WO2015090259A1 - Method of preparing anhydrous polymorphic form n-6 of dasatinib - Google Patents

Method of preparing anhydrous polymorphic form n-6 of dasatinib Download PDF

Info

Publication number
WO2015090259A1
WO2015090259A1 PCT/CZ2014/000160 CZ2014000160W WO2015090259A1 WO 2015090259 A1 WO2015090259 A1 WO 2015090259A1 CZ 2014000160 W CZ2014000160 W CZ 2014000160W WO 2015090259 A1 WO2015090259 A1 WO 2015090259A1
Authority
WO
WIPO (PCT)
Prior art keywords
dasatinib
solvate
temperature
sample
mecn
Prior art date
Application number
PCT/CZ2014/000160
Other languages
French (fr)
Inventor
Jan RYMES
Michal Zapadlo
Ondrej Dammer
Original Assignee
Zentiva, K.S.
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 Zentiva, K.S. filed Critical Zentiva, K.S.
Publication of WO2015090259A1 publication Critical patent/WO2015090259A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the invention relates to a new method of preparing the polymorphic anhydrous form N-6 of dasatinib ; chemically: (N-(2-cbioro-6-methylphenyl)-2- [[6-[4-(2-hydroxyethyl)- 1 - piperazinyl]-2-me1hyl-4-pyrirriidinyl]amino]-5-thiazole carboxamide, CAS: 302962-49-8)), described by the formula (1), and its key solvates, enabling formation of the chemically highly pure Form N-6.
  • CML chronic myeloid leukaemia
  • Hl-7 A large number of polymorphic forms of dasatinib are described in the literature, designated as: Hl-7, BU-2, E2-1, N-6, T1H1-7, and T1E2-1.
  • Patent document WO2005/077975 A2 describes the preparation of dasatinib monohydrate (Hl-7) and butanol solvate (BU-2).
  • Another patent document (US2006/0004067A1) linked up with the previous one describes two ethanol solvates (E2-1, T1E2-1) and further two anhydrous forms (N-6, T1H1-7).
  • a large number of preparations of various polymorphic forms are also described in TEVA patent application WO 2009/053854 A2.
  • the present invention relates to a new method of preparing the anhydrous Form N-6, in particular new use of selected solvate forms (MeOH, MeCN) and their mixtures, which allow conversion to the anhydrous Form N-6 under elevated temperature and vacuum. This phenomenon has also been observed under laboratory conditions, only with a lower conversion rate. From the viewpoint of industrial utilization, this is a very simple, robust, and relatively fast process of preparing the anhydrous Form N-6; its production has also been reproducibly tested in kilogram amounts.
  • selected solvate forms MeOH, MeCN
  • the preparation of the compound of formula (1) includes the following direct steps 1 to 3 (Scheme 1).
  • Next step is reaction of the (SNAT) intermediate (3) 2-(6-chloro-2-methylpyrimidine-4-ylamino)-N-(2-chloro-6- methylphenyl)thiazole-5-carboxamide with l-(2-hydroxyethyl)-piperazine (2) in an environment of NMP.
  • the reaction runs in the temperature range of 65 to 100°C.
  • Acetonitrile is used as the anti-solvent for isolation of crude dasatinib (1).
  • the crude product is dried in vacuo at a temperature of 55 - 65°C.
  • An NMP solvate is obtained with predominant amounts of two residual solvents NMP with an admixture of MeCN.
  • the final step of the synthesis is crystallization from the mixture of solvents MeOH and MeCN at a temperature of 55 - 62°C.
  • the obtained isolated wet crystalline product again shows the solvate structure corresponding to a MeCN solvate (Fig. 2) or a MeOH solvate (Fig. 3).
  • the type to be isolated depends on the selected procedure of preparation and on the ratio of both solvents (examples 3 + 4).
  • the crystallized wet product is further dried in vacuo at a temperature in the range of 55 - 65°C. After drying for 2 - 3 hours, the expected polymorphic Form N-6 is obtained. The duration of drying depends on residual moisture in the sample, type of the drying oven, attained vacuum, and heating rate to the final temperature of drying.
  • the described process is an efficient method of preparing dasatinib that is also convenient for larger scales.
  • the procedure according to Scheme 1 makes it possible to obtain, in a reproducible way, dasatinib (1) characterized by chemical purity higher than 99.8 % and containing individual impurities under 0.1 % (according to UPLC).
  • Figure 1 X-ray powder pattern of polymorphic anhydrous Form N-6 of dasatinib
  • Figure 2 X-ray powder pattern of the MeCN solvate form of dasatinib
  • Figure 3 X-ray powder pattern corresponding to the MeOH solvate of dasatinib
  • Figure 4 Evolution of X-ray powder patterns in the course of conversion of the MeCN solvate form to the Form N-6 during drying in vacuo and at a temperature of 55 °C (shelf drier).
  • Example 1 serve for a more detailed illustration of the method of producing dasatinib of the polymorphic anhydrous Form N-6 according to the present invention on the industrial scale.
  • the temperature of the reaction mixture is kept between 15 and 30°C. After the dosing is completed, the reaction mixture is stirred at 20 ⁇ 2°C for 60 min. The reaction mixture is cooled to 10 - 15°C, 12.5 1 of demineralized water is added dropwise, and the mixture is stirred for 15 min. Then, its temperature is brought to 5°C and diluted 1.5 M HCl (2,550 ml of 35 % HCl + 16.5 1 of demi water) is added dropwise within about 35 - 45 min. The amount of the HCl solution is chosen in order to reach final pH of 4.0 - 6.5 (optimally 5.0 - 5.5).
  • the stirring goes on at 5 - 8°C for 15 - 20 min (checking of pH, adjusted, as required). 22.5 1 of acetone is added, the mixture is heated to 40°C, and stirred at this temperature for 30 minutes. Then it is cooled down slowly to 10 - 15°C and stirred for 1.5 h. The separated product is centrifuged, washed with a warm (40 °C) 1 :1 mixture of acetone / demi water (1 x 5.0 1) and then with cold acetone (1 x 5.0 1). The product is dried in a vacuum drier at 55 - 65°C for 12 hours.
