compounds (I) can be prepared starting from a racemic or enantiomerically pure compound of formula (XI)
Figure imgf000009_0001
wherein n, R1 and R2 have the above mentioned meanings, and Z is oxygen or a keto-protecting group (such as ethylenedioxy or dimethyl acetal).
According to the process of the invention:
a) A compound of formula (XI) wherein Z is oxygen and n, R1 and R2 have the above mentioned meanings, is reacted with a compound of formula (XII)
Figure imgf000010_0001
wherein X and Y, which can be the same or different, are halogens (such as Cl, Br, I), in the presence of an amine such as triethylamine or pyridine, in an inert organic solvent, such as ethyl ether, tetrahydrofuran (THF) or chloroform, at a temperature from 0° to 40 ºC, to give a compound of formula (XIII)
Figure imgf000010_0002
wherein n, R 1, R3, X and Y have the above mentioned meanings. After that, aqueous hydrolysis in a saline solution, such as a sodium chloride solution at a temperature from 0° to 50 ºC, gives the compound of formula (XIV) ^
Figure imgf000010_0003
wherein R1, R2 and Y have the above mentioned meanings. Compound (XIV) is reacted with a compound of formula (XV)
Figure imgf000011_0001
wherein R4, R5 and R6 have the above mentioned meanings, in a suitable organic solvent, such as chloroform, acetonitrile, dimethylformamide (DMF) or toluene, at a temperature from 40°C to the solvent's reflux temperature, to give a compound of formula (I), wherein n, R1, R2, R4, R5 and R6 have the above mentioned meanings and R3 is hydrogen.
b) Alternatively, compound (XI), wherein Z is oxygen and n, R1 and R2 have the above mentioned meanings, is reacted with a compound of formula (XVI)
Figure imgf000011_0002
wherein X is halogen (such as Cl, Br, I) in a suitable organic solvent, such as ethyl ether or benzene, in the presence of an amine such as triethylamine or pyridine, at a temperature from 0° to 40 °C, to give a compound of formula (XVII)
Figure imgf000012_0001
wherein R1 and R2 have the above mentioned meanings. Subsequently, compound (XVII) is treated with a compound of formula (XV), in suitable organic solvents, such as dichloromethane, acetonitrile or benzene, at a temperature from 40ºC to the solvent's reflux temperature, to give a compound of formula (I) wherein n , R1 , R2 , R4 R5 and R6 have the above mentioned meanings and R3 is hydrogen .
c ) Alternatively , a compound of formula ( XI ) wherein Z is oxygen or a suitable keto-protecting group , and n ,
R1 and R2 have the above mentioned meanings, is reacted with PCI3 in the presence of a slightly nucleophilic base, such as imidazol, and an acid-binding agent, such as triethylamine or pyridine, in suitable organic solvents, such as acetonitrile or toluene, at a temperature from 0º to 40°C, to give a compound of formula (XVIII)
Figure imgf000012_0002
wherein R1 and R2 have the above mentioned meanings and A can be hydrogen or a trialkylammonium group. Compound (XVIII) is reacted with a compound of formula (XIX)
Figure imgf000013_0002
wherein R7 is an amino-protecting group, such as benzyloxycarbonyl or t-butoxycarbonyl, and R3 can be an alkoxycarbonyl group having less than 12 carbon atoms, or it can be a carboxy group protected in form of the benzyl or t-butyl esters, in the presence of a base such as pyridine and of a condensing agent, such aspivaloyl chloride or 5,5-dimethyl-2-oxo-2-chloro-1,3,2-dioxophospholane, at a temperature from 0° to 40 °C, to give a compound of formula (XX)
Figure imgf000013_0001
wherein n, Z, R1, R2 , R3 and R7 have the above mentioned meanings, which is oxidized with an oxidizing agent, such as iodine, to give a compound of formula (XXI)
Figure imgf000014_0001
wherein n , Z , R1 , R2 R3 and R7 have the above mentioned meanings. This compound is transformed into a compound of formula (I) by removing the protecting groups R7 and Y, if necessary, when R3 is a carboxy group protected in form of a benzyl or t-butyl ester.
When R7 is a t-butoxycarbonyl group and R3 is a t-butyl ester, both the protecting groups, as well as Z, can be removed in an acid medium (such as HCl). When R7 is a benzyloxycarbonyl group and R3 is a benzyl ester, they can be deprotected simultaneously with the elimination of Z, by means of catalytic hydrogenation with 10% Pd/C or Pd(OH)2/c in solvents such as methanol, water or mixtures thereof, in the presence of an acid, such as HCl, under pressures ranging from the atmospheric one to 50 psi.
The preparations of compounds (XI) starting from known or commercially available precursors are shown in the following schemes 1 and 2 Scheme 1
Figure imgf000015_0001
Scheme 2
Figure imgf000016_0001
A compound of formula (XI), wherein n, R1 and R2 have the above mentioned meanings and Z is oxygen, can be obtained according to step f) by reacting a compound of formula (VII) with a Grignard reactive R1MgX, such as alkylmagnesium bromide or phenylalkylmagnesium bromide, in a suitable solvent (such as ethyl ether, THF, benzene or mixtures thereof) at a temperature from 25ºC to the solvent's reflux one, followed by hydrolysis and detritylation in an acid medium (by HCl or H2SO4), in the presence of a suitable solvent, such as dioxane, at a temperature from the room one to the solvent's reflux temperature.
A compound of formula (VII) can be prepared, for example, following the synthetic sequence of scheme 1, by reacting the compound of formula (II), which is commercially available, with a compound Q(CH2)nBr (XXII), wherein Q is halogen (for example Cl, Br) and n has the above mentioned meanings, in a suitable organic solvent, such as DMF or THF, in the presence of an alkali hydride, such as NaH, at a temperature from 0° to 50°C, to obtain a compound of formula (III). Compound (III) is subjected to a substitution reaction with NaCN or KCN in a suitable organic solvent, such as DMSO or an ethanol-water mixture, at a temperature from the room's one to 100°C, to obtain compound (IV).
A compound of formula (V) can be obtained starting from a compound of formula (IV), by means of acid hydrolysis with HCl or H2SO4, for example, at a temperature from room temperature to the solvent's reflux temperature, in a suitable solvent such as THF.
A compound of formula (VI) can be prepared by reacting a compound of formula (V) with trityl chloride, or in the presence of bases, such as pyridine or triethylamine, with or without DMAP, at at temperature from the room's one to 100 °C.
Step e) can be carried out by reacting a compound of formula (VI) with a compound R2X, wherein X is halogen (such as Br, Cl, I) and R2 can have the above mentioned meanings, in a suitable solvent, such as DMF, THF or benzene, in the presence of sodium hydride or potassium hydride, at a temperature from the room's one to the reflux temperature of the solvent.
Alternatively, a compound of formula (XI), wherein n, Z, R1 and R2 have the above mentioned meanings, can be prepared, according to steps g) and h), by reacting an alcohol of formula (VIII), whose preparation is well disclosed in literature [M.Takatani et al., J. Med. Chem., 32, 56 (1989)], with a compound of formula (IX), in a suitable organic solvent, such as toluene, benzene, dimethylsulfoxide (DMSO) or DMF, in the presence of sodium hydride, potassium hydride or KOH, at a temperature from the room's one to the solvent's reflux temperature, then removing the benzyl group by catalytic hydrogenation under atmospheric pressure, for example with 10% Pd/C or Pd(OH)2/c is solvents such as methanol, ethanol, water or mixtures thereof and the protecting group Z can be removed, if desired, carrying out the same catalytic hydrogenation in an acid medium, such as HCl.
A compound of formula (IX) can be prepared, for example, according to the reaction scheme 3: Scheme 3
Figure imgf000019_0001
Compounds of formula (XXII), wherein Q is a hydroxy group or a bromine atom and n has the above mentioned meanings, are commercially available.
Compounds of formula (XXIII) can be obtained, according to step i), starting from compounds of formula (XXII), by reaction with benzyl alcohol (when Q = Br) or benzyl bromide (when Q = OH) in the presence of an hydride, such as sodium hydride, in a suitable organic solvent, such as THF, at a temperature ranging from room temperature to the solvent's reflux temperature.
Compounds of formula (XXIV) can be prepared by reacting compounds of formula (XXIII) with sodium or potassium cyanides, in DMSO or in an ethanol-water mixture, at a temperature ranging from 50 °C to the solvent's reflux temperature.
Compounds of formula (XXV) can be prepared, according to step k), by reacting compounds of formula
(XXIV) with a Grignard reactive R1MgX, for example alkylmagnesium bromide or phenylalkylmagnesium bromide, in a suitable solvent, such as ethyl ether, THF, benzene or mixtures thereof, at a temperature ranging from 20 °C to the solvent's reflux temperature, followed by acid hydrolysis, for example with HCl or H2SO4, in the presence of a suitable solvent, such as ethyl ether, THF or dioxane and at a temperature ranging from room temperature to the solvent's reflux temperature.
If desired, the keto function of compounds of formula (XXV) can be protected by reaction with a suitable alcohol, such as ethylene glycol or methanol in an acid medium, such as p-toluenesulfonic acid, in a suitable solvent, such as benzene or toluene, at a temperature ranging from 25 °C to the solvent's reflux temperature.
Compounds of formula (XXVII) can be prepared starting from compounds of formula (XXVI), or, when z is oxygen, starting from compounds of formula (XXV), by catalytic hydrogenation under atmospheric pressure, for example with 10% Pd/C or Pd(OH)2/C, in solvents such as methanol, ethanol, water or mixtures thereof.
Compounds of formula (IX), wherein n, Z and R1 have the above mentioned meanings, can be prepared by reacting compounds of formula (XXVII), for example, with methanesulfonyl chloride, in a suitable solvent, such as dichloromethane or chloroform, at a temperature ranging from -5ºC to room temperature .
The compounds of the present invention can be used in human therapy as antitumor, antiasthmatic, antithrombotic, anti-inflammatory, anti-allergic agents as well as in the prophylaxis of anaphylactic shock. Moreover, they can be used as hypotensive or antianginal agents.
For this purpose, the compounds of the invention can be formulated according to conventional techniques and excipients, such as those described in "Remington's Pharmaceutical Science Handbook", Mack Pub. Co., New York, U.S.A. Examples of said forms include capsules, tablets, syrups and the like, containing 1 to 1000 mg of the active ingredient per unitary dose.
The following non limiting Examples further illustrate the invention. EXAMPLE 1
1-a 1-O-(6-bromohexyl)-2,3-O-isopropylideneσlycerol (III, n = 6)
A suspension of 0.188 g (6.2 mmoles) of 80% NaH in anhydrous DMF is added with a solution of 2.73 g (11.2 mmoles) of dibromohexane in DMF. A solution of 0.74 g (5.6 mmoles) of isopropylidene glycerol in DMF is dropped therein. The reaction mixture is left under stirring at room temperature for 18 hours, after that ethanol and water are added. The solution is extracted with ether, washed with a sodium chloride saturated solution, dried and solvent is evaporated off. The obtained crude product is purified by distillation under high vacuum, the excess 1, 6-dibromohexane distils at 42-44 °C/0.0 torr and 1-O-(6-bromohexyl)-2,3-O-isopropylideneglycerol distils at 70-72 °C/0.3 torr. The distillation residue is purified by flash chromatography on silica gel, eluting with dichloromethane. 0.680 g of 1-O-(6-bromohexyl)-2,3-O-isopropylideneglycerol are obtained (41% yield).
TLC: eluent dichloromethane:ethyl acetate (1:1); Rf =
0.45
IR (NaCl) cm-1: 2960, 2900, 2840, 1430, 1360, 1240,
1200
1H NMR (300 MHz, CDCl3) δ ppm: 1.25 (s, 3H) ; 1.30 (s, 3H); 1.30 (m, 4H) ; 1.50 (quintuplet, 2H) ; 1.75 (quintuplet, 2H) ; 3.25-3.45 (m, 6H) ; 3.63 (t, 1H) ; 3.96 (t, 1H); 4.17 (quintuplet, 1H).
13C NMR (75 MHz, CDCl3) δ ppm: 25.43, 25.59, 26.94, 28.11, 29.53, 32.88, 34.03, 67.15, 72.18, 71.86, 75.06, 109.80. Following the same procedure, the following products are obtained:
2R-1-O-(6-bromohexyl)-2,3-O-isopropylideneglycerol, starting from R-isopropylideneglycerol, 35% yield;
[α]D = - 7.1 (c 3.9, CHCl3)
2S-1-O-(6-bromohexyl)-2,3-O-isopropylideneglycerol, starting from S-isopropylideneglycerol, 30% yield;
[α]D = + 8.9 (c 1.0, CHCl3)
1-O-(10-bromodecyl)-2,3-O-isopropylideneglycerol (III, n=10), starting from 1,10-dibromodecane and isopropylideneglycerol, 35% yield;
TLC: eluent, petroleum ether:ethyl ether (1:1), Rf =
0.5
IR (NaCl) cm-1: 3000, 2950, 2850, 1460, 1350
1H NMR (60 MHz, CDCl3 ) δ ppm: 1.10-1.90 (m, 22H) ; 3.10- 4.20 (m, 9H)
1-b 1-O-(6-cyanohexyl)-2,3-O-isopropylideneglycerol
(IV, n = 6)
A solution of 1.19 g (24 mmoles) of sodium cyanide in 25 ml of DMSO is heated to 90°C and added with a solution of 6 g (20 mmoles) of 1-O-(6-bromohexyl)-2,3- O-isopropylideneglycerol in DMSO. The mixture is left at 90 °C for 1 hour, then water and ethyl ether are added, the two phases are separated and the organic phase is washed with a sodium chloride saturated solution, dried, and solvent is evaporated off to obtain 4.55 g of 1-O-(6-cyanohexyl)-2,3-O-isopropylideneglycerol (95% yield).
TLC: eluent, petroleum ether:ethyl ether (1:1), Rf = 0.43
IR (NaCl) cm-1: 3000, 2950, 2875, 2260, 1465, 1425 1H-NMR (300 MHz, CDCl3) δ ppm: 1.30 (s, 3H) ; 1.37 (s,
3H); 1.30-1.47 (m, 4H); 1.50-1.70 (m, 4H); 2.30 (t,
2H); 3.24-3.50 (m, 4H); 3.67 (dd, 1H) ; 4.02 (dd, 1H);
4.22 (quintuplet, 1H).
13C NMR (75 MHz, CDCl3) δ ppm: 17.26, 25.50, 25.54,
25.62, 26.99, 28.67, 29.45, 67.16, 72.27, 71.81, 75.12,
109.92, 120.34.
Following the same procedure, the following compounds are obtained:
2R-1-O-(6-cyanohexyl)-2,3-O-isopropylideneglycerol, 94% yield;
[α]D = -5.5 (c 11.2, CHCl3)
2S-1-O-(6-cyanohexyl)-2,3-O-isopropylideneglycerol, 95% yield;
[α]D = + 5.7 (c 11.2, CDCl3)
1-O-(10-cyanodecyl)-2,3-O-isopropylideneglycerol
(IV, n = 10), 79% yield;
TLC: eluent, petroleum ether: ethyl ether (1:1), Rf = 0.3
IR (NaCl) cm-1: 2975, 2925, 2850, 2250, 1450, 1360
1H NMR (300 MHz, CDCl3) δ ppm: 1.16-1.30 (m, 12H); 1.22 (s, 3H); 1.28 (s, 3H); 1.40 (m, 2H); 1.51 (quintuplet, 2H); 2.22 (t, 2H); 3.20-3.42 (m, 4H); 3.57 (dd, 1H); 3.90 (dd, 1H); 4.13 (quintuplet, 1H).
1-c 1-O-(6-cyanohexyl)glycerol (V, n = 6)
200 ml of a 2N HCl solution are added to a solution of 4 g (16.6 mmoles) of 1-O-(6-cyanohexyl)-2,3-O)isopropylideneglycerol in 500 ml of THF. The reaction mixture is kept at room temperature and under stirring for two hours, then it is neutralized with NaHCO3, the two phases formed are separated and the aqueous phase is extracted with ethyl acetate. The two organic phases are combined, dried and concentrated, to obtain 2.78 g of 1-O-(6-cyanohexyl)glycerol as a colourless oil (83% yield).
TLC: eluent, dichloromethane: ethyl acetate (1:1), Rf = 0,09
IR (NaCl) cm-1: 3450, 2975, 2900, 2300, 1475, 1440
1H NMR (300 MHz, CDCl3) δ ppm: 1.15-1.35 (m, 4H) ; 1.35-1.55 (m, 4H); 2.20 (t, 2H) ; 3.30 (m, 4H) ; 3.38 (dd, 1H); 3.48 (dd, 1H) ; 3.67 (quintuplet, 1H).
13C NMR (75 MHz, CDCl3) δ ppm: 17.26, 25.42, 25.53, 28.61, 29.43, 64.47, 70.91, 71.73, 72.77, 120.30.
Following the same procedure, the following compounds are obtained:
