WO2017108605A1 - Pharmaceutical composition comprising amorphous dasatinib - Google Patents

Pharmaceutical composition comprising amorphous dasatinib Download PDF

Info

Publication number
WO2017108605A1
WO2017108605A1 PCT/EP2016/081369 EP2016081369W WO2017108605A1 WO 2017108605 A1 WO2017108605 A1 WO 2017108605A1 EP 2016081369 W EP2016081369 W EP 2016081369W WO 2017108605 A1 WO2017108605 A1 WO 2017108605A1
Authority
WO
WIPO (PCT)
Prior art keywords
dasatinib
tablet composition
composition according
polymer
acid
Prior art date
Application number
PCT/EP2016/081369
Other languages
French (fr)
Inventor
Sonia GARCIA JIMENEZ
Lisardo Alvarez Fernandez
Jose VELADA CALZADA
Original Assignee
Synthon B.V.
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 Synthon B.V. filed Critical Synthon B.V.
Publication of WO2017108605A1 publication Critical patent/WO2017108605A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/141Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers
    • A61K9/146Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers with organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • A61K9/2077Tablets comprising drug-containing microparticles in a substantial amount of supporting matrix; Multiparticulate tablets
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Definitions

  • CML chronic myelogenous leukaemia
  • ALL Philadelphia chromosome- positive acute lymphoblastic leukaemia
  • Ph+ Philadelphia chromosome positive chronic myelogenous leukaemia
  • Dasatinib was discovered by Bristol-Myers Squibb and is disclosed in EP1169038. Dasatinib is the active ingredient in the medicinal product sold under the brand name Sprycel ® .
  • WO2005077945 discloses a crystalline monohydrate of dasatinib and a butanol solvate of dasatinib.
  • the marketed product Sprycel ® contains the crystalline monohydrate of dasatinib.
  • WO2005077945 further discloses a crystalline ethanol solvate and two anhydrous forms of dasatinib.
  • Other forms of dasatinib are disclosed in WO2009053854, WO2010062715, WO2010067374, WO2011095059 and WO2012014149. Some of the described forms do contain unwanted solvents. Moreover, it was experienced in our laboratory that, especially under humid conditions, some of these forms are rather unstable.
  • Dasatinib monohydrate is a BCS class II compound, exhibiting low solubility and high permeability. Its low aqueous solubility affects the dissolution behavior of dasatinib monohydrate negatively.
  • solubility of amorphous forms is higher compared to the solubility of crystalline forms, thus it would be desirable to have dasatinib available in amorphous form.
  • WO2009053854 provides processes to prepare amorphous dasatinib.
  • WO2015049645 discloses processes to prepare several hydrated forms of amorphous dasatinib.
  • CN104327067 provides a process to prepare amorphous dasatinib by dissolving the compound in a solvent/co-solvent system followed by spray-drying.
  • amorphous dasatinib as such is not stable and therefore not suitable for use on pharmaceutical production scale.
  • WO2013105894 and WO2013105895 disclose pharmaceutical compositions comprising stable amorphous hybrid nanoparticles comprising dasatinib and a polymeric stabilizing and matrix-forming component and processes to prepare them.
  • the process to obtain said particles involves the provision and mixing of several pressurized streams. Such a treatment could easily affect the physical stability of dasatinib and is moreover rather costly.
  • CN 104367557 discloses a process to prepare solid dispersions comprising dasatinib and polyvinylpyrrolidone by dissolving dasatinib and the polymer in a mixture of methanol and water followed by spray drying. Since the solubility of dasatinib in this solvent mixture is low, large amounts of solvent are used. The disclosed process is therefore not suitable for production on commercial scale.
  • the present invention relates to a tablet composition
  • a solid dispersion of dasatinib and a polymer wherein the composition is obtained by:
  • polymer is selected from polyvinylpyrrolidone, copovidone and hydroxypropyl cellulose.
  • Said tablet composition may be used as a medicament, particularly in the treatment of chronic myeloid leukaemia (CML) and acute lymphoblastic leukaemia (ALL).
  • CML chronic myeloid leukaemia
  • ALL acute lymphoblastic leukaemia
  • Sprycel ® contains the monohydrated form of dasatinib as disclosed in WO2005077945, the so-called form "Hl-7".
  • Dasatinib monohydrate is a very stable compound, but being a BCS class II compound, it exhibits low aqueous solubility which affects dissolution behavior.
  • dasatinib has been disclosed in the prior art. Many of these forms are solvated forms of dasatinib and some of them do contain unwanted (e.g. toxic) solvents. Moreover, some of the forms as disclosed in the prior art are rather unstable.
  • solid dispersion has been defined as a dispersion of one or more Active Pharmaceutical Ingredients (APIs) in an inert carrier or matrix at the solid state, prepared by a solvent or melting process or a combination of the two.
  • APIs Active Pharmaceutical Ingredients
  • the solid dispersions are divided into crystalline solid dispersions and amorphous solid dispersions respectively.
  • Amorphous carriers used are mostly polymers.
  • the API is dispersed in very small size and exists in supersaturated state in amorphous carriers because of forced degradation.
  • the amorphous carriers can increase the wettability and dispersibility of drugs as well as inhibit the precipitation process of drugs when amorphous solid dispersions are dissolved in water. These properties along with the fast dissolution rate of amorphous carriers due to the low thermodynamic stability of amorphous state carriers, enhance the drug solubility and release rate.
  • the number of marketed products arising from solid dispersion approaches is very low. This low number is mainly due to scale-up problems and physicochemical instability in the manufacturing process or during storage leading to phase separation and crystallization (Vo et. al, Eur. J. Pharm. Biopharm., 85 (2013) 799-813).
  • the process selected to prepare the tablet compositions of the present invention is the solvent evaporation method, because in this method problems related to decomposition, as frequently occurring in the melting method, are prevented.