  • dasatinib intermediate (3) CAS: 302964-08-5 (4.500 kg) is suspended in 18.0 I of NMP.
  • the suspension is heated to a temperature of 67 - 68°C.
  • a solution of HEPIP (7.313 kg of HEPIP + 3.8 1 of NMP) is prepared and added dropwise to the reaction mixture within 5 - 10 min. During the addition the temperature is kept at 65 ⁇ 3°C. After the addition is completed, the mixture is stirred at 65°C for 60 min. Then, temperature is brought to 100°C and stirring goes on for 20 min. All the time, the reaction is carried out under N 2 and access of light is prevented! After the reaction is completed, the reaction mixture is cooled down to 40 - 45°C and filtered.
  • the filter is washed with 1.0 1 NMP.
  • the filtered reaction mixture is added dropwise to MeCN (113 1), heated at 25 - 30°C, within 15 - 20 min. Stirring continues at 25 - 30°C for 30 min.
  • the mixture is heated to 45 - 50°C and stirred for 0.5 h.
  • Then, is slowly cooled down to 10 - 15°C within 75 - 90 min and stirred for 1.0 - 1.5 h.
  • the separated product is centrifuged, and washed with cold (2 - 8°C) MeCN (2 x 6.0 1). It is dried in a vacuum drier at 60°C for 12 hours. Yield: 82%, sum of impurities according to UPLC: 0.2%; content of residual solvents: 30,000 ppm NMP, 17,000 ppm CH3OH; X-ray powder pattern corresponds to the NMP solvate.
  • the crude dasatinib (1.00 kg) is suspended in 10.5 1 of MeCN and 17.5 1 of MeOH. The suspension is heated up to 58 - 62°C and stirred for 0.5 h. Then, it is cooled down to 40 - 45°C and stirred for 1 hour. Cooling is continued to -5 to 0°C and stirred for 1.0 - 1.5 h. The separated product is centrifuged, and washed with cold MeCN (2 x 3.0 1). All the time, the crystallization is carried out under the inert atmosphere of N 2 and access of light is prevented! Drying is carried out as in Example 5. The X-ray pattern of the wet sample corresponds to the MeCN solvate.
  • the MeCN solvate is characterized by the reflexions listed in Table 1:
  • the crude dasatinib (1.00 kg) is suspended in 5.0 1 of MeCN and 25 1 of MeOH.
  • the suspension is heated up to 58 - 62°C and stirred for 0.5 h.
  • it is cooled down to 30 - 35°C and stirred for 1 hour. Cooling is continued to -5 to 0 °C and stirred for 1.0 - 1.5 h.
  • the separated product is centnfuged, and washed with cold MeCN (2 x 3.0 1). All the time, the crystallization is carried out under the inert atmosphere of N 2 and access of light is prevented! Drying is carried out as in Example 6.
  • the X-ray pattern of the wet sample corresponds to the MeOH solvate and is characterized by the reflexions listed in Table 2: Table 2: MeOH solvate
  • the wet crystalline dasatinib (solvate form - MeCN, Example 3) is dried in a vacuum tray drier with suction of N 2 at 55 - 65 °C for 16 h. Yield: 91% (from crude API); sum of impurities according to UPLC: 0.11%, content of residual solvents less than 500 ppm, content of water according to KF 0.5%; X-ray pattern of the dry sample corresponds to the pure anhydrous Form N-6.
  • the wet crystallized dasatinib (solvate form - MeOH, Example 4) is dried in a stirred vacuum drier with suction of N 2 at 55— 65 °C for 1 h.
  • the measurement was performed on a flat powdered sample placed on an Si plate or X-ray holders with a cavity of the diameter 16 mm and height 2.4 mm (so-called back-loading technique) were used.
  • Programmable divergent diaphragms with irradiated area of the sample of 10 mm, Soller diaphragms 0.02 rad, and an anti-scattering diaphragm 1 ⁇ 4 were used for setting the primary optics.
  • a detector X'Celerator with maximum opening of the detection slot, Soller diaphragms 0.02 rad, and an anti-scattering diaphragm 5.0 mm were used for setting the secondary optics.
  • Capillary column CP Sil 5 CB (30 m x 0.32 mm x 3.0 ⁇ ) or equivalent
  • Carrier gas helium for chromatography R; 1.6 ml/min
  • Reference solution 2.5 ml of the basic solution is diluted with ⁇ , ⁇ -dimethylacetamide to 50.0 ml.
  • Reference sample 1.0 ml of the reference solution is pipetted to the 20 ml HS vial and the vial is closed with a cap.
  • Blank experiment 1.0 ml of N,N-dimethylacetamide is pipetted to the 20 ml HS vial and the vial is closed with a cap.
  • Tested sample 20.0 mg of the tested substance is weighed into the 20 ml HS vial, 1.0 ml of N,N-dimethylacetamide is added, and the vial is closed with a cap.
  • This residual solvent was determined by the method of gas chromatography in an instrument Agilent 6890 with direct injection and FI detection.
  • Carrier gas helium for chromatography R; 40 cm/s
  • Re ference solution 125 ⁇ of the basic solution is diluted with pyridine to 25 ml.
  • Blank sample 1.0 ml of pyridine is pipetted to a 2 ml GC vial and the vial is closed with a cap.
  • Reference sample 1.0 ml of the reference solution is pipetted to a 2 ml GC vial and the vial is closed with a cap.
  • Tested sample 50.0 mg of the tested substance are dissolved in 1 ml of pyridine in a 2 ml GC vial and the vial is closed with a cap.
  • A" f peak area of N-methyl-2-pyrrolidone in the chromatogram of the reference sample; m r ...weight of N-methyl-2-pyrrolidone in the basic solution in mg;
  • Mobile phase A 1 ml of HC10 4 R is dissolved in 1000 ml of water R.
  • Test solution 15 mg of the sample are dissolved in 25 ml of the sample solvent, the mixture is placed into an ultrasound bath for 5 minutes, and, after dissolving and cooling down to room temperature, the solution is made up to 50.0 ml with the sample solvent. The sample must be well protected against light!