2S-1-O-(6-cyanohexyl)glycerol, 75% yield;
[α]D = + 1.3 (c 1.2, CHCl3)
2R-1-O-(6-cyanohexyl) glycerol, 86% yield;
[α]D = -1.4 (c 1.2, CHCl3)
1-O-(10-cyanodecyl)glycerol (V, n = 10), 88% yield.
1H NMR (300 MHz, CDCl3) δ ppm: 1.25 (broad s, 10H) ; 1.35 (m, 2H); 1.47-1.65 (m, 4H) ; 2.30 (t, 2H) ; 3.40-3.50 (m, 4H); 3.60 (dd, 1H) ; 3.67 (dd, 1H) ; 3.82 (quintuplet, 1H).
1-d 1-O-(6-cyanohexyl)-3-O-tritylglycerol (VI, n = 6) 7.24 g (26 mmoles) of trityl chloride are added to a solution of 2.6 g (13 mmoles) of 1-O-(6-cyanohexyl)glycerol in 60 ml of pyridine. The solution is stirred at room temperature for 22 hours, then it is poured onto an ice-water mixture and extracted with ethyl ether. The organic phase is washed with 0.1N HCl to acid pH, 5% NaHCO3 and a sodium chloride saturated aqueous solution, in this order. The reaction mixture is dried and solvent is evaporated off, to obtain a crude product which is purified by flash chromatography on silica gel, eluting with mixtures of petroleum ether:ethyl ether of increasing polarity, to obtain
3,98 g of 1-O-(6-cyanohexyl)-3-O-tritylglycerol (69% yield).
TLC: eluent petroleum ether:ethyl ether (1:1), Rf =
0.14
IR (NaCl) cm-1: 3650, 3475, 3100, 3050, 3000, 2950,
2850, 2250, 1600, 1475, 1430
1H NMR (300 MHz, CDCl3) δ ppm: 1.25-1.70 (m, 8H) ; 2.27
(t, 2H); 2.40 (broad s, 1H) ; 3.15 (m, 2H) ; 3.36-3.55
(m, 4H); 3.93 (quintuplet, 1H) ; 7.15-7.45 (m, 15H).
13C NMR (75 MHz, CDCl3) δ ppm: 17.28, 25.49, 25.58,
28.69, 29.54, 64.94, 70.24, 71.58, 72.50, 87.07,
120.35, 127.66, 128.43, 129.25, 144.48.
Following the same procedure, the following compounds are obtained:
2R-1-O-(6-cyanohexyl)-3-O-tritylglycerol, 66% yield;
[OC]D = + 2.9 (c 0.3, CHCl3)
2S-1-O-(6-cyanohexyl)-3-O-tritylglycerol, 60% yield;
[α]D = -3.3 (c 0.9, CHCl3)
1-O-(10-cyanodecyl)-3-O-tritylglycerol (VI, n = 10), 66% yield;
IR (NaCl) cm-1: 3500, 3100, 3060, 3040, 2950, 2850, 2250, 1590, 1385
1H NMR (300 MHz, CDCl3) δ ppm: 1.22 (broad s, 10H); 1.40 (m, 2H); 1.50 (m, 2H); 1.62 (quintuplet, 2H) ; 2.27 (t, 2H); 2.42 (broad s, 1H) ; 3.15 (m, 2H) ; 3.35-3.54 (m, 4H); 3.92 (quintuplet, 1H); 7.15-7.45 (m, 15H). 1-e 1-O- ( 6-cvanohexvl ) -2-O-methyl-3-O-tritylglycerol
(VII, n = 6, R2 = methyl)
A solution of 3.3 g (7.45 mmoles) of 1-O-(6-cyanohexyl)-3-O-tritylglycerol in 10 ml of benzene is added to a suspension of 1.78 g (8.94 mmoles) of 20% KH in anhydrous benzene. The reaction mixture is stirred for 30 minutes at room temperature, then it is added with 4.22 g (29 mmoles) of methyl iodide in 10 ml of benzene. The reaction mixture is stirred at room temperature for 3 hours, then a few methanol, water and ethyl ether are added. The two formed phases are separated and the aqueous phase is extracted with ethyl ether. The two ether phases are combined, dried and solvent is evaporated off, to obtain a crude product which is purified by silica gel chromatography, eluting with mixtures of petroleum ether :ethyl ether of increasing polarity, to obtain 2.83 g of 1-O-(6-cyanohexyl)-2-O-methyl-3-O-tritylglycerol (83% yield). TLC: eluent petroleum ether:ethyl ether (1:1), Rf = 0.3
IR (NaCl) cm-1:3030, 3020, 3010, 2900, 2850, 2250, 1600, 1475
1H NMR (300 MHz, CDCl3) δ ppm: 1.25-1.65 (m, 8H) ; 2.27 (t, 2H); 3.17 (m, 2H) ; 3.38 (s, 3H) ; 3.35-3.60 (m, 5H) ; 7.15-7.47 (m, 15H) ;.
13C NMR (75 MHz, CDCl3) δ ppm: 17.26, 25.52, 25.56, 28.70, 29.54, 58.5, 63.16, 71.33, 71.66, 80.26, 87.04, 120.38, 127.57, 128.38, 128.53, 128.60, 129.32, 144.7. Following the same procedure, the following compounds are obtained:
2R-1-O-(6-cyanohexyl)-2-O-methyl-3-O-tritylglycerol, 84% yield;
[α]D = +7.9 (c 4.8, CHCl3)
2S-1-O-(6-cyanohexyl)-2-O-methyl-3-O-tritylglycerol, 40% yield;
[α]D = -5 . 2 ( c 9.8, CHCl3)
1-O-(10-cyanodecyl)-2-O-methyl-3-O-tritylglycerol
(VII, n = 10, R2 = methyl), 91% yield;
IR (NaCl) cm-1: 3075, 3050, 2950, 2870, 2250, 1600
1H-NMR (300 MHz, CDCl3 ) δ ppm: 1.24 (broad s, 10H) ; 1.40 (quintuplet, 2H) ; 1.50 (quintuplet, 2H) ; 1.60 (m, 2H); 2.30 (t, 2H) ; 3.15 (m, 2H); 3.38 (s, 3H) ; 3.35- 3.60 (m, 5H); 7.15-7.50 (m, 15H).
1-f 1-O-(7-oxooctadecyl)-2-O-methylqlycerol (XI, n = 6,
Z = oxygen, R 1 = undecyl, R2 = methyl)
0.260 g of Mg and a crystal of I2 in anhydrous ethyl ether are placed into a 3-necked flask, then a solution of 2.060 g (8.75 mmoles) of 1-bromoundecane in ethyl ether is dropped therein. The reaction mixture is refluxed for 1 hour, after that ethyl ether is evaporated off and 10 ml of anhydrous benzene are added. A solution of 2.0 g (4.38 mmoles) of 1-O-(6-cyanohexyl)-2-O-methyl-3-O-tritylglycerol in anhydrous benzene is added. The mixture is refluxed for 6 hours and left for 3 days at room temperature, after that solvent is evaporated off and 100 ml of dioxane and 30 ml of HCl IN are added thereto. The reaction mixture is refluxed for two hours, neutralized with 5% NaHCO3 and extracted with ethyl ether. After drying and evaporating the solvent, a crude product is obtained which is purified by flash chromatography on silica gel. By eluting with mixtures of petroleum ether:ethyl ether of increasing polarity, 0.575 g of 1-O-(7-oxooctadecyl)-2-O-methylglycerol are obtained (35% yield).
IR (NaCl) cm-1: 3450, 2945, 1714, 1468, 1406, 1384, 1131
1H-NMR (300 MHz, CDCl3) δ ppm: 0.82 (t, 3H) ; 1.20 (m,
20H); 1.50 (m, 6H) ; 2.30 (t, 4H) ; 3.40 (s, 3H) ; 2.38
(broad s, 1H) ; 3.32-3.42 (m, 3H); 3.47 (m, 2H) ; 3.55
(dd, 1H); 3.69 (dd, 1H).
13C NMR (75 MHz, CDCl3) δ ppm: 14.12, 22.72-31.99,
42.72, 42.88, 57.90, 62.35, 70.59, 71.81, 80.63,
212.36.
Following the same procedure, the following compounds are obtained:
2S-1-O-(7-oxooctadecyl)-2-O-methylglycerol, 35% yield; [α]D = -7.3 (c 1.3, CHCl3)
2R-1-O-(7-oxaoctadecyl)-2-O-methylglycerol, 8.5% yield; [α]D = +7.5 (c 1.3, CDCl3)
1-O-(11-phenyl-11-oxoundecyl)-2-O-methylglycerol (XI, n = 10, Z = oxygen, R1 = phenyl, R2 = methyl), 35.5% yield;
IR (NaCl) cm-1: 3450, 3050, 2925, 2850, 1660, 1585
1H-NMR (300 MHz, CDCl3) δ ppm: 1.22 (m, 12H) ; 1.50 (m,
2H); 1.65 (quintuplet, 2H); 2.65 (broad s, 1H) ; 2.90 (t, 2H); 3.39 (s, 3H) ; 3.32-3.42 (m, 3H); 3.46 (m, 2H) ;
3.55 (dd, 1H); 3.68 (dd, 1H) ; 7.32-7.50 (m, 3H); 7.90
(d, 2H).
13C NMR (75 MHz, CDCl3) δ ppm: 24.52-29.75, 38.82,
58.07, 62.80, 70.83, 72.19, 80.43, 128.60, 129.10, 133.47, 201.51.
1-g 1-O-(7-oxooctadecyl)-2-O-methylglycerol-3-phospho choline (I, n = 6, R1 = undecyl, R2 = methyl, R3 = hydrogen, R4, R5 and R6 = methyl)
0.125 g (0.3 mmoles) of 1-O-(7-oxooctadecyl)-2-O- methylglycerol dissolved in ethyl ether is added to a suspension of 0.134 g (0.55 mmoles) of 2- bromoethyldichlorophosphate and 0.15 ml (1.098 mmoles) of Et3N in anhydrous ethyl ether, at 0ºC. The reaction mixture is stirred at room temperature for 24 hours, after that 0.54 ml of a 0.1M KCl solution are added and stirring is continued at room temperature for 1.15 hours. The reaction mixture is extracted with ethyl ether, dried and solvent is evaporated off to obtain 0.150 g of 1-O-(7-oxooctadecyl)-2-O-methylglycero-3-(2-bromoethyl)phosphate (quantitative yield).
1H NMR (300 MHz, CDCl3) δ ppm: 0.82 (t, 3H) ; 1.20 (m, 20H) ; 1.50 (m, 6H) ; 2.35 (broad, 4H) ; 3.41 (s, 3H) ; 3.35-3.57 (m, 7H) ; 4.00-4.40 (m, 4H).
0.135 g (0.24 mmoles) of the above bromoethyl phosphate are dissolved in 5 ml of anhydrous CDCl3. in a closed reactor. The reaction mixture is cooled with outer bath of dry ice-acetone and 1 ml of trimethylamine is added, the reactor is closed and heated to 65 ºC for 18 hours. The solution is evaporated to dryness, to obtain a crude product which is purified by silica gel chromatography. Eluting with a chloroform:methanol 65:25 mixture and subsequently with a chloroform:methanol:water 65:25:4 mixture, 0.730 g of 1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phosphocholine are obtained (45% yield).