  • An important prerequisite of the solvent evaporation method is the sufficient solubility of the drug and the carrier in the solvent system. Finding a suitable non-toxic solvent is sometimes difficult because carriers are hydrophilic whereas drugs are hydrophobic.
  • the present invention provides a tablet composition comprising a solid dispersion of dasatinib and a polymer, wherein the composition is obtained by:
  • the polymers to be used in accordance with the present invention are selected from polyvinylpyrrolidone (PVP), copovidone and hydroxypropyl cellulose.
  • PVP polyvinylpyrrolidone
  • Commonly used polymers like polyethylene glycol, Eudragit ® and hydroxypropyl methylcellulose were found to be unsuitable to be used in the present invention for various reasons. Some of the polymers did not result in fully amorphous solid dispersions, while other polymers did give rise to amorphous solid dispersions but wherein dasatinib started to crystallize over time. Moreover, some polymers were found to be unsuitable to be used in accordance with the present invention due to their solubility/gelling behavior in the solvent system.
  • Polyvinylpyrrolidone is a particularly preferred polymer to be used in accordance with the present invention.
  • the weight ratio of dasatinib to polymer in the solid dispersion ranges from about 1: 1 to about 1:6, preferably from 1: 1 to 1:3.
  • the tablet composition of the present invention is obtained by dissolving, in the first step of the process, dasatinib and the polymer in a solvent mixture.
  • the non-toxic solvent mixture in accordance with the present invention comprises water, an alcohol and at least one molar equivalent of acid with respect to dasatinib. Both dasatinib and the polymer dissolve well in the solvent mixture upon heating at a temperature ranging from 45 to 70°C.
  • the acid used in accordance with the present invention is a mineral acid.
  • the acid is hydrochloric acid.
  • the amount of acid present in the solvent mixture may range. However, at molar ratios of acid to dasatinib below 1: 1, dissolution is insufficient, while at too high ratios, gelling was observed.
  • the molar ratio of acid to dasatinib is ranging from 1: 1 to 3: 1. More preferably, the molar ratio of acid to dasatinib is ranging from 1: 1 to 2: 1.
  • the alcohol used in the solvent mixture of the present invention is selected from methanol, ethanol and isopropanol. Ethanol is a particularly preferred alcohol.
  • the volume ratio water to alcohol in the solvent mixture may vary, but is preferably ranging from 2:3 to 1:4. At these ratios, optimal solubility is achieved.
  • the resulting solution is added to a diluent.
  • the addition of the solution to the diluent is performed by spraying the solution over the diluent.
  • the diluent to be used in accordance with the present invention may be any diluent known to a person of ordinary skill in the art.
  • the diluent to be used in accordance with the present invention is an inorganic diluent, polysaccharide, mono- or disaccharide or sugar alcohol.
  • Microcrystalline cellulose is a particularly preferred diluent.
  • the solvent is evaporated.
  • the evaporation is carried out by techniques known to a person of ordinary skill in the art.
  • the resulting blend is then mixed with further excipients.
  • the tablet compositions according to the present invention comprise, besides a diluent, further pharmaceutically acceptable excipients.
  • the excipients to be used in accordance with the present invention are well4inown and are those excipients which are conventionally used by the person skilled in the art.
  • the pharmaceutically acceptable excipients are chosen from one or more binders, disintegrants, glidants or lubricants. Most preferably, the further excipients are chosen from one or more disintegrants and one or more lubricants.
  • the disintegrant to be used in accordance with the present invention may be any disintegrant known to a person of ordinary skill in the art. Suitable disintegrants to be used in accordance with the present invention are selected from the group consisting of
  • croscarmellose sodium is a particularly preferred disintegrant.
  • the lubricant to be used in accordance with the present invention may be any lubricant known to a person of ordinary skill in the art. Magnesium stearate is a particularly preferred lubricant.
  • the final blend is compressed into tablets, using equipment and methods well-known in the art.
  • the tablet composition of the present invention exhibits excellent long term stability. Even after 3 months at 40°C/75% RH, no conversion into any crystalline form of dasatinib was observed. Moreover, the tablet composition of the present invention is cost effective and very suitable for production on commercial scale.
  • the tablet composition of the present invention display dissolution behavior typical for immediate-release formulations.
  • the composition of the present invention exhibits a dissolution rate of at least 85% in 15-30 minutes when tested in 500 ml 0.01 N hydrochloric acid pH 2.0 in a USP apparatus II at 75 rpm, 25°C.
  • the tablet composition in accordance with the present invention may be used as a medicament.
  • the pharmaceutical composition typically may be used in the treatment of chronic myeloid leukaemia (CML) and acute lymphoblastic leukaemia (ALL).
  • CML chronic myeloid leukaemia
  • ALL acute lymphoblastic leukaemia
  • the tablets comprising a solid dispersion of dasatinib and a polymer were prepared by wet granulation and have the composition as given in table 1.
  • Table 1 tablet composition of tablets comprising a solid dispersion of dasatinib and a polymer
  • microcrystalline cellulose in a fluid bed dryer.
  • the solvent was evaporated by using a top spraying/drying process. During spraying/drying the temperature was adjusted to 45-80°C, depending on the polymer used.
  • the resulting blend was sieved through an appropriate mesh size sieve. 14.00 g of croscarmellose sodium was sieved through a suitable mesh size sieve for deagglomeration and mixed with the blend.
  • Magnesium stearate was sieved through an appropriate sieve to deagglomerate and mixed with the powder mix.
  • the homogeneous powder obtained, was compressed using a rotating tablet press using appropriate punches. The tablets were stored at 40°C/75% RH (open dish). Stability results:

Abstract

The present invention relates to a tablet composition comprising a solid dispersion of dasatinib and a polymer, wherein the composition is obtained by: (1) Dissolving dasatinib and the polymer in a solvent mixture comprising water, an alcohol and at least one molar equivalent of acid with respect to dasatinib at a temperature ranging from 45 to 70ºC; (2) Adding the resulting solution to a diluent; (3) Evaporating the solvent; (4) Mixing the resulting blend with further excipients; (5) Compressing the final blend into tablets, what wherein the polymer is selected from polyvinylpyrrolidone, copovidone and hydroxypropyl cellulose. The invention further relates to the use of said tablet composition as a medicament, particularly in the treatment of chronic myeloid leukaemia (CML) and acute lymphoblastic leukaemia (ALL).

Description

PHARMACEUTICAL COMPOSITION COMPRISING AMORPHOUS
DASATINIB BACKGROUND OF THE PRESENT INVENTION
Dasatinib, chemically N-(2-chloro-6-methylphenyl)-2-[[6-[4-(2-hydroxyethyl)-l- piperazinyl]-2-methyl-4-pyrimidinyl] amino] -5-thiazolecarboxamide of formula (I),
Figure imgf000002_0001
is a pharmaceutically active compound used for the treatment of adult patients with chronic myelogenous leukaemia (CML) after imatinib treatment, Philadelphia chromosome- positive acute lymphoblastic leukaemia (ALL) and newly diagnosed Philadelphia chromosome positive (Ph+) chronic myelogenous leukaemia (CML) in the chronic phase.
Dasatinib was discovered by Bristol-Myers Squibb and is disclosed in EP1169038. Dasatinib is the active ingredient in the medicinal product sold under the brand name Sprycel®.
Several crystalline forms of dasatinib are known and described in literature.
WO2005077945 discloses a crystalline monohydrate of dasatinib and a butanol solvate of dasatinib. The marketed product Sprycel® contains the crystalline monohydrate of dasatinib. WO2005077945 further discloses a crystalline ethanol solvate and two anhydrous forms of dasatinib. Other forms of dasatinib are disclosed in WO2009053854, WO2010062715, WO2010067374, WO2011095059 and WO2012014149. Some of the described forms do contain unwanted solvents. Moreover, it was experienced in our laboratory that, especially under humid conditions, some of these forms are rather unstable.
Dasatinib monohydrate is a BCS class II compound, exhibiting low solubility and high permeability. Its low aqueous solubility affects the dissolution behavior of dasatinib monohydrate negatively.
The solubility of amorphous forms is higher compared to the solubility of crystalline forms, thus it would be desirable to have dasatinib available in amorphous form.
WO2009053854 provides processes to prepare amorphous dasatinib. WO2015049645 discloses processes to prepare several hydrated forms of amorphous dasatinib. CN104327067 provides a process to prepare amorphous dasatinib by dissolving the compound in a solvent/co-solvent system followed by spray-drying. However, amorphous dasatinib as such is not stable and therefore not suitable for use on pharmaceutical production scale.
WO2013105894 and WO2013105895 disclose pharmaceutical compositions comprising stable amorphous hybrid nanoparticles comprising dasatinib and a polymeric stabilizing and matrix-forming component and processes to prepare them. The process to obtain said particles, involves the provision and mixing of several pressurized streams. Such a treatment could easily affect the physical stability of dasatinib and is moreover rather costly.
CN 104367557 discloses a process to prepare solid dispersions comprising dasatinib and polyvinylpyrrolidone by dissolving dasatinib and the polymer in a mixture of methanol and water followed by spray drying. Since the solubility of dasatinib in this solvent mixture is low, large amounts of solvent are used. The disclosed process is therefore not suitable for production on commercial scale.
Thus in view of the prior art cited above, there is still a need for stable pharmaceutical compositions with adequate dissolution comprising dasatinib, which are suitable for production on commercial scale. BRIEF DESCRIPTION OF THE PRESENT INVENTION
The present invention relates to a tablet composition comprising a solid dispersion of dasatinib and a polymer, wherein the composition is obtained by:
(1) Dissolving dasatinib and the polymer in a solvent mixture comprising water, an alcohol and at least one molar equivalent of acid with respect to dasatinib at a temperature ranging from 45 to 70°C;
(2) Adding the resulting solution to a diluent;
(3) Evaporating the solvent;
(4) Mixing the resulting blend with further excipients;
(5) Compressing the final blend into tablets,
wherein the polymer is selected from polyvinylpyrrolidone, copovidone and hydroxypropyl cellulose.
Said tablet composition may be used as a medicament, particularly in the treatment of chronic myeloid leukaemia (CML) and acute lymphoblastic leukaemia (ALL).
DETAILED DESCRIPTION OF THE PRESENT INVENTION
Sprycel® contains the monohydrated form of dasatinib as disclosed in WO2005077945, the so-called form "Hl-7". Dasatinib monohydrate is a very stable compound, but being a BCS class II compound, it exhibits low aqueous solubility which affects dissolution behavior.
Other forms of dasatinib have been disclosed in the prior art. Many of these forms are solvated forms of dasatinib and some of them do contain unwanted (e.g. toxic) solvents. Moreover, some of the forms as disclosed in the prior art are rather unstable.