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

A method for the preparation of the crystalline Form N-6 of dasatinib, characterized by the following reflexions in the X-ray powder pattern, measured with the beam radiation CuKα: 6.8; 12.3; 13.2; 13.8; 16.7; 21.0; 24.3, and 24.8 ± 0.2° 2-theta. The method consists in drying a solvate of dasatinib from the group of the ethanol solvate, methanol solvate, and acetonitrile solvate of dasatinib, or their mixtures.

Description

Method of preparing anhydrous polymorphic Form N-6 of dasatinib Technical Field
The invention relates to a new method of preparing the polymorphic anhydrous form N-6 of dasatinib ; chemically: (N-(2-cbioro-6-methylphenyl)-2- [[6-[4-(2-hydroxyethyl)- 1 - piperazinyl]-2-me1hyl-4-pyrirriidinyl]amino]-5-thiazole carboxamide, CAS: 302962-49-8)), described by the formula (1), and its key solvates, enabling formation of the chemically highly pure Form N-6.
State of the art During the recent decade, the therapy of the chronic myeloid leukaemia (CML) has significantly changed. Great turn in the therapy of CML was the discovery of imatinib (Gleevec), an inhibitor of BCR-ABL tyrosine kinase. Nevertheless, in spite of efficiency of imatinib, a group of patients with CML still remains who are resistant to or do not tolerate therapy with imatinib. It is these patients that the therapy with dasatinib (BMS-354825), an inhibitor of BCR-ABL, Src, and other kinases, can be mdicated for. This drug has been developed by Bristol-Myers Squibb (WO0062778A1, 2000) and sold under the trade name Sprycel.
Figure imgf000002_0001
1
A large number of polymorphic forms of dasatinib are described in the literature, designated as: Hl-7, BU-2, E2-1, N-6, T1H1-7, and T1E2-1. Patent document WO2005/077975 A2 describes the preparation of dasatinib monohydrate (Hl-7) and butanol solvate (BU-2). Another patent document (US2006/0004067A1) linked up with the previous one describes two ethanol solvates (E2-1, T1E2-1) and further two anhydrous forms (N-6, T1H1-7). A large number of preparations of various polymorphic forms are also described in TEVA patent application WO 2009/053854 A2.
Disclosure of Invention The present invention relates to a new method of preparing the anhydrous Form N-6, in particular new use of selected solvate forms (MeOH, MeCN) and their mixtures, which allow conversion to the anhydrous Form N-6 under elevated temperature and vacuum. This phenomenon has also been observed under laboratory conditions, only with a lower conversion rate. From the viewpoint of industrial utilization, this is a very simple, robust, and relatively fast process of preparing the anhydrous Form N-6; its production has also been reproducibly tested in kilogram amounts.
Detailed description of invention
The preparation of the compound of formula (1) includes the following direct steps 1 to 3 (Scheme 1).
Scheme 1 : Synthesis of dasatinib
SteP 1 Molecular Weight: 163,00
Figure imgf000003_0001
Step 2
Figure imgf000004_0001
Molecular Weight: 394,28 Molecular Weight 130,19 Molecu,ar We,9ht: 438·°1
In the first step, 2-ammo-N-(-(2-cMoro-6-memylphenyl)-thiazole-5-carboxamide (4) is reacted with 4,6-dicMoro-2-methylpyrirnidine (5). The reaction is carried out in tetrahydrofuran THF at temperature of 10 - 30°C. Sodium terc-butoxide (NaOtBu) is used as a base. The reaction is completed within 1 hour. The isolated yield ranged between 80 and 83%. Next step is reaction of the (SNAT) intermediate (3) 2-(6-chloro-2-methylpyrimidine-4-ylamino)-N-(2-chloro-6- methylphenyl)thiazole-5-carboxamide with l-(2-hydroxyethyl)-piperazine (2) in an environment of NMP. The reaction runs in the temperature range of 65 to 100°C. Acetonitrile is used as the anti-solvent for isolation of crude dasatinib (1). After isolation, the crude product is dried in vacuo at a temperature of 55 - 65°C. An NMP solvate is obtained with predominant amounts of two residual solvents NMP with an admixture of MeCN. The final step of the synthesis is crystallization from the mixture of solvents MeOH and MeCN at a temperature of 55 - 62°C. The obtained isolated wet crystalline product again shows the solvate structure corresponding to a MeCN solvate (Fig. 2) or a MeOH solvate (Fig. 3). The type to be isolated depends on the selected procedure of preparation and on the ratio of both solvents (examples 3 + 4). The crystallized wet product is further dried in vacuo at a temperature in the range of 55 - 65°C. After drying for 2 - 3 hours, the expected polymorphic Form N-6 is obtained. The duration of drying depends on residual moisture in the sample, type of the drying oven, attained vacuum, and heating rate to the final temperature of drying. Typically, as early as within 1 hour of drying at 55 °C in a shelf drier, majority of the starting MeCN/MeOH solvate has been transformed to the Form N-6 (Fig. 4). The isolated yield ranges between 73 and 77% (crude and crystallized product dasatinib). Total yield of the synthesis is then within the range of 58 - 63%. In addition, it has been verified that the polymorphic Form N-6 can be obtained under reduced vacuum and elevated temperature by drying other solvates, such as EtOH, acetone, IP A, and their mixtures. Similarly, the monohydrate, particularly containing MeCN, MeOH as residual solvents, can be converted to the Form N-6 under reduced pressure and elevated temperature.
The described process is an efficient method of preparing dasatinib that is also convenient for larger scales. The procedure according to Scheme 1 makes it possible to obtain, in a reproducible way, dasatinib (1) characterized by chemical purity higher than 99.8 % and containing individual impurities under 0.1 % (according to UPLC).
Brief Description of Drawings
Figure 1 : X-ray powder pattern of polymorphic anhydrous Form N-6 of dasatinib Figure 2: X-ray powder pattern of the MeCN solvate form of dasatinib
Figure 3: X-ray powder pattern corresponding to the MeOH solvate of dasatinib
Figure 4: Evolution of X-ray powder patterns in the course of conversion of the MeCN solvate form to the Form N-6 during drying in vacuo and at a temperature of 55 °C (shelf drier).
Examples
The following examples serve for a more detailed illustration of the method of producing dasatinib of the polymorphic anhydrous Form N-6 according to the present invention on the industrial scale. Example 1
The preparation of the intermediate (3) (2-(6-chloro-2-methylpyrimidine-4-ylamino)-N-(2- cUoro-6-methylphenyl)thiazole-5-carboxamide)
In a chemical reactor, 2-ammo-N-(-(2-chloro-6-methylphenyl)-thiazole-5-carboxamide (4) (3.750 kg) and 2,4-dichloro-2-methyIpyrimidine (5) (2.889 kg) are suspended in 50.0 1 of THF. The suspension is stirred until dissolved at 25 - 30°C; then it is cooled down to 10°C and a pre-prepared solution of 4.444 kg of NaOt-Bu in 25.0 1 of THF is added dropwise under N2. The solution of NaOt-Bu is added dropwise under N2 within 15 - 25 min (intensive stirring). Finally, the separating funnel is rinsed with 1.5 1 of THF. During the dropwise addition, the temperature of the reaction mixture is kept between 15 and 30°C. After the dosing is completed, the reaction mixture is stirred at 20 ± 2°C for 60 min. The reaction mixture is cooled to 10 - 15°C, 12.5 1 of demineralized water is added dropwise, and the mixture is stirred for 15 min. Then, its temperature is brought to 5°C and diluted 1.5 M HCl (2,550 ml of 35 % HCl + 16.5 1 of demi water) is added dropwise within about 35 - 45 min. The amount of the HCl solution is chosen in order to reach final pH of 4.0 - 6.5 (optimally 5.0 - 5.5). The stirring goes on at 5 - 8°C for 15 - 20 min (checking of pH, adjusted, as required). 22.5 1 of acetone is added, the mixture is heated to 40°C, and stirred at this temperature for 30 minutes. Then it is cooled down slowly to 10 - 15°C and stirred for 1.5 h. The separated product is centrifuged, washed with a warm (40 °C) 1 :1 mixture of acetone / demi water (1 x 5.0 1) and then with cold acetone (1 x 5.0 1). The product is dried in a vacuum drier at 55 - 65°C for 12 hours.
Yield: 4,610 g (83.5%), sum of impurities according to PLC: 1.4%; loss on drying: 0.3%.
Example 2
Preparation of crude dasatinib (NMP solvate)
In a chemical reactor, dasatinib intermediate (3), CAS: 302964-08-5 (4.500 kg) is suspended in 18.0 I of NMP. The suspension is heated to a temperature of 67 - 68°C. A solution of HEPIP (7.313 kg of HEPIP + 3.8 1 of NMP) is prepared and added dropwise to the reaction mixture within 5 - 10 min. During the addition the temperature is kept at 65 ± 3°C. After the addition is completed, the mixture is stirred at 65°C for 60 min. Then, temperature is brought to 100°C and stirring goes on for 20 min. All the time, the reaction is carried out under N2 and access of light is prevented! After the reaction is completed, the reaction mixture is cooled down to 40 - 45°C and filtered. The filter is washed with 1.0 1 NMP. The filtered reaction mixture is added dropwise to MeCN (113 1), heated at 25 - 30°C, within 15 - 20 min. Stirring continues at 25 - 30°C for 30 min. The mixture is heated to 45 - 50°C and stirred for 0.5 h. Then, is slowly cooled down to 10 - 15°C within 75 - 90 min and stirred for 1.0 - 1.5 h. The separated product is centrifuged, and washed with cold (2 - 8°C) MeCN (2 x 6.0 1). It is dried in a vacuum drier at 60°C for 12 hours. Yield: 82%, sum of impurities according to UPLC: 0.2%; content of residual solvents: 30,000 ppm NMP, 17,000 ppm CH3OH; X-ray powder pattern corresponds to the NMP solvate.
Example 3 Preparation of crystallized dasatinib - wet (MeCN solvate)
The crude dasatinib (1.00 kg) is suspended in 10.5 1 of MeCN and 17.5 1 of MeOH. The suspension is heated up to 58 - 62°C and stirred for 0.5 h. Then, it is cooled down to 40 - 45°C and stirred for 1 hour. Cooling is continued to -5 to 0°C and stirred for 1.0 - 1.5 h. The separated product is centrifuged, and washed with cold MeCN (2 x 3.0 1). All the time, the crystallization is carried out under the inert atmosphere of N2 and access of light is prevented! Drying is carried out as in Example 5. The X-ray pattern of the wet sample corresponds to the MeCN solvate. The MeCN solvate is characterized by the reflexions listed in Table 1:
Table 1: MeCN solvate
Position Inter-planar Rel. int.
[°2 Th.] distance [A] [%]
5.66 15.602 100.0
5.86 15.080 81.5
11.77 7.514 10.5
12.25 7.222 36.9
14.86 5.958 27.2
16.05 5.518 16.6
17.11 5.179 8.7
18.53 4.785 5.7
19.11 4.642 4.8
21.63 4.106 7.4
22.13 4.013 12.0
22,97 3.869 7.3
24.67 3.606 13.0
25.34 3.511 15.5 Example 4
Preparation of crystallized dasatinib - wet (MeOH solvate)
The crude dasatinib (1.00 kg) is suspended in 5.0 1 of MeCN and 25 1 of MeOH. The suspension is heated up to 58 - 62°C and stirred for 0.5 h. Then, it is cooled down to 30 - 35°C and stirred for 1 hour. Cooling is continued to -5 to 0 °C and stirred for 1.0 - 1.5 h. The separated product is centnfuged, and washed with cold MeCN (2 x 3.0 1). All the time, the crystallization is carried out under the inert atmosphere of N2 and access of light is prevented! Drying is carried out as in Example 6. The X-ray pattern of the wet sample corresponds to the MeOH solvate and is characterized by the reflexions listed in Table 2: Table 2: MeOH solvate
Position Inter-planar Rel. int.
[°2 Th.] distance [A] [%]
7.36 11.994 74.4
12.20 7.252 21.5
13.43 6.585 19.7
14.58 6.069 36.7
16.39 5.404 100.0
16.99 5.215 15.1
19.02 4.662 17.8
19.44 4.562 13.0
20.86 4.256 5.3
21.95 4.047 11.6
24.54 3.624 45.0
25.44 3.498 28.6
30.94 2.888 9.9 Example 5
Preparation of crystallized dasatinib - dry (anhydrous Form N-6)
The wet crystalline dasatinib (solvate form - MeCN, Example 3) is dried in a vacuum tray drier with suction of N2 at 55 - 65 °C for 16 h. Yield: 91% (from crude API); sum of impurities according to UPLC: 0.11%, content of residual solvents less than 500 ppm, content of water according to KF 0.5%; X-ray pattern of the dry sample corresponds to the pure anhydrous Form N-6.
Example 6 Preparation of crystallized dasatinib - dry (anhydrous Form N-6)
The wet crystallized dasatinib (solvate form - MeOH, Example 4) is dried in a stirred vacuum drier with suction of N2 at 55— 65 °C for 1 h.
Yield: 85% (from crude API); sum of impurities according to UPLC: 0.13%, content of residual solvents less than 1500 ppm, content of water according to KF 0.7%; X-ray pattern of the dry sample corresponds to the pure anhydrous Form N-6.
Overview of analytical methods
The samples mentioned in the previous examples were characterized by the method of X-ray powder diffraction. The amounts of residual solvents were determined by GC. Chemical purity of the samples was determined by means of UPLC.
The measured parameters of the X-ray powder diffraction: Diffraction patterns were measured in a diffractometer X'PERT PRO MPD PANalytical, X-ray beam CuKa (λ = 1.542 A), excitation voltage: 45 kV, anodic current: 40 mA, measured range: 2 - 40° 2Θ, step size: 0.01° 2Θ. The measurement was performed on a flat powdered sample placed on an Si plate or X-ray holders with a cavity of the diameter 16 mm and height 2.4 mm (so-called back-loading technique) were used. Programmable divergent diaphragms with irradiated area of the sample of 10 mm, Soller diaphragms 0.02 rad, and an anti-scattering diaphragm ¼ were used for setting the primary optics. A detector X'Celerator with maximum opening of the detection slot, Soller diaphragms 0.02 rad, and an anti-scattering diaphragm 5.0 mm were used for setting the secondary optics.
Method A: The following solvents are monitored: methanol ethanoU acetonitrile
These residual solvents were determined using the method of head-space gas chromatography in an instrument PerkinElmer Autosystem XL with FI detection and a head-space autosampler TurboMatrix 40.
Chromatographic conditions:
Capillary column: CP Sil 5 CB (30 m x 0.32 mm x 3.0 μηι) or equivalent
Temperature program: 40°C - 3 min, gradient 20°C/min to 160°C - 2 min
Carrier gas: helium for chromatography R; 1.6 ml/min
Injector: splitting flow rate 4.0 ml/min, 165°C
Detector: FID, 300 °C, Range: 1, Attn: -3
Conditions ofHS autosampler:
Oven temperature: 110°C
Needle temperature: 115°C
Transfer line temperature: 120°C
Time at temperature: 30.0 min
Time of pressurization: 1,0 min
Time of injection: 0.05 min
Time of rinsing: 0.2 min
Pressure: 21 psi
GC cycle: 20 min
Preparation of solutions:
Basic solution:
Methanol lOO mg
Ethanol lOO mg
Acetonitrile 100 mg
Acetone 100 mg
Dichloromethane 100 mg These weights are dissolved in N,N-diraethylacetamide and the solution is made up to 25.0 ml with this solvent.
Reference solution: 2.5 ml of the basic solution is diluted with Ν,Ν-dimethylacetamide to 50.0 ml.
Reference sample: 1.0 ml of the reference solution is pipetted to the 20 ml HS vial and the vial is closed with a cap.
Blank experiment: 1.0 ml of N,N-dimethylacetamide is pipetted to the 20 ml HS vial and the vial is closed with a cap.
Tested sample: 20.0 mg of the tested substance is weighed into the 20 ml HS vial, 1.0 ml of N,N-dimethylacetamide is added, and the vial is closed with a cap.
Test of performance of the chromatographic system:
- Repeatability: relative standard deviation of areas of the peaks of the given residual solvent - max. 15.0 %, calculated from five consecutive injections of the reference sample;
- Resolution: at least 1.5 between the peaks of individual residual solvents in the fifth injection of the reference sample.
Evaluation:
The contents of the residual solvents expressed in % are evaluated by the method of external standard according to the formula: x = x 0,2 , wherein
Figure imgf000011_0001
Af ... peak area of the individual residual solvent in the chromatogram of the tested sample; Ar ... peak area of the individual residual solvent in the chromatogram of the reference sample;
.weight of the individual residual solvent in the basic solution
mf ...weight of the tested sample in mg. Method B: l-Methyl-2-pyrrolidinone ΓΝΜΡ') is monitored
This residual solvent was determined by the method of gas chromatography in an instrument Agilent 6890 with direct injection and FI detection.
Chromatographic conditions:
Capillary column: Rtx-5 Amine (30 m χ 0.53 mm χ 3.0 μιη) or equivalent
Temperature program: 115°C - 0 min, gradient 15°C/min to 235°C - 0 min
Carrier gas: helium for chromatography R; 40 cm/s
Injector: splitting ratio 1: 2, 220°C
Injection: 1 μΐ
Detector: FID, 290°C
Preparation of solutions:
Basic solution: 53.0 mg of the reference substance N-methyl-2-pyrrolidone and 150.0 mg of the reference substance l-(2-hydroxyethyl)piperazine are dissolved in pyridine in a 10 ml volumetric flask and made up with pyridine to the mark.
Re ference solution: 125 μΐ of the basic solution is diluted with pyridine to 25 ml.
Blank sample: 1.0 ml of pyridine is pipetted to a 2 ml GC vial and the vial is closed with a cap.
Reference sample: 1.0 ml of the reference solution is pipetted to a 2 ml GC vial and the vial is closed with a cap.
Tested sample: 50.0 mg of the tested substance are dissolved in 1 ml of pyridine in a 2 ml GC vial and the vial is closed with a cap.
Orientation retention times:
N-methyl-2-pyrrolidone 4.5 min.
l-(2-hydroxyethyl)piperazine 5.6 min.
Test of performance of the chromatographic system:
- Repeatability: relative standard deviation of peak areas of N-methyl-2-pyrrolidone - at most 15.0%, calculated from five consecutive injections of the reference sample;
- Resolution: at least 10 between peaks of N-methyl-2-pyrrolidone and 1 -(2-hydroxyethyl) piperazine in the fifth injection of the reference sample. Evaluation:
The content of N-methyl-2-pyrroHdone expressed in ppm is evaluated by the method of external standard according to the formula:
A'esl ■ mref
x = - — r x °'05. wherein
Λχ ' mx
A* 51 ...peak area of N-methyl-2-pyrrolidone in the chromatogram of the tested sample;
A"f ... peak area of N-methyl-2-pyrrolidone in the chromatogram of the reference sample; mr ...weight of N-methyl-2-pyrrolidone in the basic solution in mg;
m' ...weight of the tested sample in mg
Method C: Impurities in the product are monitored
ChromatosraOhic conditions:
Instrument: UPLC UHPLC with UV/VIS or PDA detector
Column:
Stationary phase: ACQUITY CSH Phenyl-Hexyl (1.7 μιη)
Size: 1 = 0.100 m, 0 3.0 mm
Temperature: 35°C
Mobile phase A: 1 ml of HC104 R is dissolved in 1000 ml of water R.
Mobile phase B: Acetonitrile Rl Elution gradient:
Time Mobile phase A Mobile phase B
[min] [% V VJ [% V/VJ
0 90 10
10 50 50
11 50 50
12 90 10
Flow rate: 0.5 ml/min
Detection: UV 220 nm
Injection: 1.0 Temperature of autosampler 15°C
Time of analysis: 12 min
Delay of next injection: 2 min
Orientation retention time: "dasati ib " about 5.6 min
Relative retention time: (with reference to "dasatinib ")
Impurity "DAS 4" RRT 0.72 Impurity "DAS 6" RRT 0.93 Impurity "DAS 1" RRT 0.97 Impurity "DAS 3" RRT 1.07 Impurity "DAS 5" RRT 1.82
Sample solvent: Acetonitrile l / Water R 50/50 (V/V)
Test solution: 15 mg of the sample are dissolved in 25 ml of the sample solvent, the mixture is placed into an ultrasound bath for 5 minutes, and, after dissolving and cooling down to room temperature, the solution is made up to 50.0 ml with the sample solvent. The sample must be well protected against light!
Evaluation: The content of individual impurities is evaluated by the method of internal normalization:
Limits:
- Impurity DAS 1 NMT 0.15 %
- Impurity DAS 3 NMT 0.15 %
- Impurity DAS 4 NMT 0.15
- Impurity DAS 5 NMT 0.15 %
- Impurity DAS 6 NMT 0.15 %
- Any other impurity NMT 0.10 %
Water (2.5.32): Microdetermination: max. 2.0%, determination from 0.100 g.

Claims

1. A method of preparing the crystalline Form N-6 of dasatinib, characterized by the following reflexions in the X-ray powder pattern, measured with the beam radiation CuKa: 6.8; 12.3; 13.2; 13.8; 16.7; 21.0; 24.3, and 24.8 ± 0.2° 2-theta,
the method consisting in drying a dasatinib solvate from the group of the ethanol solvate, methanol solvate, and acetonitrile solvate of dasatinib, or their mixtures.
2. The method according to Claim 1, characterized in that the drying is carried out in vacuo at a temperature in the range of 55 - 65°C for at least 2 hours.
PCT/CZ2014/000160 2013-12-19 2014-12-18 Method of preparing anhydrous polymorphic form n-6 of dasatinib WO2015090259A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CZ2013-1046A CZ306732B6 (en) 2013-12-19 2013-12-19 A method of preparation of the anhydrous polymorphic form of N-6 Dasatinib
CZPV2013-1046 2013-12-19

Publications (1)

Publication Number Publication Date
WO2015090259A1 true WO2015090259A1 (en) 2015-06-25

Family

ID=52446175

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CZ2014/000160 WO2015090259A1 (en) 2013-12-19 2014-12-18 Method of preparing anhydrous polymorphic form n-6 of dasatinib

Country Status (2)

Country Link
CZ (1) CZ306732B6 (en)
WO (1) WO2015090259A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106117196A (en) * 2016-06-09 2016-11-16 青岛辰达生物科技有限公司 A kind of method preparing cancer therapy drug Dasatinib
WO2017087818A1 (en) * 2015-11-19 2017-05-26 The Regents Of The University Of Michigan Dual src/p38 kinase inhibitor compounds and their use as therapeutic agents
WO2018100585A1 (en) * 2016-12-01 2018-06-07 Natco Pharma Limited An improved process for the preparation of dasatinib polymorph
US10940149B1 (en) 2018-06-15 2021-03-09 Handa Oncology, Llc Kinase inhibitor salts and compositions thereof

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000062778A1 (en) 1999-04-15 2000-10-26 Bristol-Myers Squibb Co. Cyclic protein tyrosine kinase inhibitors
WO2005077945A2 (en) * 2004-02-06 2005-08-25 Bristol-Myers Squibb Company Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
WO2005077975A2 (en) 2004-02-10 2005-08-25 Cyclacel Limited Polypeptides comprising the pleckstrin homology domain of ect2 and their binding specificities to phosphatidylinositols
US20060004067A1 (en) 2004-02-06 2006-01-05 Bang-Chi Chen Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
WO2009053854A2 (en) 2007-10-23 2009-04-30 Teva Pharmaceutical Industries Ltd. Polymorphs of dasatinib and process for preparation thereof
WO2014086326A1 (en) * 2012-12-06 2014-06-12 Zentiva, K.S. A method for the preparation and purification of new and known polymorphs and solvates of dasatinib

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102643275B (en) * 2011-02-21 2016-04-20 江苏先声药物研究有限公司 The preparation method that a kind of Dasatinib N-6 crystal formation is new

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000062778A1 (en) 1999-04-15 2000-10-26 Bristol-Myers Squibb Co. Cyclic protein tyrosine kinase inhibitors
WO2005077945A2 (en) * 2004-02-06 2005-08-25 Bristol-Myers Squibb Company Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
US20060004067A1 (en) 2004-02-06 2006-01-05 Bang-Chi Chen Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
WO2005077975A2 (en) 2004-02-10 2005-08-25 Cyclacel Limited Polypeptides comprising the pleckstrin homology domain of ect2 and their binding specificities to phosphatidylinositols
WO2009053854A2 (en) 2007-10-23 2009-04-30 Teva Pharmaceutical Industries Ltd. Polymorphs of dasatinib and process for preparation thereof
WO2014086326A1 (en) * 2012-12-06 2014-06-12 Zentiva, K.S. A method for the preparation and purification of new and known polymorphs and solvates of dasatinib

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017087818A1 (en) * 2015-11-19 2017-05-26 The Regents Of The University Of Michigan Dual src/p38 kinase inhibitor compounds and their use as therapeutic agents
CN106117196A (en) * 2016-06-09 2016-11-16 青岛辰达生物科技有限公司 A kind of method preparing cancer therapy drug Dasatinib
WO2018100585A1 (en) * 2016-12-01 2018-06-07 Natco Pharma Limited An improved process for the preparation of dasatinib polymorph
US10800771B2 (en) 2016-12-01 2020-10-13 Natco Pharma Limited Process for the preparation of dasatinib polymorph
US10940149B1 (en) 2018-06-15 2021-03-09 Handa Oncology, Llc Kinase inhibitor salts and compositions thereof
US11007195B2 (en) 2018-06-15 2021-05-18 Handa Oncology, Llc Kinase inhibitor salts, and compositions thereof
US11052088B2 (en) 2018-06-15 2021-07-06 Handa Oncology, Llc Kinase inhibitor salts, and compositions thereof
US11160805B2 (en) 2018-06-15 2021-11-02 Handa Onocology, Llc Kinase inhibitor salts and compositions thereof

Also Published As

Publication number Publication date
CZ306732B6 (en) 2017-05-31
CZ20131046A3 (en) 2015-07-01

Similar Documents

Publication Publication Date Title
EP3231805B1 (en) Salt of pyrrolo[2,3-d]pyrimidine compound and novel polymorph of salt
US9505744B2 (en) Solid state forms of vilazodone and vilazodone hydrochloride
CA2875877A1 (en) Syk inhibitors
US20220235039A1 (en) Crystalline salt forms of 6-(cyclopropanecarboxamido)-4-((2-methoxy-3-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)amino)-n-(methyld3) pyridazine-3-carboxamide
WO2015090259A1 (en) Method of preparing anhydrous polymorphic form n-6 of dasatinib
KR20210054546A (en) Salts and crystal forms of GABAA positive allosteric modulators
CA2928287A1 (en) Solid form of axitinib
CN103360391A (en) Novel apixaban crystal form and preparation method thereof
JP2023089169A (en) Synthesis method of furoimidazopyridine compound, crystal form of furoimidazopyridine compound, and crystal form of salt thereof
CN103804357A (en) Rupatadine fumarate compound as well as synthesis method and pharmaceutical composition thereof
AU2021201177B2 (en) Processes for preparing an FGFR inhibitor
WO2018113801A1 (en) Crystalline forms of2-[1-ethylsulfonyl-3-[4-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)pyrazol-1-yl]azetidin-3-yl]acetonitrile with phosphoric acid and a method of their preparation
EP3981398A1 (en) Method for synthesizing furoimidazopyridine compound, polymorphic substance and polymorphic substance of salt
CN110305131B (en) Novel crystal form of linagliptin and preparation method thereof
US10138250B2 (en) Salt of pyrrolo[2,3-D]pyrimidine compound and novel polymorph of salt
CZ2016705A3 (en) Crystalline forms of 2-[1-ethylsulfonyl-3-[4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)pyrazol-1yl]azetidin-3yl] of acetonitrile salts and their preparation
WO2023086320A1 (en) Forms and compositions of inhibitors of jak2
CN108026087A (en) (4- ((3R, 4R) -3- methoxyl groups ttetrahydro-pyran -4- bases amino) piperidin-1-yl) (5- methyl -6- (((2R, 6S) -6- (p- tolyl) tetrahydrochysene -2H- pyrans -2- bases) methylamino) pyrimidine-4-yl) methanone citrate salt
CN105560246B (en) The preparation and its pharmaceutical composition of imatinib mesylate α crystallization
CA3080827A1 (en) Salt form and crystal form of compound as fgfr4 inhibitor and preparation method thereof
BR112020006051A2 (en) crystalline forms of lenalidomide
CZ31155U1 (en) A cocrystal of a crystal form of baricitinib and coformer
EP4230625A1 (en) Crystal form of multi-substituted benzene ring compound maleate, and preparation method therefor and use thereof
US11440911B2 (en) Salts and crystals
WO2017093773A1 (en) New polymorphic and solvate form of idelalisib

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: 14833498

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14833498

Country of ref document: EP

Kind code of ref document: A1