1H-NMR (300 MHz, CDCl3) δ ppm: 0.82 (t, 3H) ; 1.20 (broad s, 20H) ; 1.50 (quintuplet, 6H); 2.34 (broad, 4H); 3.31 (s, 9H); 3.37 (s, 3H); 3.30-3.50 (m, 5H);
3.75 (m, 2H); 3.77-3.92 (m, 2H); 4.25 (m, 2H).
13C NMR (75 MHz, CDCl3) δ ppm: 14.27, 22.84-32.09,
42.89, 43.12, 54.59, 58.04, 59.50, 65.02, 66.60, 70.60, 71.86, 80.06, 212.54.
Elementary analysis: Calculated for C 27H66NO12P: C,
51.66; H, 10.59; N, 2.23. Found: C, 51.64; H, 10.38; N,
2.41.
Following the same procedure, the following compounds are obtained:
2-O-methyl-1-O-(7-oxooctadecyl)-sn-glycero-3-phosphocholine, 36% yield;
[α]D = +1.1 (c 2.4, CHCl3)
2-O-methyl-3-O-(7-oxooctadecyl)-sn-glycero-1-phosphocholine, 56% yield;
[α]D = 0.9 (c 0.3, CHCl3)
1-O-(11-phenyl-11-oxoundecyl)-2-O-methylglycero-3-phosphocholine (I, n = 10, R1 = phenyl, R2 = methyl, R3 = hydrogen, R4, R5 and R6 = methyl), 5% yield;
1H NMR (300 MHz, CDCl3) δ ppm: 1.20 (m, 12H); 1.50 (m,
2H); 1.67 (m, 2H); 2.90 (t, 2H); 3.25 (s, 9H); 3.38 (s,
3H); 3.30-3.50 (m, 5H); 3.67 (m, 2H); 3.72-3.92 (m,
2H); 4.23 (m, 2H); 7.35-7.55 (m, 3H); 7.90 (d, 2H).
13C NMR (75 MHz, CDCl3) δ ppm : 25.79-30.95, 39.72, 54.99, 58.55, 60.68, 66.50, 67.82, 71.40, 73.06, 81.25,
129.40, 130.00, 134.40, 203.00.
Elementary analysis: Calculated for C26H52NO10P : C,
54.77; H, 9.20; N, 2.45. Found: C, 54.18; H, 9.04; N,
2.59.
EXAMPLE 2
2-a 1-Benzyloxy-6-bromohexane (XXIII, n = 6 A solution of 25 g (100 mmoles) of 1,6- dibromohexane in THF is added to a 80% NaH suspension in anhydrous THF. The reaction mixture is refluxed and 4.32 g (40 mmoles) of benzyl alcohol dissolved in THF are dropped therein. At the end of the addition, reflux is continued for 1 hour, after that the solution is poured into an ice-water mixture and extracted with ethyl ether. The ether phase is washed with a sodium chloride saturated solution, dried and concentrated. The resulting crude product is purified by distillation under high vacuum, 1-benzyloxy-6-bromohexane distilling at 86°C/l torr, to obtain 4.5 g (35% yield).
1H NMR (300 MHz, CDCl3) δ ppm: 1.42 (m, 4H) ; 1.62 (quintuplet, 2H) ; 1.85 (quintuplet, 2H) ; 3.37 (t, 2H) ; 3.45 (t, 2H) ; 4.50 (s, 2H) ; 7.35 (m, 5H).
2-b 1-Benzyloxy-3-bromopropane (XXIII, n = 3)
A solution of 4.78 g (28 mmoles) of benzyl bromide in THF and a tip of tetrabutylammonium iodide is added to a suspension of 0.474 g (15.4 mmoles) of 80% NaH in anhydrous THF. Temperature is kept at 30°C and 2 g (14 mmoles) of 3-bromopropanol are added. At the end of the addition, temperature is kept at 30 ºC for 4 hours, after that a few methanol and water are added and the reaction mixture is extracted with ethyl ether, dried and solvent is evaporated off. The resulting crude product is purified by distillation under high vacuum: 1-benzyloxy-3-bromopropane distils at 80°C/1, to obtain 1.34 g (42% yield).
1H NMR (300 MHz, CDCl3) δ ppm: 2.10 (m, 2H) ; 3.47 and 3.55 (2 m, 4H); 4.50 (s, 2H) ; 7.30 (m, 5H). Following the same procedure, the following compound is obtained:
1-benzyloxy-11-bromoundecane (XXIII, n = 11), 40% yield.
2-c 7-Benzyloxyheptanonitrile (XXIV, n = 6)
A solution of 0.576 g (11.7 mmoles) of sodium cyanide in DMSO is heated to 90°C and added with a solution of 3 g (11.7 mmoles) of 1-benzyloxy-6-bromohexane in DMSO. The reaction mixture is left at 90°C for 16 hours, then water and ethyl ether are added, the two phases are separated and the organic phase is washed with a NaCl saturated solution, dried and solvent is evaporated off to obtain 2.053 g of 7-benzyloxyheptanonitrile (87% yield).
1H NMR (300 MHz, CDCl3) δ ppm: 1.40 (m, 4H) ; 1.57 (m,
4H); 2.20 (t, 2H) ; 3.40 (t, 2H) ; 4.45 (s, 2H) ; 7.30 (m,
5H).
Following the same procedure, the following compounds are obtained:
4-benzyloxybutanonitrile (XXIV, n = 3), 66% yield;
1H NMR (300 MHz, CDCl3) δ ppm: 1.91 (quintuplet, 2H) ;
2.46 (t, 2H); 3.55 (t, 2H) ; 4.49 (s, 2H); 7.20-7.35 (m,
5H).
12-benzyloxydodecanonitrile (XXIV, n = 11), 70% yield;
1H NMR (300 MHz, CDCl3) δ ppm: 1.20-1.40 (m, 14H ) ; 1.57
(m, 4H); 2.25 (t, 2H) ; 3.42 (t, 2H) ; 4.45 (s, 2H) ;
7.20-7.35 (m, 5H).
2-d 1-Benzyloxy-7-octadecanone (XXV, n = 6, R1 = undecyl)
0.320 g (13.3 mmoles) of Mg and a iodine crystal in anhydrous ethyl ether are placed into a flask. A solution of 2.7 g (11.5 mmoles) of bromoundecane in ethyl ether is dropped therein. The mixture is refluxed for 1 hour, after that a hot solution of 1 g (4.6 mmoles) of 7-benzyloxyheptanonitrile in anhydrous benzene is added. The reaction mixture is refluxed for 6 hours, after that a 10% H2SO4 cold solution is added, keeping reflux for 1 hour. The two phases are separated and the organic phase is washed with a 5% NaHCO3 solution, dried and solvent is evaporated off, to obtain a crude product which is purified by silica gel flash chromatography. Eluting with mixtures of petroleum ether: ethyl ether of increasing polarity, 0.716 g of 1-benzyloxy-7-octadecanone are obtained (41.5% yield).
IR (NaCl) cm-1: 3000, 2900, 1707, 1460, 1410, 1260
1H NMR (300 MHz, CDCl3) δ ppm: 0.80 (t, 3H) ; 1.20 (m,
20H); 1.50 (m, 6H); 2.25 (t, 4H); 3.37 (t, 2H) ; 4.40 (s, 2H); 7.20 (m, 5H).
Following the same procedure, the following compounds are obtained:
1-benzyloxy-4-octadecanone (XXV, n = 3, R1 tetradecyl), 24% yield;
Melting point: 36-37°C
IR (NaCl) cm-1: 3000, 2950, 2850, 1714, 1450, 1360
1H NMR (300 MHz, CDCl3) δ ppm: 0.84 (t, 3H) ; 1.20
(broad s, 22H); 1.50 (quintuplet, 2H) ; 1.85
(quintuplet, 1H) ; 2.35 (t, 2H) ; 2.48 (t, 2H) ; 3.44 (t,
2H) ; 4.45 (s, 2H) ; 7.22-7.37 (m, 5H).
18-benzyloxy-7-octadecanone (XXV, n = 11, R1 = hexyl), 45% yield;
1H NMR (300 MHz, CDCl3) δ ppm: 0.85 (t, 3H) ; 1.30 (broad s, 20H); 1.55 (m, 6H); 2.35 (t, 4H); 3.42 (t, 2H); 4.45 (s, 2H); 7.20-7.32 (m, 5H).
2-e 1-Benzyloxy-7-ethylenedioxyoctadecane (XXVI, n = 6, Z = ethylenedioxy, R1 = undecyl)
A mixture of 2.7 g (7.22 mmoles) of 1-benzyloxy-7-octadecanone, 15.2 g (245 mmoles) of ethylene glycol and 1.82 g (9.6 mmoles) of p-toluenesulfonic acid in 120 ml of anhydrous benzene is refluxed for 24 hours in a Dean-Stark apparatus. At the end of this time, the reaction mixture is washed first with a 10% NaOH solution, then with water, it is dried and solvent is evaporated off, to obtain 2.51 g of 1-benzyloxy-7-ethylenedioxyoctadecane in form of an oil (83% yield). -H NMR (300 MHz, CDCl3) δ ppm: 0.85 (t, 3H); 1.30 (m, 18H); 1.55 (m, 6H); 3.42 (t, 2H); 3.90 (s, 4H); 4.47 (s, 2H); 7.20-7.35 (m, 5H).
Following the same procedure, the following compound is obtained:
l-benzyloxy-4-ethylenedioxyoctadecane (XXVI, n = 3, Z = ethylenedioxy, R1 = tetradecyl), 92% yield;
1H NMR (300 MHz, CDCl3) δ ppm: 0.90 (t, 3H); 1.30 (broad s, 24H); 1.60 (m, 2H); 1.70 (s, 4H); 3.47 (m, 2H); 3.92 (s, 4H); 4.50 (s, 2H); 7.25-7.35 (m, 5H).
2-f 7-Ethylenedioxy-1-octadecanol (XXVII, n = 6, Z = ethylenedioxy, R1 = undecyl)
0.81 g of 20% Pd(OH)2/c are added to a solution of 1.25 g (3 mmoles) of 1-benzyloxy-7-ethylenedioxyoctadecane in 81 ml of a methanol:water 9:1 mixture, and the reaction mixture is left under stirring and hydrogen atmosphere at room temperature for 16 hours. The reaction mixture is filtered and evaporated to dryness, to obtain 0.950 g of 7-ethylenedioxy-1- octadecanol (97% yield).
1H-NMR (300 MHz, CDCl3) δ ppm: 0.75 (t, 3H) ; 1.15-1.25 (m, 24H); 1.50 (m, 6H) ; 2.52 (broad s, 1H); 3.50 (t, 2H); 3.80 (s, 4H).
Following the same procedure, the following compounds are obtained:
4-ethylenedioxy-1-octadecanol (XXVII, n = 3, Z ethylenedioxy, R1 = tetradecyl), 86% yield;
Melting point: 39-41°C