Various approaches to overcome the poor aqueous solubility of drug candidates have been investigated in drug research and development. Since the solubility of amorphous forms is higher compared to the solubility of crystalline forms, it would be desirable to have the drug candidate available in amorphous form. However, drugs that can exist in either amorphous or crystalline form tend to crystallize over time when present in amorphous state because the crystalline form of the drug is a lower-energy state than the amorphous form.
One of the most successful strategies to improve the dissolution of poorly soluble drugs is the preparation of a solid dispersion. The term solid dispersion has been defined as a dispersion of one or more Active Pharmaceutical Ingredients (APIs) in an inert carrier or matrix at the solid state, prepared by a solvent or melting process or a combination of the two. Depending on the physical state of the carrier, which is crystalline or amorphous, the solid dispersions are divided into crystalline solid dispersions and amorphous solid dispersions respectively. Amorphous carriers used are mostly polymers. In amorphous solid dispersions, the API is dispersed in very small size and exists in supersaturated state in amorphous carriers because of forced degradation. The amorphous carriers can increase the wettability and dispersibility of drugs as well as inhibit the precipitation process of drugs when amorphous solid dispersions are dissolved in water. These properties along with the fast dissolution rate of amorphous carriers due to the low thermodynamic stability of amorphous state carriers, enhance the drug solubility and release rate. Despite the high active research interests, the number of marketed products arising from solid dispersion approaches is very low. This low number is mainly due to scale-up problems and physicochemical instability in the manufacturing process or during storage leading to phase separation and crystallization (Vo et. al, Eur. J. Pharm. Biopharm., 85 (2013) 799-813).
It is not self-evident that a given drug will form an amorphous solid dispersion with just any polymer, and that, even in the event the solid dispersion is formed, it will be stable over time. Factors playing a role herein are the physicochemical properties of both API and polymer, the ratio of API to polymer used and the technique used to prepare the solid dispersion. Techniques to prepare solid dispersions often require very specific conditions for each combination of API and polymer.
In order to obtain tablet compositions comprising a solid dispersion of dasatinib and a polymer, exhibiting adequate dissolution and excellent long term stability, which are suitable for production on commercial scale, the use of several polymers has been investigated. Only some of them appeared to be suitable to be used in accordance with the present invention. Use of some of the polymers resulted in tablet compositions wherein dasatinib was present only partially in amorphous form. Other polymers did provide tablet compositions comprising solid dispersions wherein dasatinib was present in fully amorphous form, but wherein upon storage dasatinib converted into its crystalline form(s).
The process selected to prepare the tablet compositions of the present invention is the solvent evaporation method, because in this method problems related to decomposition, as frequently occurring in the melting method, are prevented. An important prerequisite of the solvent evaporation method is the sufficient solubility of the drug and the carrier in the solvent system. Finding a suitable non-toxic solvent is sometimes difficult because carriers are hydrophilic whereas drugs are hydrophobic.
The present invention provides a tablet composition comprising a solid dispersion of dasatinib and a polymer, wherein the composition is obtained by:
(1) Dissolving dasatinib and the polymer in a solvent mixture comprising water, an alcohol and at least one molar equivalent of acid with respect to dasatinib at a temperature ranging from 45 to 70°C;
(2) Adding the resulting solution to a diluent;
(3) Evaporating the solvent;
(4) Mixing the resulting blend with further excipients;
(5) Compressing the final blend into tablets, wherein the polymer is selected from polyvinylpyrrolidone, copovidone and hydroxypropyl cellulose.
The polymers to be used in accordance with the present invention are selected from polyvinylpyrrolidone (PVP), copovidone and hydroxypropyl cellulose. Commonly used polymers like polyethylene glycol, Eudragit® and hydroxypropyl methylcellulose were found to be unsuitable to be used in the present invention for various reasons. Some of the polymers did not result in fully amorphous solid dispersions, while other polymers did give rise to amorphous solid dispersions but wherein dasatinib started to crystallize over time. Moreover, some polymers were found to be unsuitable to be used in accordance with the present invention due to their solubility/gelling behavior in the solvent system. Polyvinylpyrrolidone is a particularly preferred polymer to be used in accordance with the present invention.
The weight ratio of dasatinib to polymer in the solid dispersion ranges from about 1: 1 to about 1:6, preferably from 1: 1 to 1:3.
The tablet composition of the present invention is obtained by dissolving, in the first step of the process, dasatinib and the polymer in a solvent mixture. The non-toxic solvent mixture in accordance with the present invention comprises water, an alcohol and at least one molar equivalent of acid with respect to dasatinib. Both dasatinib and the polymer dissolve well in the solvent mixture upon heating at a temperature ranging from 45 to 70°C.
Preferably, the acid used in accordance with the present invention is a mineral acid. Most preferably, the acid is hydrochloric acid. The amount of acid present in the solvent mixture may range. However, at molar ratios of acid to dasatinib below 1: 1, dissolution is insufficient, while at too high ratios, gelling was observed. Preferably, the molar ratio of acid to dasatinib is ranging from 1: 1 to 3: 1. More preferably, the molar ratio of acid to dasatinib is ranging from 1: 1 to 2: 1. The alcohol used in the solvent mixture of the present invention is selected from methanol, ethanol and isopropanol. Ethanol is a particularly preferred alcohol. The volume ratio water to alcohol in the solvent mixture may vary, but is preferably ranging from 2:3 to 1:4. At these ratios, optimal solubility is achieved.
After the first step of dissolving dasatinib and the polymer in the solvent mixture, the resulting solution is added to a diluent. In a preferred embodiment of the present invention, the addition of the solution to the diluent is performed by spraying the solution over the diluent. The diluent to be used in accordance with the present invention may be any diluent known to a person of ordinary skill in the art. Particularly, the diluent to be used in accordance with the present invention is an inorganic diluent, polysaccharide, mono- or disaccharide or sugar alcohol. Microcrystalline cellulose is a particularly preferred diluent.
After the step of adding the solution, comprising of dasatinib and the polymer in the solvent mixture, to a diluent, the solvent is evaporated. The evaporation is carried out by techniques known to a person of ordinary skill in the art.
The resulting blend is then mixed with further excipients. The tablet compositions according to the present invention comprise, besides a diluent, further pharmaceutically acceptable excipients. The excipients to be used in accordance with the present invention are well4inown and are those excipients which are conventionally used by the person skilled in the art. The pharmaceutically acceptable excipients are chosen from one or more binders, disintegrants, glidants or lubricants. Most preferably, the further excipients are chosen from one or more disintegrants and one or more lubricants.
The disintegrant to be used in accordance with the present invention may be any disintegrant known to a person of ordinary skill in the art. Suitable disintegrants to be used in accordance with the present invention are selected from the group consisting of
croscarmellose sodium, crospovidone or sodium starch glycolate. Croscarmellose sodium is a particularly preferred disintegrant. The lubricant to be used in accordance with the present invention may be any lubricant known to a person of ordinary skill in the art. Magnesium stearate is a particularly preferred lubricant.
After mixing the blend, comprising of the solid dispersion of dasatinb and the polymer, with further excipients, the final blend is compressed into tablets, using equipment and methods well-known in the art.
The tablet composition of the present invention exhibits excellent long term stability. Even after 3 months at 40°C/75% RH, no conversion into any crystalline form of dasatinib was observed. Moreover, the tablet composition of the present invention is cost effective and very suitable for production on commercial scale.
The tablet composition of the present invention display dissolution behavior typical for immediate-release formulations. The composition of the present invention exhibits a dissolution rate of at least 85% in 15-30 minutes when tested in 500 ml 0.01 N hydrochloric acid pH 2.0 in a USP apparatus II at 75 rpm, 25°C.
The tablet composition in accordance with the present invention may be used as a medicament. The pharmaceutical composition typically may be used in the treatment of chronic myeloid leukaemia (CML) and acute lymphoblastic leukaemia (ALL).
The following examples are intended to illustrate the scope of the present invention but not to limit it thereto.
EXAMPLES
The tablets comprising a solid dispersion of dasatinib and a polymer were prepared by wet granulation and have the composition as given in table 1. Table 1: tablet composition of tablets comprising a solid dispersion of dasatinib and a polymer
Figure imgf000010_0001
87.5 g of the polymer was dissolved in a heated solution (60°C) of ethanol 96%
(v/v):aqueous hydrochloric acid (3M):water 60:5:35. 87.5 g of dasatinib was added. The total solution volume was 1250 ml. The resulting solution was added to 159.25 g of
microcrystalline cellulose in a fluid bed dryer. The solvent was evaporated by using a top spraying/drying process. During spraying/drying the temperature was adjusted to 45-80°C, depending on the polymer used. The resulting blend was sieved through an appropriate mesh size sieve. 14.00 g of croscarmellose sodium was sieved through a suitable mesh size sieve for deagglomeration and mixed with the blend. Magnesium stearate was sieved through an appropriate sieve to deagglomerate and mixed with the powder mix. The homogeneous powder obtained, was compressed using a rotating tablet press using appropriate punches. The tablets were stored at 40°C/75% RH (open dish). Stability results:
Table 2: Stability results at 40°C/75% RH open dish for tablets comprising dispersion of ' dasatinib:polyvinylpyrrolidone 1:1
Figure imgf000011_0001

Claims

A tablet composition comprising a solid dispersion of dasatinib and a polymer, wherein the composition is obtained by:
(1) Dissolving dasatinib and the polymer in a solvent mixture comprising water, an alcohol and at least one molar equivalent of acid with respect to dasatinib at a temperature ranging from 45 to 70°C;
(2) Adding the resulting solution to a diluent;
(3) Evaporating the solvent;
(4) Mixing the resulting blend with further excipients;
(5) Compressing the final blend into tablets,
wherein the polymer is selected from polyvinylpyrrolidone, copovidone and hydroxypropyl cellulose.
The tablet composition according to claim 1, wherein the diluent is microcrystalline cellulose.
The tablet composition according to claim 1 or 2, wherein the polymer is
polyvinylpyrrolidone.
The tablet composition according to any one of claims 1 to 3, wherein the weight ratio of dasatinib to polymer is ranging from 1: 1 to 1:3.
The tablet composition according to any one of claims 1 to 4, wherein the alcohol is selected from methanol, ethanol and isopropanol.
The tablet composition according to any one of claims 1 to 5, wherein the alcohol is ethanol.
The tablet composition according to any one of claims 1 to 6, wherein the volume ratio water to alcohol in the solvent mixture is ranging from 2:3 to 1:4.
8. The tablet composition according to any one of claims 1 to 7, wherein the molar ratio of acid to dasatinib is ranging from 1: 1 to 3: 1.
9. The tablet composition according to any one of claims 1 to 8, wherein the acid is a
mineral acid.
10. The tablet composition according to any one of claims 1 to 9, wherein the acid is
hydrochloric acid.
11. The tablet composition according to any one of claims 1 to 10, wherein the further excipients are chosen from one or more disintegrants and one or more lubricants.
12. The tablet composition according to any one of claims 1 to 11 exhibiting a dissolution rate of at least 85% in 15-30 minutes when tested in aqueous hydrochloric acid pH 2.0 in a USP apparatus II at 75 rpm, 25°C.
13. The tablet composition according to any one of claims 1 to 12, for use as a medicament.
14. The tablet composition according to claim 13 for use in the treatment of chronic
myeloid leukaemia (CML) and acute lymphoblastic leukaemia (ALL).
PCT/EP2016/081369 2015-12-22 2016-12-16 Pharmaceutical composition comprising amorphous dasatinib WO2017108605A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP15202074.9 2015-12-22
EP15202074 2015-12-22

Publications (1)

Publication Number Publication Date
WO2017108605A1 true WO2017108605A1 (en) 2017-06-29

Family

ID=55069716

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2016/081369 WO2017108605A1 (en) 2015-12-22 2016-12-16 Pharmaceutical composition comprising amorphous dasatinib

Country Status (1)

Country Link
WO (1) WO2017108605A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019209908A1 (en) 2018-04-25 2019-10-31 Johnson Matthey Public Limited Company Crystalline forms of dasatinib
CN111217807A (en) * 2018-11-26 2020-06-02 安礼特(上海)医药科技有限公司 Dasatinib amorphous form and preparation method thereof
CN111904933A (en) * 2019-05-08 2020-11-10 北京化工大学 Transparent water-dispersible dasatinib nanoemulsion and preparation method thereof
US10940149B1 (en) 2018-06-15 2021-03-09 Handa Oncology, Llc Kinase inhibitor salts and compositions thereof
WO2021150981A1 (en) * 2020-01-24 2021-07-29 Nanocopoeia, Llc Amorphous solid dispersions of dasatinib and uses thereof
US20220175774A1 (en) * 2020-12-07 2022-06-09 Alembic Pharmaceuticals Limited Bioavailable Oral Dosage Form Of Tyrosine-Kinase Inhibitor
US11389450B2 (en) 2020-01-31 2022-07-19 Nanocopoeia, Llc Amorphous nilotinib microparticles and uses thereof
WO2022157308A1 (en) * 2021-01-21 2022-07-28 Xspray Pharma Ab (Publ) Fast dissolving pharmaceutical compositions of dasatinib
WO2022254033A1 (en) 2021-06-04 2022-12-08 Bend Research, Inc. Formic acid as processing aid in spray drying for basic drugs
US11559485B2 (en) 2020-04-30 2023-01-24 Nanocopoeia, Llc Orally disintegrating tablet comprising amorphous solid dispersion of nilotinib

Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1169038A1 (en) 1999-04-15 2002-01-09 Bristol-Myers Squibb Company 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
WO2009053854A2 (en) 2007-10-23 2009-04-30 Teva Pharmaceutical Industries Ltd. Polymorphs of dasatinib and process for preparation thereof
WO2010062715A2 (en) 2008-11-03 2010-06-03 Teva Pharmaceutical Industries Ltd. Polymorphs of dasatinib and process for preparation thereof
WO2010067374A2 (en) 2008-12-08 2010-06-17 Hetero Research Foundation Polymorphs of dasatinib
WO2011095059A1 (en) 2010-02-08 2011-08-11 南京卡文迪许生物工程技术有限公司 Polymorphs of dasatinib, preparation methods and pharmaceutical compositions thereof
WO2012014149A1 (en) 2010-07-30 2012-02-02 Ranbaxy Laboratories Limited N-methylformamide solvate of dasatinib
WO2013105895A1 (en) 2012-01-13 2013-07-18 Xspray Microparticles Ab A pharmaceutical composition comprising stable, amorphous hybrid nanopraticles of at least one protein kinase inhibitor and at least one polymeric stabilizing and matrix-forming component
US20140343073A1 (en) * 2013-03-26 2014-11-20 Cadila Healthcare Limited Process for preparation of amorphous form of dasatinib
CN104327067A (en) 2014-10-11 2015-02-04 深圳市浩瑞实业发展有限公司 Preparation method of amorphous dasatinib
CN104367557A (en) 2013-08-12 2015-02-25 浙江九洲药业股份有限公司 Preparation method of amorphous composition composed of drug active component and PVP
WO2015049645A2 (en) 2013-10-04 2015-04-09 Alembic Pharmaceuticals Limited An improved process for the preparation of dasatinib

Patent Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1169038A1 (en) 1999-04-15 2002-01-09 Bristol-Myers Squibb Company 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
WO2009053854A2 (en) 2007-10-23 2009-04-30 Teva Pharmaceutical Industries Ltd. Polymorphs of dasatinib and process for preparation thereof
WO2010062715A2 (en) 2008-11-03 2010-06-03 Teva Pharmaceutical Industries Ltd. Polymorphs of dasatinib and process for preparation thereof
WO2010067374A2 (en) 2008-12-08 2010-06-17 Hetero Research Foundation Polymorphs of dasatinib
WO2011095059A1 (en) 2010-02-08 2011-08-11 南京卡文迪许生物工程技术有限公司 Polymorphs of dasatinib, preparation methods and pharmaceutical compositions thereof
WO2012014149A1 (en) 2010-07-30 2012-02-02 Ranbaxy Laboratories Limited N-methylformamide solvate of dasatinib
WO2013105895A1 (en) 2012-01-13 2013-07-18 Xspray Microparticles Ab A pharmaceutical composition comprising stable, amorphous hybrid nanopraticles of at least one protein kinase inhibitor and at least one polymeric stabilizing and matrix-forming component
WO2013105894A1 (en) 2012-01-13 2013-07-18 Xspray Microparticles Ab A method for producing stable, amorphous hybrid nanoparticles comprising at least one protein kinase inhibitor and at least one polymeric stabilizing and matrix- forming component.
US20140343073A1 (en) * 2013-03-26 2014-11-20 Cadila Healthcare Limited Process for preparation of amorphous form of dasatinib
CN104367557A (en) 2013-08-12 2015-02-25 浙江九洲药业股份有限公司 Preparation method of amorphous composition composed of drug active component and PVP
WO2015049645A2 (en) 2013-10-04 2015-04-09 Alembic Pharmaceuticals Limited An improved process for the preparation of dasatinib
CN104327067A (en) 2014-10-11 2015-02-04 深圳市浩瑞实业发展有限公司 Preparation method of amorphous dasatinib

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
VO, EUR. J. PHARM. BIOPHARM., vol. 85, 2013, pages 799 - 813

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019209908A1 (en) 2018-04-25 2019-10-31 Johnson Matthey Public Limited Company Crystalline forms of dasatinib
US11440908B2 (en) 2018-04-25 2022-09-13 Johnson Matthey Public Limited Company Crystalline forms of dasatinib
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
US11007195B2 (en) 2018-06-15 2021-05-18 Handa Oncology, Llc Kinase inhibitor salts, and compositions thereof
US10940149B1 (en) 2018-06-15 2021-03-09 Handa Oncology, Llc Kinase inhibitor salts and compositions thereof
CN111217807A (en) * 2018-11-26 2020-06-02 安礼特(上海)医药科技有限公司 Dasatinib amorphous form and preparation method thereof
CN111904933A (en) * 2019-05-08 2020-11-10 北京化工大学 Transparent water-dispersible dasatinib nanoemulsion and preparation method thereof
CN111904933B (en) * 2019-05-08 2023-06-13 北京化工大学 Transparent water-dispersed dasatinib nanoemulsion and preparation method thereof
US11202778B2 (en) 2020-01-24 2021-12-21 Nanocopoeia, Llc Amorphous solid dispersions of dasatinib and uses thereof
US11298356B1 (en) 2020-01-24 2022-04-12 Nanocopoeia, Llc Amorphous solid dispersions of dasatinib and uses thereof
US11324745B2 (en) 2020-01-24 2022-05-10 Nanocopoeia, Llc Amorphous solid dispersions of dasatinib and uses thereof
WO2021150981A1 (en) * 2020-01-24 2021-07-29 Nanocopoeia, Llc Amorphous solid dispersions of dasatinib and uses thereof
US11413290B2 (en) 2020-01-24 2022-08-16 Nanocopoeia, Llc Amorphous solid dispersions of dasatinib and uses thereof
US11633398B2 (en) 2020-01-24 2023-04-25 Nanocopoeia, Llc Amorphous solid dispersions of dasatinib and uses thereof
US11389450B2 (en) 2020-01-31 2022-07-19 Nanocopoeia, Llc Amorphous nilotinib microparticles and uses thereof
US11559485B2 (en) 2020-04-30 2023-01-24 Nanocopoeia, Llc Orally disintegrating tablet comprising amorphous solid dispersion of nilotinib
US20220175774A1 (en) * 2020-12-07 2022-06-09 Alembic Pharmaceuticals Limited Bioavailable Oral Dosage Form Of Tyrosine-Kinase Inhibitor
WO2022157308A1 (en) * 2021-01-21 2022-07-28 Xspray Pharma Ab (Publ) Fast dissolving pharmaceutical compositions of dasatinib
US11529351B2 (en) 2021-01-21 2022-12-20 Xspray Pharma Ab Fast dissolving pharmaceutical compositions
WO2022254033A1 (en) 2021-06-04 2022-12-08 Bend Research, Inc. Formic acid as processing aid in spray drying for basic drugs

Similar Documents

Publication Publication Date Title
WO2017108605A1 (en) Pharmaceutical composition comprising amorphous dasatinib
CN107530348B (en) Pharmaceutical composition containing JAK kinase inhibitor or pharmaceutically acceptable salt thereof
US9511026B2 (en) Poorly soluble drug containing microspheres with improved bioavailability and method of preparing the same
JP2012512840A (en) Solid composition containing rasagiline component
US20090088465A1 (en) Pharmaceutical Compositions of Amorphous Atorvastatin and Process for Preparing Same
EP3389640B1 (en) Pharmaceutical composition comprising anhydrous dasatinib
WO2017032908A1 (en) Pharmaceutical composition comprising amorphous posaconazole
WO2008027600A2 (en) Imatinib compositions
CN107320460B (en) Oral nilotinib nano preparation and preparation method thereof
WO2018069937A1 (en) Solid dispersions of trisodium sacubitril valsartan and process for the preparation thereof
US10660963B2 (en) Pharmaceutical composition containing tacrolimus and preparation methods thereof
US20180000827A1 (en) Pharmaceutical composition comprising gefitinib
KR101331723B1 (en) Pharmaceutical compositions comprising a hcv polymerase inhibitor prodrug
CN110121333B (en) Formulations containing benzimidazole derivatives
US20220347102A1 (en) Pharmaceutical composition comprising enzalutamide
US9561186B2 (en) Method for improving the bioavailability of low aqueous solubility drugs
WO2017037645A1 (en) Stable pharmaceutical formulations of teriflunomide
KR101282847B1 (en) Solid dispersion containing cilostazol and pharmaceutical composition comprising the same
EP3171861A1 (en) Solid pharmaceutical composition comprising pi3k-inhibitor
CN111511365A (en) Improved pharmaceutical formulations
WO2013169523A1 (en) Solubility enhanced compositions
WO2015086768A1 (en) Pharmaceutical composition comprising a pharmaceutically acceptable salt of rasagiline
WO2013100876A1 (en) Risperidone formulations
US20200093804A1 (en) Pharmaceutical composition of everolimus
Verma et al. Available Online Through Review Article www. ijptonline. com

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: 16809858

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: 16809858

Country of ref document: EP

Kind code of ref document: A1