WO2012073254A1 - A process for the preparation of pazopanib using novel intermediate - Google Patents

A process for the preparation of pazopanib using novel intermediate Download PDF

Info

Publication number
WO2012073254A1
WO2012073254A1 PCT/IN2011/000781 IN2011000781W WO2012073254A1 WO 2012073254 A1 WO2012073254 A1 WO 2012073254A1 IN 2011000781 W IN2011000781 W IN 2011000781W WO 2012073254 A1 WO2012073254 A1 WO 2012073254A1
Authority
WO
WIPO (PCT)
Prior art keywords
pazopanib
solvent
methanol
formula
methyl
Prior art date
Application number
PCT/IN2011/000781
Other languages
French (fr)
Inventor
Bandi Parthasaradhi Reddy
Kura Rathnakar Reddy
Dasari Muralidhara Reddy
Thungathurthy Srinivasa Rao
Bandi Vamsi Krishna
Original Assignee
Hetero Research Foundation
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 Hetero Research Foundation filed Critical Hetero Research Foundation
Priority to US13/989,541 priority Critical patent/US9150547B2/en
Priority to EP11845075.8A priority patent/EP2646431B1/en
Priority to CA2819118A priority patent/CA2819118A1/en
Publication of WO2012073254A1 publication Critical patent/WO2012073254A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/42One nitrogen atom

Definitions

  • the present invention provides a commercially viable process for preparing pazopanib and its pharmaceutically acceptable acid addition salts thereof in high yields using novel intermediate.
  • the present invention also provides a process for the purification of pazopanib hydrochloride.
  • U.S. patent no. 7,105,530 disclosed pyrimidineamines and their derivatives thereof. These compounds are antineoplastic agents, and are useful in the treatment of various cancers and renal cell carcinoma.
  • pazopanib hydrochloride chemically 5-[4-[N-(2,3-Dimethyl-2H-indazol-6-yl)-N-methylamino]pyrimidin-2- ylamino]-2-methylbenzenesulfonamide hydrochloride.
  • Pazopanib hydrochloride is represented by the following structure:
  • Pazopanib hydrochloride is a potent and selective multi-targeted receptor tyrosine kinase inhibitor of VEGFR (Vascular endothelial growth factor receptors)- 1, VEGFR-2, VEGFR-3, PDGFR (Platelet-derived growth factor receptors )-a/p, and c-kit that blocks tumor growth and inhibits angiogenesis. It has been approved for renal cell carcinoma by the U.S. Food and Drug Administration. Pazopanib hydrochloride may also be active in ovarian cancer and soft tissue sarcoma. Pazopanib hydrochloride also appears effective in the treatment of non-small cell lung carcinoma. Pazopanib hydrochloride is marketed under the brand name Votrient ® by Glaxosmithkline in the form of tablet.
  • pazopanib hydrochloride can be prepared by reacting the N-(2-chloropyrimidin-4-yl)-N,2,3-trimethyl-2H-indazol-6-amine with 5- amino-2-methylbenzenesulfonamide in the presence of hydrochloric acid in isopropanol and ether.
  • pazopanib hydrochloride can be prepared by reacting the N-(2-chloropyrimidin-4-yl)-N,2,3- trimethyl-2H-indazol-6-amine with 5-amino-2-methylbenzenesulfonamide in the presence of hydrochloric acid in ethanol or methanol or tetrahydrofuran or acetonitrile and dioxane.
  • one object of the present invention is to provide a novel process for preparing pazopanib and pharmaceutically acceptable acid addition salts of pazopanib in high yields using novel intermediate.
  • Another object of the present invention is to provide a process for the purification of pazopanib hydrochloride.
  • room temperature refers to a temperature of about 25°C to about 35°C.
  • the base used in step (a) may preferably be an organic base or inorganic base and more preferably the base is inorganic base selected from alkali metal hydroxides, alkali metal carbonates or alkali metal bicarbonates. Still more preferably the inorganic base is sodium bicarbonate or potassium bicarbonate.
  • the solvent used in step (a) may be a solvent or mixture of solvents selected from methanol, ethanol, isopropyl alcohol, n-butanol, toluene, xylene, n-hexane, cyclohexane, methylene chloride, ethylene dichloride, chloroform, carbon tetrachloride, acetone, diethyl ketone, methyl ethyl ketone, methyl propyl ketone, methyl isobutyl ketone, methyl tert-butyl ketone, ethyl acetate, methyl acetate, isopropyl acetate, tert- butyl acetate, ethyl formate, methyl formate, tetrahydrofuran, diethyl ether, diisopropyl ether, tert-butyl methyl ether, dimethylformamide, N, N-dimethyl acetamide and
  • the reaction in step (a) may preferably be carried out at ambient temperatures in the range from about -25°C to 100°C, more preferably at about 0°C to 90°C and still more preferably at about 0°C to 80°C.
  • the alcoholic solvent used in step (b) may preferably be a solvent or mixture of solvents selected from methanol, ethanol, isopropyl alcohol and n-butanol, and more preferably the alcoholic solvent is ethanol or methanol.
  • step (b) may preferably be carried out at an elevated temperature.
  • elevated temperature refers to temperature at above 25°C. More preferably the step (b) is carried out at reflux.
  • a pazopanib or a salt thereof having a purity of at least 99.9%.
  • a process for the purification of pazopanib hydrochloride which comprises crystallizing pazopanib hydrochloride from a solvent system comprising alcohol solvent and water and isolating substantially pure pazopanib hydrochloride.
  • substantially pure pazopanib hydrochloride refers to pazopanib hydrochloride having the purity greater than about 98% by weight, preferably greater than about 99% by weight, and more preferably greater than about 99.9% by weight.
  • the alcoholic solvent used in the process may preferably be a solvent or mixture of solvents selected from methanol, ethanol, isopropanol and n-butanol, and more preferably the alcoholic solvent is methanol or isopropanol. Still more preferably the alcoholic solvent is methanol.
  • Isolation of substantially pure pazopanib hydrochloride in the process can be performed by conventional methods such as cooling, removal of solvents, concentrating the reaction mass, adding an anti-solvent, extraction with a solvent and the like.
  • the pH of the reaction mass was adjusted to 8.0 to 8.5 with hydrochloric acid solution (40 ml) and then added ethyl acetate (400 ml). Then the layers were separated and the aqueous layer was extracted with ethyl acetate. The organic layer was dried with sodium sulfate and treated with carbon. The combined organic layers were washed with sodium chloride solution and dried with sodium sulfate. The organic layer was treated with carbon and filtered through hi-flow bed. The solvent was distilled off under vacuum at below 50°C to obtain a residual mass. To the residual mass was added diisopropyl ether (75 ml) and stirred for 1 hour, filtered. The solid obtained was dried to give 10 gm of N,2,3-trimethyl-2H-indazol-6-amine.
  • Pazopanib hydrochloride (5 gm; HPLC Purity: 97.5%) as obtained in example 3 was dissolved in a mixture of methanol (100 ml) and water (10 ml) at room temperature and then heated to reflux. The reaction mass was maintained for 30 minutes at reflux and filtered. The filtrate obtained was cooled to room temperature and maintained for 2 hours at room temperature. The solid obtained was collected by filtration and dried to obtain 3.5 gm of pazopanib hydrochloride (HPLC Purity: 99.9%).
  • Example 5 Example 5:
  • Pazopanib hydrochloride 22 gm; HPLC Purity: 98%), methanol (528 ml), water (55 ml) and concentrated hydrochloric acid (0.2 ml) were added at room temperature. The contents were heated to reflux and maintained for 30 minutes, filtered. Take the filtrate and the solvent was distilled off under vacuum to obtain a residual mass. The residual mass was then cooled to room temperature and stirred for 30 minutes at room temperature. The contents were further cooled to 0 to 5°C, stirred for 1 hour and filtered. The solid obtained was dried to give 19 gm of pazopanib hydrochloride (HPLC Purity: 99.85%).
  • Pazopanib hydrochloride (10 gm; HPLC Purity: 96%), methanol (250 ml), water (25 ml) and concentrated hydrochloric acid (0.1 ml) were added at room temperature. The contents were heated to reflux and maintained for 30 minutes, filtered. The filtrate obtained was then cooled to room temperature and stirred for 30 minutes at room temperature. The contents further cooled to 0 to 10°C and stirred for 1 hour. The separated solid was filtered and dried to obtain 6.6 gm of pazopanib hydrochloride (HPLC Purity: 99.8%).
  • Pazopanib hydrochloride (22 gm; HPLC Purity: 97%) was dissolved in a mixture of isopropanol (132 ml) and water (20 ml) at room temperature and then heated to reflux. The reaction mass was maintained for 1 hour at reflux and then cooled to room temperature. The reaction mass was stirred for 1 hour at room temperature and filtered. The solid obtained was dried to give 18 gm of pazopanib hydrochloride (HPLC Purity: 99.8%).

Abstract

The present invention provides a commercially viable process for preparing pazopanib and its pharmaceutically acceptable acid addition salts thereof in high yields using novel intermediate. The present invention also provides a process for the purification of pazopanib hydrochloride.

Description

A PROCESS FOR THE PREPARATION OF PAZOPANIB USING NOVEL
INTERMEDIATE
Filed of the Invention
The present invention provides a commercially viable process for preparing pazopanib and its pharmaceutically acceptable acid addition salts thereof in high yields using novel intermediate. The present invention also provides a process for the purification of pazopanib hydrochloride.
Background of the Invention
U.S. patent no. 7,105,530 disclosed pyrimidineamines and their derivatives thereof. These compounds are antineoplastic agents, and are useful in the treatment of various cancers and renal cell carcinoma. Among them pazopanib hydrochloride, chemically 5-[4-[N-(2,3-Dimethyl-2H-indazol-6-yl)-N-methylamino]pyrimidin-2- ylamino]-2-methylbenzenesulfonamide hydrochloride. Pazopanib hydrochloride is represented by the following structure:
Figure imgf000002_0001
Pazopanib hydrochloride is a potent and selective multi-targeted receptor tyrosine kinase inhibitor of VEGFR (Vascular endothelial growth factor receptors)- 1, VEGFR-2, VEGFR-3, PDGFR (Platelet-derived growth factor receptors )-a/p, and c-kit that blocks tumor growth and inhibits angiogenesis. It has been approved for renal cell carcinoma by the U.S. Food and Drug Administration. Pazopanib hydrochloride may also be active in ovarian cancer and soft tissue sarcoma. Pazopanib hydrochloride also appears effective in the treatment of non-small cell lung carcinoma. Pazopanib hydrochloride is marketed under the brand name Votrient® by Glaxosmithkline in the form of tablet.
Processes for the preparation of pazopanib hydrochloride and related compounds were disclosed in U.S. patent no. 7,105,530 and U.S. patent no. 7,262,203.
According to U.S. patent no. 7,105,530, pazopanib hydrochloride can be prepared by reacting the N-(2-chloropyrimidin-4-yl)-N,2,3-trimethyl-2H-indazol-6-amine with 5- amino-2-methylbenzenesulfonamide in the presence of hydrochloric acid in isopropanol and ether.
U.S. patent application publication no. 2006/0252943 disclosed a process for the preparation of pazopanib hydrochloride. According to this patent, pazopanib hydrochloride can be prepared by reacting the N-(2-chloropyrimidin-4-yl)-N,2,3- trimethyl-2H-indazol-6-amine with 5-amino-2-methylbenzenesulfonamide in the presence of hydrochloric acid in ethanol or methanol or tetrahydrofuran or acetonitrile and dioxane.
We have discovered a novel process for the preparation of pazopanib using novel intermediate. The process of the invention results in higher yields compared with the known process.
We have also discovered a process for the purification of pazopanib hydrochloride.
Thus, one object of the present invention is to provide a novel process for preparing pazopanib and pharmaceutically acceptable acid addition salts of pazopanib in high yields using novel intermediate.
Another object of the present invention is to provide a process for the purification of pazopanib hydrochloride. Detailed Description of the Invention
As used herein the term "room temperature" refers to a temperature of about 25°C to about 35°C.
According to one aspect of the present invention, there is provided a novel process for the preparation of pazopanib of formula I:
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof:
which comprises:
a) reacting 5-amino-2-methylbenzenesulfonamide of formula II:
Figure imgf000004_0001
with a 2,4-dichloropryrimidine of formula III:
Figure imgf000004_0002
in the presence of a base and a solvent to give 5-(4-chloropyrimidin-2ylamino)-2- methylbenzenesulfonamide of formula IV; and
Figure imgf000004_0003
b) condensing the 5-(4-chloropyrimidin-2ylamino)-2-methylbenzenesulfonamide of formula IV with the N,2,3-trimethyl-2H-indazol-6-amine of formula V:
Figure imgf000004_0004
in an alcoholic solvent to give a pazopanib of formula I and optionally converting pazopanib formed into the pharmaceutically acceptable acid addition salt of pazopanib. The base used in step (a) may preferably be an organic base or inorganic base and more preferably the base is inorganic base selected from alkali metal hydroxides, alkali metal carbonates or alkali metal bicarbonates. Still more preferably the inorganic base is sodium bicarbonate or potassium bicarbonate.
Preferably the solvent used in step (a) may be a solvent or mixture of solvents selected from methanol, ethanol, isopropyl alcohol, n-butanol, toluene, xylene, n-hexane, cyclohexane, methylene chloride, ethylene dichloride, chloroform, carbon tetrachloride, acetone, diethyl ketone, methyl ethyl ketone, methyl propyl ketone, methyl isobutyl ketone, methyl tert-butyl ketone, ethyl acetate, methyl acetate, isopropyl acetate, tert- butyl acetate, ethyl formate, methyl formate, tetrahydrofuran, diethyl ether, diisopropyl ether, tert-butyl methyl ether, dimethylformamide, N, N-dimethyl acetamide and dimethyl sulfoxide. Most preferably the solvents are methanol, ethanol, tetrahydrofuran and diisopropyl ether, and still more preferably the solvents are ethanol and tetrahydrofuran.
The reaction in step (a) may preferably be carried out at ambient temperatures in the range from about -25°C to 100°C, more preferably at about 0°C to 90°C and still more preferably at about 0°C to 80°C.
The alcoholic solvent used in step (b) may preferably be a solvent or mixture of solvents selected from methanol, ethanol, isopropyl alcohol and n-butanol, and more preferably the alcoholic solvent is ethanol or methanol.
The reaction in step (b) may preferably be carried out at an elevated temperature. The term "elevated temperature" refers to temperature at above 25°C. More preferably the step (b) is carried out at reflux.
According to another aspect of the present invention, there is provided a novel compound of formula IV:
Figure imgf000005_0001
According to another aspect of the present invention, there is provided a pazopanib or a salt thereof having a purity of at least 99.9%.
According to another aspect of the present invention, there is provided a process for the purification of pazopanib hydrochloride, which comprises crystallizing pazopanib hydrochloride from a solvent system comprising alcohol solvent and water and isolating substantially pure pazopanib hydrochloride.
The term "substantially pure pazopanib hydrochloride" refers to pazopanib hydrochloride having the purity greater than about 98% by weight, preferably greater than about 99% by weight, and more preferably greater than about 99.9% by weight.
The alcoholic solvent used in the process may preferably be a solvent or mixture of solvents selected from methanol, ethanol, isopropanol and n-butanol, and more preferably the alcoholic solvent is methanol or isopropanol. Still more preferably the alcoholic solvent is methanol.
Isolation of substantially pure pazopanib hydrochloride in the process can be performed by conventional methods such as cooling, removal of solvents, concentrating the reaction mass, adding an anti-solvent, extraction with a solvent and the like.
The purity of pazopanib hydrochloride is measured by High performance liquid chromatography (HPLC).
The invention will now be further described by the following example, which is illustrative rather than limiting.
Examples
Example 1:
Preparation of 5-(4-chloropyrimidin-2ylamino)-2-methyIbenzenesulfonamide
To a mixture of 5-amino-2-methylbenzenesulfonamide (20 gm) in ethanol (208 ml) and tetrahydrofuran (52 ml) was added 2,4-dichloropryrimidine (44 gm) and sodium bicarbonate (36 gm) at room temperature. The contents were heated to 70 to 75°C and maintained for 13 hours. The reaction mass was then cooled to 10°C and maintained for 2 hours. The reaction mass was filtered and the solvent was distilled off under vacuum at below 50 to 55°C to obtain a residual mass. To the residual mass was added ethyl acetate (100 ml) and stirred for 1 hour, filtered. The solid obtained was dried to give 15.5 gm of 5-(4-chloropyrimidin-2ylamino)-2-methylbenzenesulfonamide. Example 2:
Preparation of N,2,3-trimethyI-2H-indazol-6-amine
Sodium methoxide (19 gm) was dissolved in methanol (610 ml) and then added 2,3-dimethyl-2H-indazol-6-amine (13 gm). The reaction mixture was stirred for 15 minutes and then added paraformaldehyde (3.9 gm). The contents were heated to 60°C and stirred for 10 hours. The reaction mass was then cooled to room temperature and maintained for 4 hours 30 minutes. Sodium borohydride (2.8 gm) was added to the reaction mass slowly at room temperature and then heated to reflux. The reaction mass was maintained for 2 hours at reflux and then cooled to room temperature. The reaction mass was stirred for 14 hours at room temperature and then added sodium hydroxide solution (1M, 100 ml). The pH of the reaction mass was adjusted to 8.0 to 8.5 with hydrochloric acid solution (40 ml) and then added ethyl acetate (400 ml). Then the layers were separated and the aqueous layer was extracted with ethyl acetate. The organic layer was dried with sodium sulfate and treated with carbon. The combined organic layers were washed with sodium chloride solution and dried with sodium sulfate. The organic layer was treated with carbon and filtered through hi-flow bed. The solvent was distilled off under vacuum at below 50°C to obtain a residual mass. To the residual mass was added diisopropyl ether (75 ml) and stirred for 1 hour, filtered. The solid obtained was dried to give 10 gm of N,2,3-trimethyl-2H-indazol-6-amine.
Example 3:
Preparation of pazopanib hydrochloride
5-(4-Chloropyrimidin-2ylamino)-2-methylbenzenesulfonamide (17 gm) as obtained in example 1, N,2,3-trimethyl-2H-indazol-6-amine (10 gm) as obtained in example 2 and ethanol (166 ml) were added at room temperature and then heated to reflux. The reaction mass was maintained for 3 hours at reflux and then added concentrated hydrochloric acid (1 ml). The reaction mass was maintained for 10 hours at reflux and then cooled to room temperature. The separated solid was filtered and dried to obtain 17 gm of pazopanib hydrochloride (HPLC Purity: 97.5%). Example 4:
Purification of pazopanib hydrochloride
Pazopanib hydrochloride (5 gm; HPLC Purity: 97.5%) as obtained in example 3 was dissolved in a mixture of methanol (100 ml) and water (10 ml) at room temperature and then heated to reflux. The reaction mass was maintained for 30 minutes at reflux and filtered. The filtrate obtained was cooled to room temperature and maintained for 2 hours at room temperature. The solid obtained was collected by filtration and dried to obtain 3.5 gm of pazopanib hydrochloride (HPLC Purity: 99.9%). Example 5:
Purification of pazopanib hydrochloride
Pazopanib hydrochloride (22 gm; HPLC Purity: 98%), methanol (528 ml), water (55 ml) and concentrated hydrochloric acid (0.2 ml) were added at room temperature. The contents were heated to reflux and maintained for 30 minutes, filtered. Take the filtrate and the solvent was distilled off under vacuum to obtain a residual mass. The residual mass was then cooled to room temperature and stirred for 30 minutes at room temperature. The contents were further cooled to 0 to 5°C, stirred for 1 hour and filtered. The solid obtained was dried to give 19 gm of pazopanib hydrochloride (HPLC Purity: 99.85%).
Example 6:
Purification of pazopanib hydrochloride
Pazopanib hydrochloride (10 gm; HPLC Purity: 96%), methanol (250 ml), water (25 ml) and concentrated hydrochloric acid (0.1 ml) were added at room temperature. The contents were heated to reflux and maintained for 30 minutes, filtered. The filtrate obtained was then cooled to room temperature and stirred for 30 minutes at room temperature. The contents further cooled to 0 to 10°C and stirred for 1 hour. The separated solid was filtered and dried to obtain 6.6 gm of pazopanib hydrochloride (HPLC Purity: 99.8%).
Example 7: Purification of pazopanib hydrochloride
Pazopanib hydrochloride (22 gm; HPLC Purity: 97%) was dissolved in a mixture of isopropanol (132 ml) and water (20 ml) at room temperature and then heated to reflux. The reaction mass was maintained for 1 hour at reflux and then cooled to room temperature. The reaction mass was stirred for 1 hour at room temperature and filtered. The solid obtained was dried to give 18 gm of pazopanib hydrochloride (HPLC Purity: 99.8%).

Claims

We claim:
1. A novel p :
Figure imgf000010_0001
or a pharmaceutically acceptable salt thereof:
which comprises:
a) reacting 5-amino-2-methylbenzenesulfonamide of formula II:
Figure imgf000010_0002
with a 2,4-dichloropryrimidine of formula III:
Figure imgf000010_0003
in presence of a base and a solvent to give 5-(4-chloropyrimidin-2ylamino)-2- methylbenzenesulfonamide of formula IV; and
Figure imgf000010_0004
b) condensing the 5-(4-chloropyrimidin-2ylamino)-2-methylbenzenesulfonamide of formula IV with the N,2,3-trimethyl-2H-indazol-6-amine of formula V:
■V
Figure imgf000010_0005
in an alcoholic solvent to give a pazopanib of formula I and optionally converting pazopanib formed into the pharmaceutically acceptable acid addition salt of pazopanib.
2. The process as claimed in claim 1, wherein the base used in step (a) is an organic base or inorganic base.
3. The process as claimed in claim 2, wherein the base is inorganic base selected from alkali metal hydroxides, alkali metal carbonates or alkali metal bicarbonates.
4. The process as claimed in claim 3, wherein the inorganic base is sodium bicarbonate or potassium bicarbonate.
5. The process as claimed in claim 1, wherein the solvent is a solvent or mixture of solvents selected from methanol, ethanol, isopropyl alcohol, n-butanol, toluene, xylene, n-hexane, cyclohexane, methylene chloride, ethylene dichloride, chloroform, carbon tetrachloride, acetone, diethyl ketone, methyl ethyl ketone, methyl propyl ketone, methyl isobutyl ketone, methyl tert-butyl ketone, ethyl acetate, methyl acetate, isopropyl acetate, tert-butyl acetate, ethyl formate, methyl formate, tetrahydrofuran, diethyl ether, diisopropyl ether, tert-butyl methyl ether, dimethylformamide, N, N-dimethyl acetamide and dimethyl sulfoxide.
6. The process as claimed in claim 5, wherein the solvents are methanol, ethanol, tetrahydrofuran and diisopropyl ether.
7. The process as claimed in claim 1, wherein the alcoholic solvent used in step (b) is a solvent or mixture of solvents selected from methanol, ethanol, isopropyl alcohol and n-butanol.
8. The process as claimed in claim 7, wherein the alcoholic solvent is ethanol or methanol.
9. The process as claimed in claim 1, wherein the reaction in step (b) is carried out at an elevated temperature.
10. The process as claimed in claim 9, wherein the reaction in step (b) is carried out at above 25°C.
11. A novel compound of formula IV:
Figure imgf000012_0001
12. A pazopanib or a salt thereof having a purity of at least 99.9%.
13. A process for the purification of pazopanib hydrochloride, which comprises crystallizing pazopanib hydrochloride from a solvent system comprising alcohol solvent and water and isolating substantially pure pazopanib hydrochloride.
14. The process as claimed in claim 13, wherein the alcoholic solvent used in the process is a solvent or mixture of solvents selected from methanol, ethanol, isopropanol and n-butanol.
15. The process as claimed in claim 14, wherein the alcoholic solvent is methanol or isopropanol.
PCT/IN2011/000781 2010-11-29 2011-11-11 A process for the preparation of pazopanib using novel intermediate WO2012073254A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US13/989,541 US9150547B2 (en) 2010-11-29 2011-11-11 Process for the preparation of pazopanib using novel intermediate
EP11845075.8A EP2646431B1 (en) 2010-11-29 2011-11-11 A process for the preparation of pazopanib using novel intermediate
CA2819118A CA2819118A1 (en) 2010-11-29 2011-11-11 A process for the preparation of pazopanib using novel intermediate

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN3590/CHE/2010 2010-11-29
IN3590CH2010 2010-11-29

Publications (1)

Publication Number Publication Date
WO2012073254A1 true WO2012073254A1 (en) 2012-06-07

Family

ID=46171268

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IN2011/000781 WO2012073254A1 (en) 2010-11-29 2011-11-11 A process for the preparation of pazopanib using novel intermediate

Country Status (4)

Country Link
US (1) US9150547B2 (en)
EP (1) EP2646431B1 (en)
CA (1) CA2819118A1 (en)
WO (1) WO2012073254A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103232443A (en) * 2013-02-01 2013-08-07 天津药物研究院 Indazole derivative crystal and its preparation method and use
WO2014085373A1 (en) * 2012-11-27 2014-06-05 Glaxosmithkline Llc Combination
WO2015031604A1 (en) 2013-08-28 2015-03-05 Crown Bioscience, Inc. Gene expression signatures predictive of subject response to a multi-kinase inhibitor and methods of using the same
CN104557881A (en) * 2014-12-30 2015-04-29 山东博迈康药物研究有限公司 Preparation method of pazopanib hydrochloride crystal form
CN110028495A (en) * 2019-05-24 2019-07-19 济南爱思医药科技有限公司 The environment-friendly preparation method thereof of pazopanib hydrochloride
US10730859B2 (en) 2013-11-05 2020-08-04 Laurus Labs Limited Process for the preparation of pazopanib or a pharmaceutically acceptable salt thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060252943A1 (en) * 2002-06-17 2006-11-09 Amogh Boloor Chemical process
US20080269170A1 (en) * 2005-07-11 2008-10-30 Sanofi-Aventis Novel 2,4-Dianilinopyrimidine Derivatives, the Preparation Thereof, Their Use as Medicaments, Pharmaceutical Compositions and, in Particular, as IKK Inhibitors

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE122010000038I1 (en) 2000-12-21 2011-01-27 Glaxosmithkline Llc PYRIMIDINAMINES AS ANGIOGENESIS MODULATORS
US8084457B2 (en) * 2003-09-15 2011-12-27 Lead Discovery Center Gmbh Pharmaceutically active 4,6-disubstituted aminopyrimidine derivatives as modulators of protein kinases

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060252943A1 (en) * 2002-06-17 2006-11-09 Amogh Boloor Chemical process
US20080269170A1 (en) * 2005-07-11 2008-10-30 Sanofi-Aventis Novel 2,4-Dianilinopyrimidine Derivatives, the Preparation Thereof, Their Use as Medicaments, Pharmaceutical Compositions and, in Particular, as IKK Inhibitors

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
LUO G. ET AL.: "Microwave-assisted synthesis of aminopyrimidines", TETRAHEDRON LETTERS, vol. 43, no. 33, 2002, pages 5739 - 5742, XP004372432 *
See also references of EP2646431A4 *

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014085373A1 (en) * 2012-11-27 2014-06-05 Glaxosmithkline Llc Combination
CN103232443A (en) * 2013-02-01 2013-08-07 天津药物研究院 Indazole derivative crystal and its preparation method and use
WO2015031604A1 (en) 2013-08-28 2015-03-05 Crown Bioscience, Inc. Gene expression signatures predictive of subject response to a multi-kinase inhibitor and methods of using the same
US10730859B2 (en) 2013-11-05 2020-08-04 Laurus Labs Limited Process for the preparation of pazopanib or a pharmaceutically acceptable salt thereof
US11299477B2 (en) 2013-11-05 2022-04-12 Laurus Labs Limited Process for the preparation of Pazopanib or a pharmaceutically acceptable salt thereof
US11427570B2 (en) 2013-11-05 2022-08-30 Laurus Labs Limited Process for the preparation of pazopanib or a pharmaceutically acceptable salt thereof
CN104557881A (en) * 2014-12-30 2015-04-29 山东博迈康药物研究有限公司 Preparation method of pazopanib hydrochloride crystal form
CN104557881B (en) * 2014-12-30 2017-08-11 山东博迈康药物研究有限公司 A kind of preparation method of pazopanib hydrochloride crystal formation
CN110028495A (en) * 2019-05-24 2019-07-19 济南爱思医药科技有限公司 The environment-friendly preparation method thereof of pazopanib hydrochloride

Also Published As

Publication number Publication date
US9150547B2 (en) 2015-10-06
US20130245262A1 (en) 2013-09-19
EP2646431A4 (en) 2014-04-30
EP2646431A1 (en) 2013-10-09
CA2819118A1 (en) 2012-06-07
EP2646431B1 (en) 2016-04-06

Similar Documents

Publication Publication Date Title
EP2646431B1 (en) A process for the preparation of pazopanib using novel intermediate
JP5535082B2 (en) Method for synthesizing bosentan, polymorphic forms thereof and salts thereof
CA2848631C (en) Rilpivirine hydrochloride
AU2002360386A1 (en) Amorphous and crystalline forms of losartan potassium and process for their preparation
WO2012082972A1 (en) Crystalline forms of 5-chloro-n2-(2-isopropoxy-5-methyl-4-piperidin-4-yl-phenyl)-n4[2-(propane-2-sulfonyl)-phenyl]-pyrimidine-2,4-diamine
EP1476442A2 (en) Lansoprazole polymorphs and processes for preparation thereof
CZ2005679A3 (en) Method of removing triphenylmethane protecting group in anti-hypertension medicament precursors
EP2294056A1 (en) Bosentan salts
WO2011018101A1 (en) Lenalidomide salts
US9624207B2 (en) Polymorphs of azilsartan medoxomil
CA2912902A1 (en) Polymorphic forms of nilotinib hydrochloride
WO2012001695A1 (en) A process for etra virine intermediate and polymorphs of etravirine
JP2007524643A (en) Preparation of tegaserod and tegaserod maleate
WO2017191539A1 (en) Process for the preparation dl-proline co-crystal of dapagliflozin
CN104829590B (en) Method for purifying trelagliptin
WO2008111016A1 (en) Process for the preparation of pure prulifloxacin
WO2015049698A2 (en) Process for regorafenib
WO2013150544A2 (en) Ivabradine hydrochloride solid dispersion
WO2010146428A1 (en) An improved process for the preparation of rabeprazole
WO2015092624A1 (en) Nilotinib mono-oxalate and its crystalline form
WO2013162390A1 (en) Process for preparation of high purity n- [2- (diethylamine) ethyl] - 5 - formyl - 2, 4 - dimethyl - 1h - pyrrole - 3 - carboxyamide
WO2011020189A1 (en) Forms of dexlansoprazole and processes for the preparation thereof
WO2014136126A2 (en) A process for preparing erlotinib hydrochloride form a
US20150336900A1 (en) Process for the Synthesis of Etravirine and Its Intermediates
US20150291574A1 (en) Novel polymorphs of azilsartan

Legal Events

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

Ref document number: 11845075

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2011845075

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 13989541

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2819118

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE