WO2005085243A2 - Process for the preparation of cabergoline - Google Patents

Process for the preparation of cabergoline Download PDF

Info

Publication number
WO2005085243A2
WO2005085243A2 PCT/HU2005/000022 HU2005000022W WO2005085243A2 WO 2005085243 A2 WO2005085243 A2 WO 2005085243A2 HU 2005000022 W HU2005000022 W HU 2005000022W WO 2005085243 A2 WO2005085243 A2 WO 2005085243A2
Authority
WO
WIPO (PCT)
Prior art keywords
formula
cabergoline
compound
catalyst
solvent
Prior art date
Application number
PCT/HU2005/000022
Other languages
French (fr)
Other versions
WO2005085243A3 (en
Inventor
János Galambos
László Czibula
Ferenc SEBÕK
Sándorné BÁLINT
Ferencné KASSAI
Györgyi IGNÁCZNÉ SZENDREI
Ádám DEMETER
Original Assignee
Richter Gedeon Vegyészeti Gyár Rt.
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 Richter Gedeon Vegyészeti Gyár Rt. filed Critical Richter Gedeon Vegyészeti Gyár Rt.
Priority to US10/591,202 priority Critical patent/US7858791B2/en
Priority to DE602005021461T priority patent/DE602005021461D1/en
Priority to AT05718154T priority patent/ATE469149T1/en
Priority to EP05718154A priority patent/EP1720869B1/en
Priority to EA200601628A priority patent/EA010689B1/en
Publication of WO2005085243A2 publication Critical patent/WO2005085243A2/en
Publication of WO2005085243A3 publication Critical patent/WO2005085243A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D457/00Heterocyclic compounds containing indolo [4, 3-f, g] quinoline ring systems, e.g. derivatives of ergoline, of the formula:, e.g. lysergic acid
    • C07D457/04Heterocyclic compounds containing indolo [4, 3-f, g] quinoline ring systems, e.g. derivatives of ergoline, of the formula:, e.g. lysergic acid with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 8
    • C07D457/06Lysergic acid amides

Definitions

  • This invention relates to a new process for the preparation of cabergoline of formula
  • Cabergoline (I) was firstly prepared according to United States Patent 4,526,892 by reaction of 6-allyl-ergoline-8 ⁇ -carboxylic acid (II) with l-[3-(dimethylamino)propyl)-3- ethylcarbodiimide (EDC) (Scheme 1). Scheme 1
  • J. Org. Chem. 2002, 67, 7147-7150 describes an ethyl isocyanate- free method for the production of cabergoline (I) that solves the problem of completing acylation of indole nitrogen, too.
  • the first step is the protection of indole nitrogen of amide (IN) preferably as tert-butyl carbamate (Nil).
  • Extension of the amide side chain is done by deprotonation of compound (Nil) with sodium hexamethyldisilazide ( ⁇ aHMDS) followed by trapping the anion with phenyl chloroformate (PhOCOCl) to yield the phenyl carbamate (VIII).
  • Reaction of compound (VIII) with ethylamine hydrochloride (Et ⁇ H 2 xHCl) gives
  • Al Patent Application cabergoline (I) may be prepared by silylating amide (TV) with a silylating agent (e.g. trimethylsilyl trifluoromethane sulfonate - TMSOTf), reacting the obtained product (XI) with ethyl isocyanate (EtNCO) followed by desilylation of intermediate (XII) (Scheme 4).
  • a silylating agent e.g. trimethylsilyl trifluoromethane sulfonate - TMSOTf
  • WO 01/70740 Al Patent Application describes a new process for the preparation of crystalline form I from the new crystalline form V.
  • the form V - which is toluene solvate - is prepared from the mixture of the purified cabergoline (I) with toluene and diethyl ether by a long-lasting complicated process, at low reaction temperature, and the yield is only 45%.
  • the crystalline form I is prepared by drying the form V in vacuum.
  • WO 01/72746 Al Patent Application describes the preparation of crystalline form VII from the crystalline form I.
  • the suspension of form I in n-heptane or 1,4-dioxane is stirred for 48 hours, and then the suspension was filtered to obtain the crystalline form VII.
  • the yield is 45.2%.
  • WO 01/72747 Al Patent Application describes the crystalline form II and a process for its preparation with approx. 70% yield by stirring the cabergoline (I) for several days in an organic solvent (eg. diethyl ether) at low temperature.
  • organic solvent eg. diethyl ether
  • cabergoline (I) with a method via new intermediates proposed by the present invention is commercially more advantageous than with the previously disclosed known methods due to the high yield (approx.78%), the milder reaction conditions and the shorter reaction time.
  • Another advantage of the use of these intermediates is that they have high hydrophobicity - in comparison to known intermediates bearing two basic function - so their purification by normal phase chromatography (if necessary) is highly effective due to the expanded difference in retention between them and the by-products.
  • the present invention relates to a process for preparing cabergoline (I) from ergoline- 8 ⁇ -carboxylic acid C ⁇ alkyl esters via new intermediates and the polymorphic amorphous form of cabergoline (I).
  • the process comprises protecting the secondary amine and the indole nitrogen functions of ergoline-8 ⁇ -carboxylic acid . 4 alkyl esters as carbamate derivatives, amidating the obtained protected compound with 3-(dimethylamino)propylamine, reacting the amide with ethyl isocyanate, cleaving the protecting groups and reacting the obtained deprotected secondary amine with an electrophyl allyl alcohol derivative to obtain cabergoline (I).
  • the present invention also relates to the new intermediates used in this process.
  • the invention also relates to the new amorphous form of cabergoline (I) and the preparation thereof. DETAILED DESCRIPTION OF THE INVENTION
  • the invention relates to a process for preparing cabergoline (I) comprising the following steps: (a) Reacting a compound of formula (XIII) - wherein Ri represents a C 1 - 4 alkyl group - in the presence of a catalyst (i) with a compound of formula (XTV), X-COOR 2 (XTV)
  • R 2 represents an optionally substituted straight or branched Ci. 6 alkyl group
  • X represents a bromine or chlorine atom, or (ii) with a compound of formula (XV), O(COOR 2 ) 2 (XV) wherein R 2 is a group as defined above for formula (XIN);
  • Step (a) of the process refers to reacting an ergoline-8 ⁇ -carboxylic acid ester of formula (XIII) with a compound of formula (XIN) or (XV).
  • the starting materials, ergoline-8 ⁇ -carboxylic acid esters of formula (XIII) can be prepared according to any method known in the state of the art: e.g. US 4,166,182 Patent or Collect. Czech. Chem. Commun. 1983, 48(5), 1483-1489. Methods that serve for protection of both secondary amines and indole nitrogen are well known from the art. Thus, these functionalities can be protected e.g. as N-alkyl, N-aryl,
  • the reaction may be carried out from -50°C to the reflux temperature of the reaction mixture in a suitable aprotic solvent in the presence of an organic or inorganic base as catalyst.
  • the reaction step (a) is carried out at a temperature of from 0°C to 50°C in the presence of 4-dimethylaminopyridine catalyst in a hydrocarbon halide solvent.
  • is methyl and R 2 is tert-butyl.
  • Step (b) of the process refers to reacting the carbamate derivative of formula (XVI) obtained in the step (a) with 3-(dimemylamino)propylamine.
  • the amidation of ergoline-8 ⁇ -carboxylic acid methyl ester derivatives containing a basic nitrogen in position 6 of the ergoline skeleton with 3-(dimethylamino)propylamine can be accomplished by know methods.
  • the reactions can be carried out with a large excess of the amine (i) refluxing the reaction mixture for 10-12 hours without solvent (approx. at 135°C) in the presence of acetic acid catalyst; (ii) heating the reactants for 18 hours at 100°C in ethylene glycol as a solvent with catalytic amount of 2-hydroxypyridine.
  • catalysts can be used provided they do not hurt the tert-butoxycarbonyl protecting groups.
  • examples of catalysts are organic and inorganic bases such as alkali metal or earth metal hydroxides or carbonates, alkali metal or earth metal alcoholates, pyridine or its derivatives, tertiary amines, etc.; salts such as ammonium chloride, copper(II) acetate, magnesium chloride; and other catalysts such as boron tribromide, dimethylaluminium amides, mixed tin(II) amides or mixtures thereof.
  • the amidation can be carried out without solvent or in the presence of a suitable solvent.
  • the step (b) is carried out at a temperature of from 50°C to 70°C in an Ci. 6 alkyl alcohol solvent in the presence of 2-hydroxypyridine catalyst.
  • the resultant amide of formula (XVII) may be used in the following step after isolation from the reaction mass following conventional procedures, or may be subjected to the subsequent step without isolation.
  • Step (c) of the process refers to reacting the ergoline-8 ⁇ -carboxamide derivative of formula (XVII) obtained in the step (b) with ethyl isocyanate (EtNCO).
  • the ethyl isocyanate may be used in a 1 to 4 fold molar amount, preferably 2 to 3 molar amount relative to the amount of the amide (XVII).
  • the reaction of amide (XVII) with ethyl isocyanate may be accelerated by metal catalysis in the presence of coordination compound(s).
  • Suitable metal catalysts include lb and lib metal group salts, preferably copper(I) and copper(II) salts.
  • Most preferred salts are copper(I) chloride, co ⁇ per(II) chloride, co ⁇ per(I) bromide and copper(I) iodide.
  • the ligands in the coordination compound(s) with lb and lib metals preferably contain phophorous, nitrogen and/or oxygen atoms.
  • Examples of the ligands include triarylphophines, tertiary amines, nitriles, amides and ether-type compounds.
  • Preferred ligands are triarylphophines, most preferred ligands are triphenylphosphine and tri-p-tolylphophine.
  • the reaction is carried out in the presence of a suitable aprotic organic solvent from 0°C to the reflux temperature of the reaction mixture.
  • a suitable aprotic organic solvent from 0°C to the reflux temperature of the reaction mixture.
  • the step (c) is carried out in hydrocarbon halide solvent, in the presence of cop ⁇ er(I) chloride and/or copper(II) chloride and/or copper(I) bromide and/or copper(l) iodide catalysts and triphenylphosphine or ligand at a temperature of from 30°C to 50°C.
  • the reaction product may be isolated and purified following conventional procedures.
  • the obtained protected N-acylurea derivatives of formula (XVIII) have high hydrophobicity (in comparison to known intermediates bearing two basic function), their purification by normal phase chromatography (if necessary) is highly effective due to the expanded difference in retention between it and the by-products.
  • Step (d) of the process refers to reacting the protected N-acylurea derivative of formula (XVIII) obtained in the step (c), with a strong aqueous inorganic acid to form N-[3- (dimethylamino)propyl]-N-[(ethylamino)carbonyl)-ergoline-8 ⁇ -carboxamide (XIX).
  • Methods cleaving the carbamate-type protecting groups from basic nitrogen and indole nitrogen are well known from the art, but not each is suitable for both purposes. Otherwise, with some commonly used methods (e.g. with formic acid or trichloroacetic acid) no significant conversion was observed, while other methods (e.g.
  • the strong acid means an acid which has a pK value less than 2 in water.
  • Examples for strong inorganic acid are hydrochloric acid, hydrobromic acid, sulfuric acid, hydrochloric acid. The deprotection may be carried out from 0 °C to the reflux temperature of the reaction mixture.
  • step (d) is carried out at a temperature of from 40°C to 80°C in aqueous hydrochloric acid.
  • the resultant compound of formula (XIX) may be used in the following step after isolation from the reaction mass following conventional procedures, or may be subjected to the subsequent step without isolation.
  • Step (e) of the process refers to reacting the obtained 6-deallyl-cabergoline of formula
  • XIX 6-deallyl-cabergoline
  • electrophyl allyl alcohol derivative in an organic solvent in the presence of palladium or nickel catalyst and ligand(s) yields Cabergoline (I) in high purity.
  • electrophyl allyl alcohol derivative are allylic carboxylates such as allyl acetate or allyl benzoate and allyl phenyl ether.
  • the catalytic system may be homogeneous or heterogeneous, preferably the catalytic system is homogeneous.
  • heterogeneous catalyst examples include palladium on activated carbon or polystyrene in the presence of phosphorous containing ligands, palladium ligated by phosphinated polystyrene or phosphinated silica.
  • homogeneous catalyst examples include tetrakis(triphenylphos ⁇ hine)palladium(0), tetraMs(triphenylphosphine)nickel, bis(cycloocta-l,5-diene)nickel, [l,4-bis(diphenyl- phosphino)butane]nickel, allyl palladium chloride dimer and cis,cis,cis- 1,2,3, 4-tetrakis- (diphenylphosphinometyl)cyclopentane.
  • the reaction may be carried out in a suitable aprotic organic solvent from 0 °C to the reflux temperature of the reaction mixture.
  • the electrophyl allyl alcohol derivative is allyl acetate
  • the catalyst is tetrakis(triphenylphosphine)palladium(0)
  • the reaction is carried out in an aromatic hydrocarbon solvents at a temperature of from 20°C to 50°C.
  • the reaction product may be isolated and purified following conventional procedures.
  • the cabergoline (I) can be converted into pharmaceutically acceptable salts.
  • the cabergoline (I) or pharmaceutically acceptable salts thereof, can be subsequently formulated with a pharmaceutically acceptable carrier or diluent to provide a pharmaceutical composition.
  • the used solvent is acetone, methyl acetate or dichloromethane.
  • the polymorphic amorphous form of Cabergoline (I) is very stable, the dissolution rate and absorption properties of the amorphous form of Cabergoline (I) are favourable. So the use of the amorphous form of
  • Cabergoline (I) in pharmaceutical compositions is advantageous in comparison to known crystalline forms.
  • N(6)-COOC(CH 3 ) 3 1.66 (s, 9 ⁇ , N(l)-COOC(CH 3 ) 3 ); 1.78-1.96 (m, 2 ⁇ ,
  • the reaction mixture was cooled to ambient temperature, 200 ml of dichloromethane was added and the pH was adjusted to 11 with concentrated aqueous ammonia solution.
  • the organic layer was separated and the aqueous layer was extracted with 2x60 ml of dichloromethane.
  • the combined organic layer was dried over anhydrous sodium sulphate. The dried solution was either subjected to the subsequent step without isolation of the product or it was concentrated in vacuum to give 8.1 g (94.8%) of the title compound.
  • Example 6a The same as in Example 6a, but employing methyl acetate as solvent, 9.85 g (98.5%) of the title compound was obtained.
  • XRD XRD
  • DSC IR analytical studies the crystalline form of the product is amorphous.
  • Figure 1 shows the XRD Spectra of amorphous form of Cabergoline (I).
  • Figure 2 shows the DSC Spectra of amorphous form of Cabergoline (I).
  • Figure 3 shows the IR Spectra of amorphous form of Cabergoline (I).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

A process for preparing cabergoline (I) from ergoline-8β-carboxylic acid ester (XIII) comprising the following steps. (XIII), (XVI), (XVII), (XVIII), (XIX), (I). The present case also relates to the intermediates (XVI), (XVII), (XVIII) and (XIX) as well as the polymorphic amorphous form of Cabergoline (I) and the production thereof.

Description

NOVELPROCESS FORPRODUCTION OF CABERGOLINE
FIELD OF THE INVENTION
This invention relates to a new process for the preparation of cabergoline of formula
(I), to novel intermediates used in this process, to the polymorphic amorphous form of cabergoline (I) and the preparation thereof.
BACKGROUND OF THE INVENTION
6- Allyl-N- [3 -(dimethylamino)propyl] -N- [(ethylamino)carbonyl] -ergoline-8 β- carboxamide - international non-proprietary name cabergoline - of formula (I)
Figure imgf000003_0001
cabergoline (I)
is a potent dopamine agonist and is useful as anti-Parkinson drug and as prolactin inhibitor (Eur. J. Med. Chem. 1989, 24, 421-426 and United States Patent 5,382,669). Cabergoline (I) was firstly prepared according to United States Patent 4,526,892 by reaction of 6-allyl-ergoline-8β-carboxylic acid (II) with l-[3-(dimethylamino)propyl)-3- ethylcarbodiimide (EDC) (Scheme 1). Scheme 1
Figure imgf000004_0001
(III) In this case both regioisomers (I) and (III) were obtained and the yield of the isolated cabergoline (I) is only approx. 21% as a consequence of isolation difficulties, considering that the yield of compound (II) prepared from (XIII) according to the state of the art is 70%. Eur. J. Med. Chem. 1989, 24, 421-426 describes another method for the preparation of Cabergoline (I), which is based on the direct reaction of 6-allyl-N-[3-(dimethylamino)propyl]- ergoline-8β-carboxamide (IN) with ethyl isocyanate (EtΝCO) (Scheme 2).
Scheme 2
EtΝCO
Figure imgf000005_0001
Figure imgf000005_0002
(IN) (I) Ri = H, R2 = EtΝHCO (cabergoline) (N) R1 = EtΝHCO, R2 = H (VI) Ri = EtNHCO, R2 = EtNHCO
Since this reaction leads to equilibrium, it requires the use of a large excess of ethyl isocyanate (up to 40 equivalents) for reasonable conversion and must be conducted at above 100°C in toluene for several hours. The use of large quantities of toxic ethyl isocyanate under drastic reaction conditions presents a serious hazard for the large-scale preparation of cabergoline (I). In addition, conversion to (I) is incomplete and competitive acylation of the indole nitrogen forming compounds (V) and (VI) occurs. This side reaction complicates the product purification and reduces the yield, which is only approx. 58%, considering that the yield of compound (IN) prepared from (XIII) according to the state of the art is 72%.
The method proposed in United States Patent 5,382,669 and Syn. Lett. 1995, 605-606 showed that catalysis by copper salts in the presence of phosphine ligands permitted the ethyl isocyanate reaction to be run at room temperature with only 3 equivalents of ethyl isocyanate. However, despite of moderation in reaction conditions the conversion and the ratio of cabergoline (I) and the byproducts (V and VI) are not much different from the uncatalyzed thermal reaction. The yield is only approx. 48% and 57%, considering that the yield of compound (TV) prepared from (XIII) according to the state of the art is 72%.
J. Org. Chem. 2002, 67, 7147-7150 describes an ethyl isocyanate- free method for the production of cabergoline (I) that solves the problem of completing acylation of indole nitrogen, too. The first step is the protection of indole nitrogen of amide (IN) preferably as tert-butyl carbamate (Nil). Extension of the amide side chain is done by deprotonation of compound (Nil) with sodium hexamethyldisilazide (ΝaHMDS) followed by trapping the anion with phenyl chloroformate (PhOCOCl) to yield the phenyl carbamate (VIII). Reaction of compound (VIII) with ethylamine hydrochloride (EtΝH2xHCl) gives
BOC-cabergoline (IX) but also generates the ethylamide (X). The deprotection is done from the mixture of (DC) and (X) with IN aqueous hydrochloric acid. The purified cabergoline (I) is then isolated by basification followed by chromatography on silica. (Scheme 3). Scheme 3
Figure imgf000007_0001
(IX) (X)
In this approach the deprotonating step requires special cold reactor and strictly anhydrous circumstances. These requirements can hardly be satisfied in the course of large- scale preparation and the yield is only approx. 52%, considering that the yield of compound (VII) prepared from (XIII) according to the state of the art is 66%.
According to US 2002/0177709 Al Patent Application cabergoline (I) may be prepared by silylating amide (TV) with a silylating agent (e.g. trimethylsilyl trifluoromethane sulfonate - TMSOTf), reacting the obtained product (XI) with ethyl isocyanate (EtNCO) followed by desilylation of intermediate (XII) (Scheme 4). Scheme 4
Figure imgf000008_0001
(IV) (XI)
desilylation (I) cabergoline
Figure imgf000008_0002
(xπ)
The disadvantage of this process is, that the silylating step requires strictly anhydrous circumstances. Otherwise, the reaction with ethyl isocyanate runs too long (24 hours) raising the safety hazard in the course of large-scale preparation and the yield is approx. 65%, considering that the yield of compound (IV) prepared from (XIII) according to the state of the art is 72%. Several crystalline forms of Cabergoline (I) are known.
IL Farmaco 1995, 50 (3), 175-178 describes the preparation of crystalline form I. This solvated anhydrate product is crystallized from diethyl ether.
WO 01/70740 Al Patent Application describes a new process for the preparation of crystalline form I from the new crystalline form V. The form V - which is toluene solvate - is prepared from the mixture of the purified cabergoline (I) with toluene and diethyl ether by a long-lasting complicated process, at low reaction temperature, and the yield is only 45%. The crystalline form I is prepared by drying the form V in vacuum.
WO 01/72746 Al Patent Application describes the preparation of crystalline form VII from the crystalline form I. By this process the suspension of form I in n-heptane or 1,4-dioxane is stirred for 48 hours, and then the suspension was filtered to obtain the crystalline form VII. The yield is 45.2%.
WO 01/72747 Al Patent Application describes the crystalline form II and a process for its preparation with approx. 70% yield by stirring the cabergoline (I) for several days in an organic solvent (eg. diethyl ether) at low temperature.
SUMMARY OF THE INVENTION
Surprisingly it was found that the preparation of cabergoline (I) with a method via new intermediates proposed by the present invention is commercially more advantageous than with the previously disclosed known methods due to the high yield (approx.78%), the milder reaction conditions and the shorter reaction time. Another advantage of the use of these intermediates is that they have high hydrophobicity - in comparison to known intermediates bearing two basic function - so their purification by normal phase chromatography (if necessary) is highly effective due to the expanded difference in retention between them and the by-products.
The present invention relates to a process for preparing cabergoline (I) from ergoline- 8β-carboxylic acid Cμ alkyl esters via new intermediates and the polymorphic amorphous form of cabergoline (I).
The process comprises protecting the secondary amine and the indole nitrogen functions of ergoline-8β-carboxylic acid .4 alkyl esters as carbamate derivatives, amidating the obtained protected compound with 3-(dimethylamino)propylamine, reacting the amide with ethyl isocyanate, cleaving the protecting groups and reacting the obtained deprotected secondary amine with an electrophyl allyl alcohol derivative to obtain cabergoline (I).
The present invention also relates to the new intermediates used in this process.
The invention also relates to the new amorphous form of cabergoline (I) and the preparation thereof. DETAILED DESCRIPTION OF THE INVENTION
The invention relates to a process for preparing cabergoline (I) comprising the following steps: (a) Reacting a compound of formula (XIII) - wherein Ri represents a C1-4 alkyl group - in the presence of a catalyst (i) with a compound of formula (XTV), X-COOR2 (XTV)
wherein R2 represents an optionally substituted straight or branched Ci.6 alkyl group, X represents a bromine or chlorine atom, or (ii) with a compound of formula (XV), O(COOR2)2 (XV) wherein R2 is a group as defined above for formula (XIN);
(b) reacting the obtained carbamate derivative of formula (XVI) - wherein Ri and R2 is a group as defined above - with 3-(dimethylamino)propylamine (DMAPA) in the presence of a catalyst;
(c) reacting the obtained ergoline-8β-carboxamide derivative of formula (XVII) - wherein R2 is a group as defined above - with ethyl isocyanate (EtΝCO) in the presence of ligand(s) and lb and lib metal group salt as catalyst, (d) reacting the obtained protected N-acylurea derivative of formula (XVIII) - wherein R2 is a group as defined above - with a strong aqueous inorganic acid (aq./acid);
(e) reacting the obtained secondary amine of formula (XLX) with an elecfrophyl allyl alcohol derivative in the presence of a palladium or nickel containing catalyst and optionally in the presence of ligand(s) to form cabergoline (I).
The reaction procedure is shown in Scheme 5.
Figure imgf000013_0001
(xvπ) (xvm)
Figure imgf000013_0002
(XLX) (I)
Step (a) of the process refers to reacting an ergoline-8β-carboxylic acid ester of formula (XIII) with a compound of formula (XIN) or (XV). The starting materials, ergoline-8β-carboxylic acid esters of formula (XIII) can be prepared according to any method known in the state of the art: e.g. US 4,166,182 Patent or Collect. Czech. Chem. Commun. 1983, 48(5), 1483-1489. Methods that serve for protection of both secondary amines and indole nitrogen are well known from the art. Thus, these functionalities can be protected e.g. as N-alkyl, N-aryl,
N-silyl, N-sulfonyl derivatives or as carbamates. A number of protecting groups were examined, and it was found to be best to protect the starting material of formula (XIII) as the carbamate of formula (XVI). Compounds of formula (XIV) and (XV) are commercially available or may be prepared by well known procedures or by procedures analogous to those described in the literature. Compounds of formula (XIV) and (XV) may be used in a 2 to 10 fold molar amount, preferably from 2 to 5 fold molar amount, relative to the amount of ergoline-8β-carboxylic acid ester of formula (XIII). The reaction may be carried out from -50°C to the reflux temperature of the reaction mixture in a suitable aprotic solvent in the presence of an organic or inorganic base as catalyst. Preferably the reaction step (a) is carried out at a temperature of from 0°C to 50°C in the presence of 4-dimethylaminopyridine catalyst in a hydrocarbon halide solvent. Preferably \ is methyl and R2 is tert-butyl.
Step (b) of the process refers to reacting the carbamate derivative of formula (XVI) obtained in the step (a) with 3-(dimemylamino)propylamine. The amidation of ergoline-8β-carboxylic acid methyl ester derivatives containing a basic nitrogen in position 6 of the ergoline skeleton with 3-(dimethylamino)propylamine can be accomplished by know methods. The reactions can be carried out with a large excess of the amine (i) refluxing the reaction mixture for 10-12 hours without solvent (approx. at 135°C) in the presence of acetic acid catalyst; (ii) heating the reactants for 18 hours at 100°C in ethylene glycol as a solvent with catalytic amount of 2-hydroxypyridine. In both cases the yields are about 85% due to the decomposition reactions caused by the elevated temperature. It has been found that the lack of the basic nitrogen in the D ring of the ergoline skeleton has a significant effect on the reaction time and temperature required in the amidation reaction. So the reaction of carbamate derivatives of formula (XVI) with 3-(dimethylamino) propylamine carried out by known methods can be completed below 70°C in a few hours. Due to the reduced temperature the obtained ergoline-8β-carboxamide derivative of formula (XVII) has high purity and the yield is over 95%. The amidation may be accomplished at a temperature of from 40°C to 70°C. Catalysts accelerate the amidation reaction. All known catalysts can be used provided they do not hurt the tert-butoxycarbonyl protecting groups. Examples of catalysts are organic and inorganic bases such as alkali metal or earth metal hydroxides or carbonates, alkali metal or earth metal alcoholates, pyridine or its derivatives, tertiary amines, etc.; salts such as ammonium chloride, copper(II) acetate, magnesium chloride; and other catalysts such as boron tribromide, dimethylaluminium amides, mixed tin(II) amides or mixtures thereof. The amidation can be carried out without solvent or in the presence of a suitable solvent. Preferably the step (b) is carried out at a temperature of from 50°C to 70°C in an Ci.6 alkyl alcohol solvent in the presence of 2-hydroxypyridine catalyst. The resultant amide of formula (XVII) may be used in the following step after isolation from the reaction mass following conventional procedures, or may be subjected to the subsequent step without isolation.
Step (c) of the process refers to reacting the ergoline-8β-carboxamide derivative of formula (XVII) obtained in the step (b) with ethyl isocyanate (EtNCO). The ethyl isocyanate may be used in a 1 to 4 fold molar amount, preferably 2 to 3 molar amount relative to the amount of the amide (XVII). Optionally, the reaction of amide (XVII) with ethyl isocyanate may be accelerated by metal catalysis in the presence of coordination compound(s). Suitable metal catalysts include lb and lib metal group salts, preferably copper(I) and copper(II) salts. Most preferred salts are copper(I) chloride, coρper(II) chloride, coρper(I) bromide and copper(I) iodide. The ligands in the coordination compound(s) with lb and lib metals preferably contain phophorous, nitrogen and/or oxygen atoms. Examples of the ligands include triarylphophines, tertiary amines, nitriles, amides and ether-type compounds. Preferred ligands are triarylphophines, most preferred ligands are triphenylphosphine and tri-p-tolylphophine. The reaction is carried out in the presence of a suitable aprotic organic solvent from 0°C to the reflux temperature of the reaction mixture. Preferably the step (c) is carried out in hydrocarbon halide solvent, in the presence of copρer(I) chloride and/or copper(II) chloride and/or copper(I) bromide and/or copper(l) iodide catalysts and triphenylphosphine or
Figure imgf000017_0001
ligand at a temperature of from 30°C to 50°C.
The reaction product may be isolated and purified following conventional procedures. As the obtained protected N-acylurea derivatives of formula (XVIII) have high hydrophobicity (in comparison to known intermediates bearing two basic function), their purification by normal phase chromatography (if necessary) is highly effective due to the expanded difference in retention between it and the by-products.
Step (d) of the process refers to reacting the protected N-acylurea derivative of formula (XVIII) obtained in the step (c), with a strong aqueous inorganic acid to form N-[3- (dimethylamino)propyl]-N-[(ethylamino)carbonyl)-ergoline-8β-carboxamide (XIX). Methods cleaving the carbamate-type protecting groups from basic nitrogen and indole nitrogen are well known from the art, but not each is suitable for both purposes. Otherwise, with some commonly used methods (e.g. with formic acid or trichloroacetic acid) no significant conversion was observed, while other methods (e.g. with trifluoroacetic acid or trifluoromethanesulfonic acid) were not tolerated by the substrate (XVIII) and/or by the product (XIX). It has been found that carbamate-type protecting groups of compounds (XVIII), can be readily removed in a clean reaction with strong aqueous inorganic acids. In this term the strong acid means an acid which has a pK value less than 2 in water. Examples for strong inorganic acid are hydrochloric acid, hydrobromic acid, sulfuric acid, hydrochloric acid. The deprotection may be carried out from 0 °C to the reflux temperature of the reaction mixture. Because of the basic nitrogen functionality, both substrate (XVIII) and product (XIX) are fully soluble in aqueous acidic medium so no organic solvent is required to accomplish the deprotection reaction. Preferably the step (d) is carried out at a temperature of from 40°C to 80°C in aqueous hydrochloric acid. The resultant compound of formula (XIX) may be used in the following step after isolation from the reaction mass following conventional procedures, or may be subjected to the subsequent step without isolation.
Step (e) of the process refers to reacting the obtained 6-deallyl-cabergoline of formula
(XIX) with an electrophyl allyl alcohol derivative to form Cabergoline (I). Due to the formation of considerable amount of quaternary ammonium derivatives the allylation of the secondary amine functionality of (XIX) can not be accomplished by commonly used allylating agents (e.g. allyl halides, allyl arylsulfonates, allyl alkylsulfonates). However, no quaternary by-products may be observed when the product (I) is produced by nucleophilic allylic substitution reaction. So the reaction of 6-deallyl-cabergoline (XIX) with an electrophyl allyl alcohol derivative in an organic solvent in the presence of palladium or nickel catalyst and ligand(s) yields Cabergoline (I) in high purity. Examples of electrophyl allyl alcohol derivative are allylic carboxylates such as allyl acetate or allyl benzoate and allyl phenyl ether. The catalytic system may be homogeneous or heterogeneous, preferably the catalytic system is homogeneous. Examples of heterogeneous catalyst are palladium on activated carbon or polystyrene in the presence of phosphorous containing ligands, palladium ligated by phosphinated polystyrene or phosphinated silica. Examples for homogeneous catalyst are tetrakis(triphenylphosρhine)palladium(0), tetraMs(triphenylphosphine)nickel, bis(cycloocta-l,5-diene)nickel, [l,4-bis(diphenyl- phosphino)butane]nickel, allyl palladium chloride dimer and cis,cis,cis- 1,2,3, 4-tetrakis- (diphenylphosphinometyl)cyclopentane. The reaction may be carried out in a suitable aprotic organic solvent from 0 °C to the reflux temperature of the reaction mixture. Preferably at step (e) the electrophyl allyl alcohol derivative is allyl acetate, the catalyst is tetrakis(triphenylphosphine)palladium(0), and the reaction is carried out in an aromatic hydrocarbon solvents at a temperature of from 20°C to 50°C.
The reaction product may be isolated and purified following conventional procedures. The cabergoline (I) can be converted into pharmaceutically acceptable salts. The cabergoline (I) or pharmaceutically acceptable salts thereof, can be subsequently formulated with a pharmaceutically acceptable carrier or diluent to provide a pharmaceutical composition.
The intermediate compounds of formula (XVI), (XVII), (XVIII) and (XLX) are new.
Surprisingly it was found that if the chromatographically purified oily Cabergoline (I) is dissolved in a suitable organic solvent and from the obtained solution the solvent is partially removed several times in vacuum at a temperature of from 0°C to 30°C until the residue is not oily but solid product, a new polymorphic amorphous form of Cabergoline (I) is obtained which is identified by XRD, DSC and IR analytical methods. The chemical purity of the polymorphic amorphous form of Cabergoline (I) obtained by the above method is >99.5% (HPLC). The advantage of the process for production of amorphous Cabergoline (I) is the short technological time and the high yield which is practically quantitative. Preferably the used solvent is acetone, methyl acetate or dichloromethane. According to our analytical and pharmacological studies the polymorphic amorphous form of Cabergoline (I) is very stable, the dissolution rate and absorption properties of the amorphous form of Cabergoline (I) are favourable. So the use of the amorphous form of
Cabergoline (I) in pharmaceutical compositions is advantageous in comparison to known crystalline forms.
The invention is illustrated further by the following non-limiting examples:
EXAMPLE 1 Synthesis of l,6-di(før?-butoxycarbonyl)-ergoline-8β-carboxyIic acid methyl ester (XVI, Ri = methyl, R2 = tert-butyϊ) To a solution of 13.05 g (48.2 mmol) of ergoline-8β-carboxylic acid methyl ester (XIII, Ri = methyl) in 400 ml of dichloromethane 20 ml of triethylamine, 1.0 g of 4- dimethylaminopyridine, 42.1 g (193.1 mmol) of di-tert-butyl dicarbonate was sequentially added, and the reaction mixture was stirred at 40°C for 5 hours. The mixture was cooled to ambient temperature and it was washed with 3x100 ml of sodium chloride solution. The organic layer was dried over anhydrous sodium sulphate, and concentrated in vacuum. Crystallization from hexane gave 21.6 g (95.2%) of the title compound. 1H NMR (CDCI3, TMS, 500MHz) δ 1.46 (s, 9H, N(6)-COOC(CH3)3); 1.66 (s, 9Η, N(l)- COOC(CH3)3); 1.88 (td, IH, J=13.1Hz, 9.5Hz, Hβ-9); 2.81 (ddd, IH, J=13.1Hz, 8.1Hz, 3.7Hz, Hα-9); 2.86-2.94 (m, IH, H-8); 3.06 (ddd, IH, J=14.9Hz, 11.6Hz, 2.1Hz, Hα-4); 3.14 (td, IH, J=13.2Hz, 3.6Hz, H-10); 3.31 (dd, IH, J=15.1Hz, 4.0Hz, Hβ-4); 3.65 (td, IH, J=11.4Hz, 4.0Hz, H-5); 3.70 (dd, IH, J=14.2Hz, 5.6Hz, Hα-7); 3.73 (s, 3H, COOCH3); 3.92 (dd, 1Η, J=14.2Ηz, 4.5Hz, Hp-7); 7.03 (d, IH, J=7.2Hz, H-12); 7.20-7.30 (m, 2H, H-2, H-13); 7.80 (br d, 1H, H-14).
EXAMPLE 2 Synthesis of N-[3-(dimethylamino)propyl]-l,6-di(ter -butoxycarbonyl)-ergoIine-8β- carboxamide (XVII, R2 = tert-butyϊ) A mixture of 14.7 g (31.24 mmol) 1.6-di(tert-butoxycarbonyl)-ergoline-8β-carboxylic acid methyl ester (XVI, Ri = methyl,R2 = tert-butyl), 58.8 ml of 3-(dimethylamino)propyl- amine, 29.4 ml of 2-propanol and 3.68 g of 2-hydroxypyridine was stirred at 70°C for 8 hours. The reaction mixture was cooled to ambient temperature, and 230 ml of dichloromethane was added. The resulting mixture was washed with 3x120 ml of sodium chloride solution. The organic layer was dried over anhydrous sodium sulphate. The dried solution was either subjected to the subsequent step without isolation of the product or it was concentrated in vacuum to give 16.4 g (97.1%) of the title compound. 1H NMR (CDCI3, TMS, 500MHz) δ 1.47 (s, 9H, N(6)-COOC(CH3)3); 1.62 (td, 1Η, J=l 3.1 Ηz, 10.4Hz, Hp-9); 1.67 (s, 9H, N(l)-COOC(CH3)3); 1.66-1.76 (m, 2Η, CONHCH2CH2CH2N(CH3)2); 2.25 (s, 6H, CONHCH2CH2CH2N(CH3)2); 2.36-2.45 (m, 2Η, CONHCH2CH2CH2N(CH3)2); 2.75-2.83 (m, IH, H-8); 2.98 (ddd, IH, J=13.4Hz, 8.2 Hz, 3.1Hz, Hα-9); 3.05 (ddd, IH, J=14.2Hz, 11.8Hz, 2.1Hz, Hα-4); 3.13 (td, IH, J=13.3Hz, 3.6Hz, H-10); 3.28 (dd, IH, J=14.8Hz, 3.9Hz, Hβ-4); 3.28-3.42 (m, 2H, CONHCH2CH2CH2N(CH3)2); 3.60 (dd, IH, J=14.8Hz, 6.0Hz, Hα-7); 3.65 (td, IH, J=11.4Hz, 3.9Hz, H-5); 3.92 (dd, IH, J=14.8Hz, 3.7Hz, Hβ-7); 7.04 (d, IH, J=7.4Hz, H-12); 7.22-7.32 (m, 2H, H-2, H-13); 7.62 (t, IH, J=5.0Hz, CONHCH2CH2CH2N(CH3)2); 7.79 (br d, IH, H-
14).
EXAMPLE 3 Synthesis of N-[3-(dimethylamino)propyl]-N-[(ethylamino)carbonyl)-l,6-di(ter^- butoxycarbonyl)-ergoline-8β-carboxa ide (XVIII, R2 = tert-butyϊ) To a solution of 15.5 g (28.67 mmol) of N-[3-(dimethylamino)propyl]-l,6-di(tert- butoxycarbonyl)-ergoline-8β-carboxamide (XVII, R2 = tert-butyl) in 350 ml of dichloromethane 0.8 g of triphenylphosphine, 0.3 g of copper(I) chloride and 6.8 ml (86mmol) of ethyl isocyanate was sequentially added, and the reaction mixture was stirred at 35°C for 4 hours. The reaction mixture was concentrated in vacuum and the product was purified on a silica plug to give 16.9 g (96.4%) of the title compound.
1H NMR (CDC13, TMS, 500MHz) δ 1.19 (t, 3H, J=7.5Hz, CONHCH2CH3); 1.46 (s, 9Η,
N(6)-COOC(CH3)3); 1.66 (s, 9Η, N(l)-COOC(CH3)3); 1.78-1.96 (m, 2Η,
CONCH2CH2CH2N(CH3)2); 1.98 (td, IH, J=13.0Hz, 9.9Hz, Hβ-9); 2.29 (s, 6H,
CONCH2CH2CH2N(CH3)2); 2.32-2.52 (m, 2H, CONCH2CH2CH2N(CH3)2); 2.77 (ddd, IH,
J=13.2Hz, 7.1Hz, 3.7Hz, Hα-9); 3.12-3.24 ( , 2H, Hα-4, H-10); 3.26-3.34 (m, 3H, Hβ-4,
CONHCH2CH3); 3.65 (td, IH, J=11.7Hz, 4.2Hz, H-5); 3.50-3.88 (br m, 5H, H-8, H-7, CONCH2CH2CH2N(CH3)2); 7.03 (d, IH, J=7.5Hz, H-12); 7.20-7.32 (m, 2H, H-2, H-13); 7.79 (br d, IH, H-14); 9.40 (br t, IH, CONHCH2CH3). EXAMPLE 4 Synthesis of N-[3-(dimethylamino)propyl]-N-[(ethylamino)carbonyl)-ergoline-8β- carboxamide (XIX) To 12.7 g (20.76 mmol) of N-[3-(dimethylamino)propyl]-N-[(ethylamino)carbonyl)- l,6-di(tert-butoxycarbonyl)-ergoline-8β-carboxamide (XVIII, R2 = tert-butyl) 230ml of 4M aqueous hydrochloric acid was added, and the reaction was stirred at 35°C for 2 hours. The reaction mixture was cooled to ambient temperature, 200 ml of dichloromethane was added and the pH was adjusted to 11 with concentrated aqueous ammonia solution. The organic layer was separated and the aqueous layer was extracted with 2x60 ml of dichloromethane. The combined organic layer was dried over anhydrous sodium sulphate. The dried solution was either subjected to the subsequent step without isolation of the product or it was concentrated in vacuum to give 8.1 g (94.8%) of the title compound.
1H NMR (CDC13, TMS, 500MHz) δ 1.19 (t, 3H, J=7.3Hz, CONHCH2CH3); 1.80-1.92 (m,
3H, Hp-9, CONCH2CH2CH2N(CH3)2); 2.27 (s, 6H, CONCH2CH2CH2N(CH3)2); 2.39 (t, 2Η,
J=6.7Hz, CONHCH2CH2CH2N(CH3)2); 2.60 (br s, IH, N(6)H); 2.68-2.90 (m, 4H, Hα-4, Hα-
9, H-5, H-10); 2.97 (t, IH, J=12.3Hz, Hβ-7); 3.07 (dd, IH, J=14.5Hz, 4.0Hz, Hβ-4); 3.26-3.42
(m, 4H, Hα-7, H-8, CONHCH2CH3); 3.74-3.92 (m, 2H, CONCH2CH2CH2N(CH3)2); 6.84- 6.90 ( , 2H, H-2, H-12); 7.10-7.20 (m, 2H, H-13, H-14); 8.16 (s, IH, N(l)H); 9.45 (br t, IH, CONHCH2CH3). EXAMPLE 5
Synthesis of 6-aUyl-N-[3-(dimethylamino)propyl]-N-[(ethylamino)carbonyl]-ergoline-8β- carboxamide (T) (Cabergoline). To a suspension of 9.0 g (21.87 mmol) N-[3-(dimethylamino)propyl]-N- [(ethylamino)carbonyl)-ergoline-8β-carboxamide (XLX) in 250 ml of toluene 0.5 g of tetrakis(triphenylphosphine)ρalladium(0) and 5 ml of allyl acetate was added, and the reaction mixture was stirred at ambient temperature for 2 hours. The resulting mixture was washed with 100 ml of water. The organic layer was dried over anhydrous sodium sulphate. The dried solution was concentrated in vacuum and the product was purified on a silica plug to give 9.1 g (92.3 %) of the title compound.
1H NMR (DMSO-d6, TMS, 500MHz) δ 1.10 (t, 3H, J=7.2Hz, CONHCH2CH3); 1.47 (q, 1Η,
J=12.4Ηz, Hp-9); 1.62-1.72 (m, 2H, CONCH2CH2CH2N(CH3)2); 2.15 (s, 6H,
CONCH2CH2CH2N(CH3)2); 2.20-2.30 (m, 2Η, CONHCH2CH2CH2N(CH3)2); 2.32-2.40 (m,
2H, H-5, Hβ-7); 2.54 (dd, IH, J=14.3Hz, 11.2Hz, Hα-4); 2.68-2.84 (m, 2H, Hα-9, H-10); 3.08
(ddd, IH, J=l 1.3Hz, 3.1Hz, 1.8Hz, Hα-7); 3.14-3.22 (m, 2H, CONHCJJ2CH3); 3.26 (dd, IH,
J=14.7Hz, 7.3Hz, HX-N(6)CH2CH=CH2); 3.28-3.38 (m, 2H, Hβ-4, H-8); 3.48 (dd, IH,
J=14.7Hz, 5.8Hz, Hy-N(6)CH2CH=CH2); 3.58-3.68 (m, 2H, CONCH2CH2CH2N(CH3)2); 5.15 (d, IH, J=10.3Hz, HX-N(6)CH2CH=CH2); 5.24 (d, 1Η, J=17.2Ηz, Hy-N(6)CH2CH=CH2); 5.88-5.98 ( , IH, N(6)CH2CH=CH2); 6.75 (d, IH, J=7.0Hz, H-12); 6.97 (s, IH, H-2); 7.01 (t, IH, J=7.5Hz, H-13); 7.13 (d, IH, J=8.0Hz, H-14); 9.04 (t, IH, J=5.0Hz, CONHCH2CH3); 10.60 (s, IH, N(l)H). EXAMPLE 6 Production of polymorphic amorphous form of Cabergoline (I)
a) 10 g of chromatographically purified oily Cabergoline (I) was dissolved in 50 ml of acetone. The solution was concentrated in vacuum at 25-30°C to approx. 15 g. The obtained oily residue was dissolved in 40 ml of acetone, and the solution was concentrated in vacuum at 25-30°C to approx. 12 g. The obtained oily residue was dissolved in 30 ml of acetone again, and the solution was concentrated in vacuum at 25-30°C to 10 g. The obtained solid Cabergoline (I) was dried in vacuum at 25-30°C to solvent-free, to give 9.8 g (98%) of the title compound. b) The same as in Example 6a, but employing methyl acetate as solvent, 9.85 g (98.5%) of the title compound was obtained. c) The same as in Example 6a, but employing dichloromethane as solvent, 9.82 g (98.2%) of the title compound was obtained. According to XRD, DSC, IR analytical studies the crystalline form of the product is amorphous.
X -Ray Powder Diffraction (XRD):
XRD was performed by using Philips PW 1840 Compact powder diffractometer. Differential Scanning Calorimeter (DSC):
The DSC investigations were determined on Mettler Toledo DSC 821 Instrument.
Figure 1 shows the XRD Spectra of amorphous form of Cabergoline (I). Figure 2 shows the DSC Spectra of amorphous form of Cabergoline (I). Figure 3 shows the IR Spectra of amorphous form of Cabergoline (I).

Claims

CLAIMS:
A process for preparing cabergoline (I)
Figure imgf000026_0001
cabergoline (I) comprising the following steps: (a) reacting the compound of formula (XIII)
Figure imgf000026_0002
(XIII) wherein Ri is a Cμ alkyl group, in the presence of a catalyst (i) with a compound of formula (XrV), X-COOR2 (XTV) wherein R2 is an optionally substituted straight or branched Cι-6 alkyl group, X represents a bromine or chlorine atom, or (ii) with a compound of formula (XV), O(COOR2)2 (XV) wherein R2 is a group as defined above for formula (XTV); (b) reacting the obtained carbamate derivative of formula (XVI)
Figure imgf000027_0001
(XVI) wherein Ri and R2 is a group as defined above, with 3-(dimethylamino)propylamine in the presence of a catalyst;
(c) reacting the obtained ergoline-8β-carboxamide derivative of formula (XVII)
Figure imgf000027_0002
(XVII) wherein R2 is a group as defined above, with ethyl isocyanate in the presence of ligand(s) and lb and lib metal group salt catalyst;
(d) reacting the obtained protected N-acylurea derivative of formula (XVIII)
Figure imgf000028_0001
(XVIII) wherein R2 is a group as defined above, with a strong aqueous inorganic acid;
(e) reacting the obtained secondary amine of formula (XLX)
Figure imgf000028_0002
(XLX) with an electrophyl allyl alcohol derivative in the presence of a palladium or nickel containing catalyst and optionally in the presence of ligand(s) to form cabergoline (I).
2. A process according to claim 1 wherein Ri is methyl and R2 is tert-butyl.
3. A process according to any of claims 1 to 2 wherein step (a) is carried out at a temperature of from 0°C to 50°C in the presence of 4-dimethylaminopyridine catalyst in a hydrocarbon halide solvent.
4. A process according to any of claims 1 to 2 wherein step (b) is carried out at a temperature of from 50°C to 70°C in an Ci-6 alkyl alcohol solvent in the presence of 2-hydroxypyridine catalyst.
5. A process according to any of claims 1 to 2 wherein step (c) is carried out in hydrocarbon halide solvent, in the presence of copper(I) chloride and/or copper(II) chloride and/or copper(I) bromide and/or copper(I) iodide catalysts and triphenylphosphine or tri-p- tolylphophine ligand at a temperature of from 30°C to 50°C.
6. A process according to any of claims 1 to 2 wherein step (d) is carried out at a temperature of from 40°C to 80°C in aqueous hydrochloric acid.
7. A process according to any of claims 1 to 2 wherein at step (e) the electrophyl allyl alcohol derivative is allyl acetate, the catalyst is tetrakis(triphenylphosphine)palladium(0), and the reaction is carried out in an aromatic hydrocarbon solvent at a temperature of from 20°C to 50°C.
8. Compounds of formula (XVI)
Figure imgf000030_0001
(XVI) wherein Ri represents a Cμ alkyl group and R2 represents an optionally substituted -β alkyl group.
9. Compound according to claim 8 wherein Ri is methyl and R2 is tert-butyl.
10. Compound of formula (XVII)
Figure imgf000030_0002
(XVII) wherein R2 represents an optionally substituted Ci_6 alkyl group.
11. Compound according to claim 10 wherein R2 is tert-butyl.
12. Compounds of formula (XVIII)
Figure imgf000031_0001
(XVIII) wherein R2 represents an optionally substituted Ci_6 alkyl group.
13. Compound according to claim 12 wherein R2 is tert-butyl.
14. Compound of formula (XLX)
Figure imgf000031_0002
(XLX)
15. The polymorphic amorphous form of Cabergoline (I).
16. Process for the preparation of the polymorphic amorphous form of Cabergoline (I) wherein the chromatographically purified oily Cabergoline (I) is dissolved in a suitable organic solvent and from the obtained solution the solvent is partially removed several times in vacuum at a temperature of from 0°C to 30°C, until not oily but solid product is obtained.
17. A process according to claim 16 wherein the solvent is acetone, methyl acetate or dichloromethane.
PCT/HU2005/000022 2004-03-04 2005-03-02 Process for the preparation of cabergoline WO2005085243A2 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US10/591,202 US7858791B2 (en) 2004-03-04 2005-03-02 Process for the preparation of cabergoline
DE602005021461T DE602005021461D1 (en) 2004-03-04 2005-03-02 PROCESS FOR PREPARING CABERGOLIN
AT05718154T ATE469149T1 (en) 2004-03-04 2005-03-02 METHOD FOR PRODUCING CABERGOLINE
EP05718154A EP1720869B1 (en) 2004-03-04 2005-03-02 Process for the preparation of cabergoline
EA200601628A EA010689B1 (en) 2004-03-04 2005-03-02 Process for the preparation of cabergoline

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
HU0400517A HUP0400517A3 (en) 2004-03-04 2004-03-04 Process for producing cabergoline
HUP0400517 2004-03-04

Publications (2)

Publication Number Publication Date
WO2005085243A2 true WO2005085243A2 (en) 2005-09-15
WO2005085243A3 WO2005085243A3 (en) 2006-04-06

Family

ID=89982034

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/HU2005/000022 WO2005085243A2 (en) 2004-03-04 2005-03-02 Process for the preparation of cabergoline

Country Status (7)

Country Link
US (1) US7858791B2 (en)
EP (1) EP1720869B1 (en)
AT (1) ATE469149T1 (en)
DE (1) DE602005021461D1 (en)
EA (1) EA010689B1 (en)
HU (1) HUP0400517A3 (en)
WO (1) WO2005085243A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1620101A2 (en) * 2003-05-08 2006-02-01 IVAX Pharmaceuticals s.r.o. Polymorphs of cabergoline
WO2008104956A2 (en) * 2007-02-28 2008-09-04 Ranbaxy Laboratories Limited Process for the preparation of amorphous cabergoline
EP2067780A1 (en) * 2007-12-07 2009-06-10 Axxonis Pharma AG Ergoline derivatives as selective radical scavengers for neurons
JP2010507623A (en) * 2006-10-26 2010-03-11 レツク・フアーマシユーテイカルズ・デー・デー A method for preparing crystalline forms of cabergoline via a novel stable solvate of cabergoline
JP2015107980A (en) * 2010-06-11 2015-06-11 ローズ テクノロジーズ Transition metal-catalyzed process for preparation of n-allyl compound and use thereof
JP2015525239A (en) * 2012-06-22 2015-09-03 マップ・ファーマシューティカルズ・インコーポレイテッド New cabergoline derivatives

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5382669A (en) * 1992-03-12 1995-01-17 Farmitalia Carlo Erba S.R.L. Process for the preparation of ergoline derivatives
WO2002085902A1 (en) * 2001-04-16 2002-10-31 Finetech Laboratories Ltd. Process and intermediates for production of cabergoline and related compounds
US20020177709A1 (en) * 2001-04-16 2002-11-28 Gutman Arie L. Novel process and intermediates for production of cabergoline and related compounds

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5382669A (en) * 1992-03-12 1995-01-17 Farmitalia Carlo Erba S.R.L. Process for the preparation of ergoline derivatives
WO2002085902A1 (en) * 2001-04-16 2002-10-31 Finetech Laboratories Ltd. Process and intermediates for production of cabergoline and related compounds
US20020177709A1 (en) * 2001-04-16 2002-11-28 Gutman Arie L. Novel process and intermediates for production of cabergoline and related compounds

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BATTAGLIA R ET AL: "DISPOSITION AND URINARY METABOLIC PATTERN OF CABERGOLINE, A POTENT DOPAMINERGIC AGONIST, IN RAT, MONKEY AND MAN" XENOBIOTICA, TAYLOR AND FRANCIS, LONDON,, GB, vol. 23, no. 12, 1993, pages 1377-1389, XP009040296 ISSN: 0049-8254 *

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1620101A2 (en) * 2003-05-08 2006-02-01 IVAX Pharmaceuticals s.r.o. Polymorphs of cabergoline
EP1620101A4 (en) * 2003-05-08 2008-07-09 Ivax Pharmaceuticals Sro Polymorphs of cabergoline
US7531551B2 (en) 2003-05-08 2009-05-12 Ivax Pharmaceuticals S.R.O. Polymorphs of cabergoline
JP2010507623A (en) * 2006-10-26 2010-03-11 レツク・フアーマシユーテイカルズ・デー・デー A method for preparing crystalline forms of cabergoline via a novel stable solvate of cabergoline
WO2008104956A3 (en) * 2007-02-28 2009-08-13 Ranbaxy Lab Ltd Process for the preparation of amorphous cabergoline
WO2008104956A2 (en) * 2007-02-28 2008-09-04 Ranbaxy Laboratories Limited Process for the preparation of amorphous cabergoline
WO2009071607A2 (en) * 2007-12-07 2009-06-11 Axxonis Pharma Ag Ergoline derivatives as selective radical scavengers for neurons
EP2067780A1 (en) * 2007-12-07 2009-06-10 Axxonis Pharma AG Ergoline derivatives as selective radical scavengers for neurons
WO2009071607A3 (en) * 2007-12-07 2009-09-24 Axxonis Pharma Ag Ergoline derivatives as selective radical scavengers for neurons
JP2015107980A (en) * 2010-06-11 2015-06-11 ローズ テクノロジーズ Transition metal-catalyzed process for preparation of n-allyl compound and use thereof
JP2017137317A (en) * 2010-06-11 2017-08-10 ローズ テクノロジーズ Transition metal-catalyzed processes for preparation of n-allyl compounds and use thereof
JP2015525239A (en) * 2012-06-22 2015-09-03 マップ・ファーマシューティカルズ・インコーポレイテッド New cabergoline derivatives
EP2863747A4 (en) * 2012-06-22 2015-12-23 Map Pharmaceuticals Inc Novel cabergoline derivatives

Also Published As

Publication number Publication date
EA200601628A1 (en) 2007-02-27
HUP0400517A3 (en) 2006-05-29
HUP0400517A2 (en) 2005-11-28
US20070293677A1 (en) 2007-12-20
ATE469149T1 (en) 2010-06-15
DE602005021461D1 (en) 2010-07-08
EP1720869A2 (en) 2006-11-15
WO2005085243A3 (en) 2006-04-06
EA010689B1 (en) 2008-10-30
EP1720869B1 (en) 2010-05-26
HU0400517D0 (en) 2004-05-28
US7858791B2 (en) 2010-12-28

Similar Documents

Publication Publication Date Title
CA2699438C (en) Method for preparing disubstituted piperidine and intermediates
EP2145890B1 (en) Crystallization of hydrohalides of pharmaceutical compounds
EP2062874B1 (en) Process and intermediates for the preparation of aliskiren
EP1720869B1 (en) Process for the preparation of cabergoline
WO2006097345A9 (en) Improved process for making cabergoline
US9403785B2 (en) Process for preparing amorphous cabazitaxel
EP1963254A2 (en) Process for the preparation of lercanidipine and amorphous form of lercanidipine hydrochloride
EP4028397A1 (en) Processes for the synthesis of valbenazine
WO2007072507A2 (en) Polymorphic forms of dolasetron base and processes of preparing dolasetron base, its polymorphic forms and salt thereof
JP2022505626A (en) N-nitrosaccharins
WO2002085902A1 (en) Process and intermediates for production of cabergoline and related compounds
CA1142176B (en) 2-azaergolines and 2-aza-8 (or 9)-ergolenes
US6696568B2 (en) Process and intermediates for production of cabergoline and related compounds
US5338867A (en) Preparation of 4β- amino podophyllotoxin compounds
CA2591071A1 (en) Process for producing [1,4'] bipiperidinyl-1'-carbonyl chloride or hydrochloride thereof
WO2006129781A1 (en) Process for production of dibenzoxepin derivative
US7544802B2 (en) Process for the preparation of 2-(ethoxymethyl)-tropane derivatives
CA3108134A1 (en) Crystalline eltrombopag monoethanolamine salt form d
KR100469030B1 (en) Synthesis of cisapride
WO2008014249A2 (en) Improved preparations of 2 -substituted pyrrolo [2, 3-b] pyrazine ( 4, 7 -diazaindole ) compounds
AU2007213571A1 (en) Production of cabergoline and novel polymorphic form thereof
WO2022241188A1 (en) Enantioselective synthesis of aminotropane compound
JP3864482B2 (en) Method for producing optically active indole derivative
EP4330255A1 (en) Processes for the synthesis of valbenazine
AU2012203941A1 (en) New method for salt preparation

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG 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 MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

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: 2135/KOLNP/2006

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2005718154

Country of ref document: EP

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

WWE Wipo information: entry into national phase

Ref document number: 200601628

Country of ref document: EA

WWP Wipo information: published in national office

Ref document number: 2005718154

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 10591202

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 10591202

Country of ref document: US