1H NMR (300 MHz, CDCl3) δ ppm: 0.82 (t, 3H) ; 1.20 (broad s, 24H) ; 1.52-1.75 (m, 6H) ; 1.87 (broad s, 1H) ; 3.60 (t, 2H); 3.91 (s, 4H).
18-hydroxy-7-octadecanone (XXVII, n = 11 , Z = oxygen, R1 = hexyl) starting from XXV (n, 11, R1 = hexyl), 80% yield;
1H NMR (300 MHz, CDCl3) δ ppm: 0.85 (t, 3H) ; 1.25 (broad s, 20H) ; 1.55 (s, 6H) ; 2.35 (t, 4H) ; 3.60 (t, 2H).
2-g 7-Ethylenedioxyoctadecyl methanesulfonate (IX, n = 6, Z = ethylenedioxy, R1 = undecyl)
1.45 ml (10.74 mmoles) of Et3N are added to a solution of 1.85 g (5.37 mmoles) of 7-ethylenedioxy-1-octadecanol in anhydrous dichloromethane. The reaction mixture is cooled to 0°C and 1.23 g (10.74 mmoles) of methanesulfonyl chloride are added. At the end of the addition, the reaction mixture is maintained at 0°C for 4 hours, after that it is washed with water, dried and solvent is evaporated off. The obtained crude product is purified by silica gel chromatography. Eluting with mixtures of petroleum ether:ethyl ether of increasing polarity, 1.79 g of 7-ethylenedioxyoctadecyl methanesulfonate are obtained (79% yield).
1H NMR (300 MHz, CDCl3) δ ppm: 0.82 (t, 3H) ; 1.15-1.40 (m, 24H); 1.52 (m, 4H) ; 1.72 (quintuplet, 2H) ; 2.95 (s, 3H); 3.87 (s, 4H) ; 4.18 (t, 2H) .
Following the same procedure, the following compounds are obtained:
4-ethylenedioxyoctadecyl methanesulfonate (IX, n - 3, Z - ethylenedioxy, R1 = tetradecyl), 60% yield;
Melting point: 52-55ºC
1H NMR (300 MHz, CDCl3) & ppm: 0.84 (t, 3H) ; 1.20 (broad s, 24H) ; 1.55 (m, 2H) ; 1.70 (m, 2H) ; 1.80 (m, 2H); 2.95 (s, 3H) ; 3.90 (s, 4H) ; 4.22 (t, 2H).
12-oxooctadecyl methanesulfonate (IX, n = 11, Z oxygen, R1 = hexyl) starting from XXVII (n, 11, Z = oxygen, R1 = hexyl), 87% yield;
1H NMR (300 MHz, CDCl3) δ ppm: 0.80 (t, 3H) ; 1.15-1.35 (m, 14H); 1.48 (m, 4H) ; 1.67 (quintuplet, 2H) ; 2.32 (t, 4H); 2.92 (s, 3H) ; 4.15 (t, 2H).
EXAMPLE 3
3-a 1-O-(4-ethylenedioxyoctadecyl)-2-O-methyl-3-O-benzylqlycerol (X, n = 3, Z ethylenedioxy, R1 tetradecyl, R2 = methyl)
A solution of 0.580 g (2.9 mmoles) of 3-O-benzyl-2-O-methylglycerol in toluene is added to a suspension of 0.684 g (12.2 mmoles) of powder KOH in 24 ml of anhydrous toluene. The reaction mixture is refluxed for 1 hour in a Dean-Stark apparatus. After that, 1 g (2.4 mmoles) of 4-ethylenedioxyoctadecyl methanesulfonate dissolved in 16 ml of toluene is added. Reflux in the Dean-Stark apparatus is maintained for 5 hours, then toluene is evaporated off, some water is added and the mixture is extracted with ethyl ether. The ether phase is washed with water to neutral pH. The organic phase is dried and evaporated, to obtain a crude product which is purified by silica gel chromatography. Eluting with mixtures of petroleum ether: ethyl ether of increasing polarity, 0.569 g of 1-O-(4- ethylenedioxyoctadecyl)-2-O-methyl-3-O-benzylglycerol are obtained (46% yield).
1H NMR (300 MHz, CDCl3) δ ppm: 0.85 (t, 3H) ; 1.20 (broad s, 24H) ; 1.50-1.62 (m, 6H) ; 3.40 (s, 3H) ; 3.35- 3.57 (m, 7H); 3.90 (s, 4H) ; 4.52 (s, 2H) ; 7.20-7.35 (m, 5H).
13C NMR (75 MHz, CDCl3) δ ppm: 14.31, 22.90-30.18, 32.16, 33.70, 37.53, 58.37, 65.28, 70.16, 70.70, 72.04, 73.81, 79.78, 112.19, 128.17, 128.22, 128.94, 138.88. Following the same procedure, the following compounds are obtained:
1-O-(7-ethylenedioxyoctadecyl)-2-O-methyl-3-O-benzylglycerol (X, n = 6, Z = ethylenedioxy, R1 = undecyl, R2 = methyl), 30% yield;
1H NMR (300 MHz, CDCl3) δ ppm: 0.85 (t, 3H) ; 1.20-1.35 (m, 24H); 1.52 (m, 6H) ; 3.40 (s, 3H) ; 3.35-3.55 (m, 7H); 3.87 (s, 4H) ; 4.50 (s, 2H) ; 7.20-7.35 (m, 5H).
1-O-(12-oxooctadecyl)-2-O-methyl-3-O-benzylglycerol (X, n = 11, Z = oxygen, R1 = hexyl, R2 = methyl) starting from IX (n = 11, Z = oxygen, R1 = hexyl), 31% yield;
1H NMR (300 MHz, CDCl3)δ ppm: 0.85 (t, 3H) ; 1.22 (m, 20H); 1.65 (m, 6H) ; 2.35 (t, 4H) ; 3.43 (s, 3H); 3.35-3.57 (m, 7H) ; 4.50 (s, 2H) ; 7.30 (m, 5H).
13C NMR (75 MHz, CDCl3) δ ppm : 14.21, 22.70-31.84, 43.08, 58.32, 70.15, 70.68, 72.09, 73.79, 79.78, 128.15, 128.22, 128.92, 138.89, 212.65.
3-b 1-O-(4-oxooctadecyl)-2-O-methylglycerol (XI, n = 3,
Z = oxygen, R1 = tetradecyl, R2 = methyl)
0.3 ml of HCl (35%) and 0.345 g of 20% Pd(OH)2/C are added to a solution of 0.648 g (1.28 mmoles) of 1- O-(4-ethylenedioxyoctadecyl)-2-O-methyl-3-O-benzylglycerol in 34 ml of a methanol :water 9:1 mixture. The reaction mixture is left under stirring and hydrogen atmosphere at room temperature for 15 hours, then it is filtered and evaporated to dryness, to obtain 0.425 g of 1-O-(4-oxooctadecyl)-2-O-methylglycerol (89% yield). 1H NMR (300 MHz, CDCl3) δ ppm: 0.82 (t, 3H); 1.20 (broad s, 22H); 1.50 (quintuplet, 2H); 1.79 (quintuplet, 2H); 2.34 (t, 2H); 2.42 (t, 2H); 3.40 (s, 3H); 3.30-3.42 (m, 3H); 3.45 (m, 2H); 3.57 (dd, 1H); 3.68 (dd, 1H).
13C NMR (75 MHz, CDCl3) δ ppm: 14.28, 22.88, 23.86, 24.06, 29.46-32.14, 39.38, 43.19, 58.10, 62.65, 70.60, 71.09, 80.36, 212.05.
Following the same procedure, the following product is obtained:
1-O-(7-oxooctadecyl)-2-O-methylglycerol (XI, n = 6, Z = oxygen, R1 = undecyl, R2 = methyl), 90% yield, compound which is described in example 1-f.
Following the same procedure, except for the acid medium, the following compounds are obtained:
1-O-(12-oxooctadecyl)-2-O-methylglycerol (XI, n = 11, Z = oxygen, R1 = hexyl, R2 = methyl), 85% yield;
1H-NMR (300 MHz, CDCl3) δ ppm: 0.80 (t, 3H); 1.15 (broad s, 20H); 1.47 (m, 6H); 2.30 (t, 4H); 2.50 (broad s, 1H); 3.35 (s, 3H) ; 3.30-3.38 (m, 3H) ; 3.44 (m, 2H) ; 3.55 (dd, 1H); 3.66 (dd, 1H).
13C NMR (75 MHz, CDCl3) δ ppm: 14.16, 22.65-31.80, 43.02, 58.04, 62.80, 70.86, 72.20, 80.36, 212.68.
1-O-(7-ethylenedioxyoctadecyl)-2-O-methylglycerol (XI, n = 6, Z = ethylenedioxy, R1 = undecyl, R2 = methyl), 85% yield;
1H-NMR (300 MHz, CDCl3) δ pPm: 0.82 (t, 3H) ; 1.20-1.35 (m, 24H); 1.52 (m, 6H) ; 3.42 (s, 3H) ; 3.35-3.42 (m, 3H); 3.47 (m, 2H) ; 3.59 (dd, 1H) ; 3.71 (dd, 1H) ; 3.90 (s, 4H).
13C NMR (75 MHz, CDCl3) δ ppm: 14.29, 22.88-32.14, 37.30, 37.40, 58.10, 62.95, 65.23, 70.94, 72.22, 80.25, 112.36.
3-c 1-O-(4-oxooctadecyl)-2-O-methylglycero-3-phosphocholine
(I, n = 3, R1 = tetradecyl, R2 = methyl, R3 = hydrogen, R4, R5 and R6 = methyl)
0.382 g (1.02 mmoles) of 1-O-(4-oxooctadecyl)-2-O-methylglycerol dissolved in ethyl ether are added to a suspension of 0.457 g (1.89 mmoles) of 2-bromoethyl dichlorophosphate and 0.53 ml (3.75 mmoles) of Et3N in anhydrous ethyl ether, at 0°C. The reaction mixture is left under stirring at room temperature for 24 hours, after that 1,84 ml of a 0.1M KCl solution are added and the mixture is left under stirring at room temperature for two hours. The reaction mixture is extracted with ethyl ether, dried and solvent is evaporated off, to obtain 0.471 g of 1-O-(4-oxooctadecyl)-2-O-methylglycero-3-(2-bromoethyl)phosphate (quantitative yield). 1H-NMR (300 MHz, CDCl3) δ ppm: 0.80 (t, 3H); 1.20 (broad s. 22H); 1.45 (m, 2H); 1.75. (quintuplet, 2H); 2.31 (t, 2H); 2.39 (t, 2H); 3.40 (s, 3H); 3.30-3.52 (m, 7H); 3.95-4.12 (m, 2H); 4.20 (m, 2H); 9.60 (broad s, 1H).
0.471 g (0.84 mmole) of the above bromophosphate dissolved in 20 ml of anhydrous CDCl3 are placed into a closed reactor, which is cooled with a dry ice-acetone outer bath. 1.1 ml of trimethylamine are added, the reactor is closed and heated to 65 °C for 24 hours. The reaction mixture is evaporated to dryness, to obtain a crude product which is purified by silica gel chromatography. Eluting with a chloroform:methanol 95:5 mixture, increasing polarity to a chloroform:methanol 65:25 mixture, then with a chloroform:methanol: water 65:25:1 mixture, increasing polarity to a chloroform:methanol:water 65:35:4 final mixture, 0.295 g of 1-O-(4-oxooctadecyl)-2-O-methylglycero-3-phosphocholine are obtained (54% yield).
1H-NMR (300 MHz, CDCl3) δ ppm: 0.80 (t, 3H); 1.10-1.25 (m, 22H); 1.43 (m, 2H); 1.72 (quintuplet, 2H); 2.29 (t, 2H); 2.35 (t, 2H); 3.32 (s, 9H); 3.36 (s, 3H); 3.25- 3.47 (m, 5H); 3.75 (m, 4H); 4.21 (m, 2H).
13C NMR (75 MHz, CDCl3) δ ppm: 14.25, 22.83-32.1, 39.34, 43.12, 54.50, 58.05, 59.55, 64.78, 66.51, 70.60, 70.92, 79.98, 211.91.
Elementary analysis: calculated for C27H63NO10·5P : C, 53.98; H, 10.57; N, 2.33. Found: C, 54.17; H, 10.52; N, 2.34.
Following the same procedure, the following compound is obtained: 1-O-(12-oxooctadecyl)-2-O-methylglycero-3- phosphocholine (I, n = 11, R1 = hexyl, R2 = methyl, R3 = hydrogen, R4, R5 and R6 = methyl), 25% yield;
1H-NMR (300 MHz, CDCl3) δ ppm: 0.80 (t, 3H) ; 1.17 (m, 20H); 1.47 (m, 6H) ; 2.30 (t, 4H) ; 3.34 (s, 9H) ; 3.35 (s, 3H); 3.27-3.47 (m, 5H) ; 3.70-3.90 (m, 4H) ; 4.25 (m, 2H).
13C NMR (75 MHz, CDCl3) δ ppm: 14.07, 22.52-31.67, 43.06, 54.59, 57.99, 59.70, 65.52, 66.45, 70.06 72.01, 19 . 10 , 213.0.
Elementary analysis: calculated for C27H62NO10·P : C, 54.82; H, 10.49; N, 2.36. Found: C, 54.63; H, 10.44; N, 2.58.
EXAMPLE 4
1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phosphoserine (I, n = 6, R1 = undecyl, R2 = methyl, R3 = carboxyl, R4, R5 and R6 = hydrogen)
0.150 g (0.36 mmole) of 1-O-(7-ethylenedioxyoctadecyl)-2-O-methylglycerol dissolved in anhydrous acetonitrile is added to a suspension of 0.350 g (5.14 mmoles) of imidazole, 0.135 ml (1.54 mmoles) of PCl3
0.756 ml (5.43 mmoles) of Et3N and anhydrous acetonitrile, at 0°C and under inert atmosphere. The reaction mixture is stirred for 4 hours at room temperature; 4 ml of water are added and stirring is continued at room temperature for 45 minutes. The reaction mixture is evaporated to dryness and redissolved in 11 ml of a pyridine:triethylamine 4:1 mixture, evaporated to dryness, dissolved in CHCl3 and the organic phase is washed with water, dried and solvent is evaporated off to obtain the phosphonate triethylammonium salt corresponding to the starting alcohol (quantitative yield).
1H-NMR (300 MHz, CDCl3) δ ppm: 0.80 (t, 3H) ; 1.20 (m, 24H); 1.30 (t, 9H); 1.50 (m, 6H) ; 3.20 (q, 6H); 3.30 (s, 3H); 3.25-3.45 (m, 5H); 3.85 (s, 4H) ; 3.75-3.90 (m, 2H); 6.63 (d, 1H).
Following the same procedure, the triethylammonium salt corresponding to 1-O-(12-oxooctadecyl)-2-O-methylglycerol phosphonate is obtained:
1H-NMR (300 MHz, CDCl3) δ ppm: 0.65 (t, 3H) ; 0.95-1.15 (m, 33H); 1.35 (m, 6H) ; 2.15 (t, 4H) ; 2.85 (q. 6H) ; 3.20 (s, 3H); 3.10-3.40 (m, 5H) ; 3.75-4.00 (m, 2H) ; 6.62 (d, 1H).
0.061 ml (0.5 mmole) of pivaloyl chloride is added to a mixture of 0.120 g (0.25 mmole) of 1-O-(7-ethylenedioxyoctadecyl)-2-O-methylglycerol phosphonate, 0.132 g (0.4 mmole) of N-benzyloxycarbonylserine benzyl ester and 3.5 ml of pyridine. The reaction mixture is stirred for 1.5 hours at room temperature, 0.2 ml of water and 0.127 g (0.5 mmole) of iodine are added. The mixture is stirred at room temperature for 25 minutes; then it is added with CDCl3, washed with sodium metabisulfite, dried and solvent is evaporated off, to obtain a crude product which is purified by flash chromatography on a silica gel column. Eluting with petroleum ether:chloroform mixtures of increasing polarity and then with chloroform:methanol mixtures of increasing polarity, 0.035 g of 1-O-(7-ethylenedioxyoctadecyl)-2-O-methylglycerol-3-phosphobenzyl(N-ca(bobenzyloxy)serine are obtained (18% yield). 1H-NMR (300 MHz, CDCl3) δ ppm: 0.85 (t, 3H); 1.10-1.35 (m, 24H); 1.40-1.60 (m, 6H); 3.30 (s, 3H); 3.25-3.42 (m, 5H); 3.86 (s, 4H); 3.85-4.00 (m, 2H); 4.1-5-4.35 (m, 2H); 4.55 (m, 1H); 4.70-5.20 (m, 4H); 7.15-7.30 (m, 10H).
Following the same procedure, the following compound is obtained:
1-O-(12-oxooctadecyl)-2-O-methylglycerol-3- phosphobenzyl(N-carbobenzyloxy)serine, 1% yield;
1H-NMR (300 MHz, CDCl3) δ ppm: 0.85 (t, 3H); 1.20 (broad s, 24H); 1.52 (m, 6H); 2.35 (t, 4H); 3.40 (s, 3H); 3.30-3.55 (m, 5H); 3.92-4.17 (m, 2H); 4.30 (m, 1H); 4.44 (m, 1H); 4.58 (m, 1H); 5.05-5.22 (m, 4H); 7.20-7.35 (m, 10H).
0.035 g (0.045 mmole) of 1-O-(7- ethylenedioxyoctadecyl)-2-O-methylglycerol-3-phosphobenzyl(N-carbobenzyloxy) serine are dissolved in 7.5 ml of a methanol :water 9:1 mixture, 0.2 ml of HCl (35%) and 0.005 g of 20% Pd(OH)2/C are added. The reaction mixture is left under stirring and hydrogen atmosphere at room temperature for 16 hours, then it is filtered, evaporated to dryness and purified by preparative chromatography (silica gel, fluorescence developer), extracted with a chloroform:methanol 1:1 mixture. 0.005 g of 1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phosphoserine is obtained (20% yield). TLC: eluent chloroform:methanol:water (65:25:4), Rf = 0.09
According to the same procedure, except for the acid medium, the following product is obtained:
1-O-(12-oxooctadecyl)-2-O-methylglycerol-3-phosphose- rine
(I, n = 11, R1 = hexyl, R2 = methyl, R3 = carboxyl, R4, R5 and R6 = hydrogen), 57% yield;
TLC: eluent chloroform:methanol:water (65:25:4), Rf = 0.11.
EXAMPLE 5
Evaluation of cytotoxic activity
Suspensions of HL-60 line cells, obtained starting from human promyelocyte leukemia, are incubated in 6- well culture plates for 24 hours at 37°C, at a concentration of 1 × 106 cells/ml, in the presence of the test products, which have previously been dissolved in the culture medium. Cytotoxicity is measured by means of the Trypan Blue exclusion technique (Practical Immunology, pp 29-32, Oxford, England : Blackwell Scientific Publications, 1976). Cytotoxic activities of the products, expressed as IC50, are reported in Tables IA and IB.
EXAMPLE 6
Evaluation of the anti-platelet action
Platelets are prepared starting from blood samples obtained by means of a carotid cannula from male albino New Zealand rabbits (2-2.5 kg), subjected to anticoagulant treatment with sodium citrate dihydrate (3.2%) 1:10. Blood is centrifuged (150xg, 10 min, 22°C) to separate the platelet-rich plasma fraction (PRP) from erythrocytes. Platelets are separated from plasmatic components by serial washings and centrifugations (Blood, 1986, 62:337), thereafter they are suspended in a physiological buffer at a concentration of 5 × 105/ml. A 400 μl volume of the above mentioned suspension is placed into the cuvette of a multicanal aggregometer (Aggrecoder HU) where it is kept at constant temperature and stirring. After stabilization, platelets are added with 50 μl of physiological buffer containing 100 μM acetylsalicylic acid and apirase 5 mg/ml, in order to block any non PAF-related platelet activation metabolic pathways. Subsequently the platelet suspensions are added with 50 μl of the different solutions to be tested. Changes in light transmission, due to platelet aggregation in the suspension, are recorded and expressed as percent platelet aggregation, according to the procedure by Born and Cross (J. Physiol., 1962, 162:67). Standard curve is performed with PAF within a concentration range of 0.1-1 nM.
The results from the tests in which PAF activity was measured are reported in Tables IIA, IIB.
EXAMPLE 7
Inhibition of PAF-induced platelet aggregation
Capability to inhibit PAF-induced platelet aggregation is evaluated by means of the aggregometry technique in suspensions of rabbit washed platelets, obtained by means of the same procedure as described in Example 6. For this purpose, the inhibition potency of the products under test, expressed as the corresponding IC50, is evaluated on the maximum aggregation values obtained in PAF standard curve. The results obtained from the tests, in which PAF-antagonist activity was measured, are reported in Tables IIA, IIB.
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001

Claims

1. Compounds of general formula (I)
Figure imgf000051_0001
wherein R1 is a C1-C16 straight or branched alkyl group, a phenyl group or a phenyl-C6-C16alkyl group; n is an integer from 3 to 18, inclusive;
R2 is a C1-C12 straight or branched alkyl group;
R3 is hydrogen, a carboxy, alkoxycarbonyl, arylalkoxycarbonyl or aryloxycarbonyl group of less than 12 carbon atoms in the last three cases;
R4, R5 and R6, which can be the same or different, are hydrogen, a C1-C6 alkyl group or N+R4R5R6 is a cyclic ammonium group, which can be aromatic or non-aromatic, wherein two of the R4, R5 or R6 groups form a ring together with the nitrogen atom, and the other group is hydrogen or C1-C6 alkyl,
and the salts, enantiomers and diastereomers thereof.
2. Compounds as claimed in claim 1, wherein R2 is a C1- C3 alkyl group.
3. Compounds as claimed in claim 1, wherein R4, R5 and
R6 are hydrogen, methyl or ethyl.
4. Compounds as claimed in claim 1, wherein, when R3 is hydrogen, N+R4R5R6 is a cyclic aromatic ammonium group as claimed in claim 1.
5. Compounds as claimed in claim 4, wherein the cyclic aromatic group is 1-pyridinium, 1-imidazolium or 3- thiazolium.
6. Compounds as claimed in claim 1, wherein, when R3 is hydrogen, N+R4R5R6 is a cyclic non-aromatic ammonium group as claimed in claim 1.
7. Compounds as claimed in claim 6, wherein the cyclic ammonium group is 1-pyrrolidinium, 1-piperidinium or 4- morpholinium.
8. Compounds as claimed in claim 1, wherein R3 is a carboxy, aryloxycarbonyl, alkoxycarbonyl or arylakoxycarbonyl group, the latter two groups having less than 12 carbon atoms.
9. Compounds as claimed in any one of claims 1-8, selected from:
1-O-(4-oxooctadecyl)-2-O-methylglycero-3-phosphocholine 1-O-(4-oxooctadecyl)-2-O-methylglycero-3-phosphoserine
1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phosphocholine 1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phosphoserine 1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phosphoserine methyl ester
1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phospho(N,N,N-trimethyl)serine
2-O-methyl-3-O-(7-oxooctadecyl)-sn-glycero-1-phospho-choline
2-O-methyl-1-O-(7-oxooctadecyl)-sn-glycero-3-phospho-choline
1-O-(7-oxooctadecyl)-2-O-ethylglycero-3-phosphocholine
1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phospho[2-(3-thiazolium)ethyl]
1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phospho[2-(1- imidazolium)ethyl]
1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phospho[2-(N-methylpiperidinium)ethyl]
1-O-(12-oxooctadecyl)-2-O-methylglycero-3-phospho-choline
1-O-(12-oxooctadecyl)-2-O-methylglycero-3-phosphoserine 1-O-(11-phenyl-11-oxoundecyl)-2-O-methylglycero-3-phosphocholine
1-O-(14-phenyl-11-oxotetradecyl)-2-O-methylglycero-3-phosphocholine
1-O-(14-phenyl-11-oxotetradecyl)-2-O-methylglycero-3-phosphoserine.
10. As a compound as claimed in claim 9,
1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phosphocholine in form of the R(+) enantiomer.
11. As a compound as claimed in claim 9,
1-O-(7-oxooctadecyl)-2-O-methylglycero-3-phosphocholine in form of the S(-) enantiomer.
12. A process for the preparation of the compounds claimed in claim 1, in which process:
a compound of formula (XI)
Figure imgf000053_0001
wherein Z is oxygen and n, R1 and R2 have the above mentioned meanings, is reacted with a compound of formula (XII)
Figure imgf000054_0001
wherein X and Y, which can be the same or different, are halogens, in the presence of an amine, in an inert organic solvent, at a temperature from 0° to 40 °C, to give a compound of formula (XIII)
Figure imgf000054_0002
wherein n , R1, R2, X and Y have the above mentioned meanings, which is subjected to aqueous hydrolysis in a saline solution, to give the compound of formula (XIV)
Figure imgf000054_0003
wherein R1 , R2 and Y have thhee aatbove mentioned meanings, which compound (XIV) is reacted with a compound of formula (XV)
NR4R5R6
XV
wherein R4, R5 and R6 have the above mentioned meanings, in a suitable organic solvent, at a temperature from 40ºC to the reflux one, to give a compound of formula I, wherein n, R1, R2 , R4 , R5 and R6 have the above mentioned meanings and R3 is hydrogen.
13. A process for the preparation of the compounds claimed in claim 1, in which process:
compound (XI), wherein X is oxygen and n, R1 and R2 have the above mentioned meanings, is reacted with a compound of formula (XVI)
Figure imgf000055_0001
wherein X is halogen (such as Cl, Br, I) in a suitable organic solvent, in the presence of an amine, at a temperature from 0º to 40°C, to give a compound of formula (XVII)
Figure imgf000056_0001
wherein R1 and R2 have the above mentioneα meanings, which compound (XVII) is treated with a compound of formula (XV), in suitable organic solvents, at a temperature from 40 °C to the reflux one, to give a compound of formula (I) wherein n, R1, R2, R4, R5 and
R6 have the above mentioned meanings and R3 is hydrogen.
14. A process for the preparation of the compounds claimed in claim 1, in which process:
a compound of formula (XI), wherein Z is oxygen or a suitable keto-protecting group, and n, R1 and R2 have the above mentioned meanings, is reacted with PCI, in the presence of a slightly nucleophilic base, such as imidazol, and an acid-binding agent, in suitable organic solvents, at a temperature from 0° to 40 °C, to give a compound of formula (XVIII)
Figure imgf000056_0002
wherein R1 and R2 have the above mentioned meanings and A can be hydrogen or a triethylammonium group, which compound (XVIII) is reacted with a compound of formula (XIX)
Figure imgf000057_0002
wherein R7 is an amino-protecting group, such as benzyloxycarbonyl or t-butyloxycarbonyl, and R3 can be an alkoxycarbonyl group having less than 12 carbon atoms, or it can be a carboxy group protected in form of the benzyl or t-butyl esters, in the presence of a base such as pyridine and of a condensing agent, such as pivaloyl chloride or 5,5-dimethyl-2-oxo-2-chloro-1,3,2-dioxaphospholane, at a temperature from 0º to 40ºC, to give a compound of formula (XX)
Figure imgf000057_0001
wherein n, Z, R1, R2, R3 and R7 have the above mentioned meanings, which is oxidized with an oxidizing agent, such as iodine, to give a compound of formula
Figure imgf000058_0001
wherein n, Z, R1, R2, R3 and R7 have the above mentioned meanings, which compound is converted into a compound of formula (I) by removing the protecting groups R7 and Y, if necessary, when R3 is a carboxy group protected in form of a benzyl or t-butyl ester.
15. A process as claimed in claims 12-14, in which the amine or acid-binding agent is triethylamine or pyridine .
16. A process as claimed in claim 14, wherein the protecting groups are removed by catalytic hydrogenation in an acid medium.
17. Pharmaceutical compositions containing as the active principle a compound as claimed in claims 1-11 in admixture with a suitable carrier.
18. The use of the compounds as claimed in claims 1-11 for the preparation of a medicament having antitumor and anti-platelet aggregating activities.
PCT/EP1992/001502 1991-07-10 1992-07-03 Ketoalkylglycerophospholipids having antitumor and anti-platelet aggregation activities, processes for the preparation thereof and pharmaceutical compositions therefrom WO1993001196A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
ES9101611A ES2034885B1 (en) 1991-07-10 1991-07-10 PROCEDURE FOR THE PREPARATION OF CETOALQUILGLICEROFOSFOLIPIDOS.
ES9101611 1991-07-10

Publications (1)

Publication Number Publication Date
WO1993001196A1 true WO1993001196A1 (en) 1993-01-21

Family

ID=8272953

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP1992/001502 WO1993001196A1 (en) 1991-07-10 1992-07-03 Ketoalkylglycerophospholipids having antitumor and anti-platelet aggregation activities, processes for the preparation thereof and pharmaceutical compositions therefrom

Country Status (3)

Country Link
AU (1) AU2229292A (en)
ES (1) ES2034885B1 (en)
WO (1) WO1993001196A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1583661A (en) * 1976-05-04 1981-01-28 Max Planck Gesellschaft Tumour antidote
EP0138559A2 (en) * 1983-10-11 1985-04-24 Takeda Chemical Industries, Ltd. Ketoalkylphospholipids and their production and use
WO1987000173A1 (en) * 1985-07-03 1987-01-15 Chemie Linz Aktiengesellschaft Derivatives of glycero-3(2)-phospho-l-serine and pharmaceutical preparations containing them
EP0225129A1 (en) * 1985-11-29 1987-06-10 Takeda Chemical Industries, Ltd. Phospholipid derivatives, their production and use

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2019552A6 (en) * 1990-04-11 1991-06-16 Menarini Lab Process for the preparation of glycerophospholipids

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1583661A (en) * 1976-05-04 1981-01-28 Max Planck Gesellschaft Tumour antidote
EP0138559A2 (en) * 1983-10-11 1985-04-24 Takeda Chemical Industries, Ltd. Ketoalkylphospholipids and their production and use
WO1987000173A1 (en) * 1985-07-03 1987-01-15 Chemie Linz Aktiengesellschaft Derivatives of glycero-3(2)-phospho-l-serine and pharmaceutical preparations containing them
EP0225129A1 (en) * 1985-11-29 1987-06-10 Takeda Chemical Industries, Ltd. Phospholipid derivatives, their production and use

Also Published As

Publication number Publication date
ES2034885B1 (en) 1994-03-01
ES2034885A1 (en) 1993-04-01
AU2229292A (en) 1993-02-11

Similar Documents

Publication Publication Date Title
Lindh et al. A general method for the synthesis of glycerophospholipids and their analogs via H-phosphonate intermediates
IL90166A (en) Antiretroviral pharmaceutical compositions comprising 9-(2,4-disubstituted butyl)-9H purine derivatives some such novel compounds and their preparation
JPS58189191A (en) Phospholipid derivative, its preparation, and antitumor agent containing it
CH664158A5 (en) DERIVATIVES PROPYLIDENEDIPHOSPHONATES-1,3 SUBSTITUTED IN POSITION 2, THEIR PREPARATION METHOD AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
WO1992019610A1 (en) 2- and 5-alkyl and phenyl substituted 4-(1-hydroxy, 1-acyloxy or 1-carbamoyloxy)-5-hydroxy-2(5h)-furanones as anti-inflammatory agents
JPH069668A (en) Aryl and heteroaryl (phosphinylmethyl)phosphonate squalene synthetase inhibitors
Kawashima et al. Optical resolution studies of cyclophosphamide
WO1993001196A1 (en) Ketoalkylglycerophospholipids having antitumor and anti-platelet aggregation activities, processes for the preparation thereof and pharmaceutical compositions therefrom
KR100541590B1 (en) Sulfurpentafluoride compounds and methods for making and using same
Ponpipom et al. Synthesis of azide and amide analogs of platelet-activating factor and related derivatives
CA2180754C (en) Heteroaryl-substituted deoxy glycero-phosphoethanolamines
EP0257762B1 (en) Novel glycerol derivative and anti-hypertensive agent
CA1234834A (en) Phospholipids
WO2004041167A2 (en) Antiprotozoal ring-substituted phospholipids
FR2571374A1 (en) NOVEL 5-FLUORO-2'-DESOXYURIDINE DERIVATIVES AND THEIR SALTS, PREPARATION METHOD THEREOF, AND ANTI-TUMOR AGENTS CONTAINING SAME
EP0494283B1 (en) 1,4-benzodioxane derivatives, process for the preparation thereof and pharmaceutical compositions therefrom
Garcia et al. Synthesis of new ether glycerophospholipids structurally related to modulator
PL184728B1 (en) Novel intermediate compounds as their application in production of bisindoliloamaleimides with a n,n'-bridge
US4659859A (en) 2-alkoxy-1-alkoxy phosphoryl dichlorides
FR2656307A1 (en) (N-METHYL-N-ALCOHYL) AMINO-3-METHOXYMETHYLENE-2-PROPANE-1-OL DERIVATIVES AND METHOD FOR THE PREPARATION THEREOF
KR20020094000A (en) Bis-(n,n'-bis-(2-haloethyl)amino)phosphoramidates as antitumor agents
TW399055B (en) 1, 9-diazabicyclo [4.3.0] nona-3, 8-diene derivatives having cardioprotective activity
EP1615937A2 (en) Analogs of lysophosphatidic acid and methods of making and using thereof
Fos et al. Synthesis of alkyl chain-modified ether lipids and evaluation of their in vitro cytotoxicity
EP0607374B1 (en) Novel cyclic lipid derivatives as potent paf antagonists

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU BB BG BR CA CS FI HU JP KP KR LK MG MN MW NO PL RO RU SD US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IT LU MC NL SE BF BJ CF CG CI CM GA GN ML MR SN TD TG

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA