WO2010136522A2 - A piperazine derivative free, or essentially free, of potential genotoxicity, and a process for preparing the same - Google Patents

A piperazine derivative free, or essentially free, of potential genotoxicity, and a process for preparing the same Download PDF

Info

Publication number
WO2010136522A2
WO2010136522A2 PCT/EP2010/057315 EP2010057315W WO2010136522A2 WO 2010136522 A2 WO2010136522 A2 WO 2010136522A2 EP 2010057315 W EP2010057315 W EP 2010057315W WO 2010136522 A2 WO2010136522 A2 WO 2010136522A2
Authority
WO
WIPO (PCT)
Prior art keywords
ranolazine
iii
compound
free
organic solvent
Prior art date
Application number
PCT/EP2010/057315
Other languages
French (fr)
Other versions
WO2010136522A3 (en
Inventor
Iolanda CHAMORRO GUTIÉRREZ
Raquel Bou Bosch
Original Assignee
Medichem S.A.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Medichem S.A. filed Critical Medichem S.A.
Publication of WO2010136522A2 publication Critical patent/WO2010136522A2/en
Publication of WO2010136522A3 publication Critical patent/WO2010136522A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D295/00Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms
    • C07D295/04Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms
    • C07D295/14Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D295/145Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with the ring nitrogen atoms and the carbon atoms with three bonds to hetero atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings
    • C07D295/15Heterocyclic compounds containing polymethylene-imine rings with at least five ring members, 3-azabicyclo [3.2.2] nonane, piperazine, morpholine or thiomorpholine rings, having only hydrogen atoms directly attached to the ring carbon atoms with substituted hydrocarbon radicals attached to ring nitrogen atoms substituted by carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with the ring nitrogen atoms and the carbon atoms with three bonds to hetero atoms attached to the same carbon chain, which is not interrupted by carbocyclic rings to an acyclic saturated chain
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • Ranolazine (Compound I) is the international common accepted name for ( ⁇ )- 1 -[3-(2-methoxyphenoxy)-2-hydroxypropyl]-4-[ ⁇ /-(2,6-dimethylphenyl)- carbamoyl-methyljpiperazine, and has an empirical formula of C24H33N3O4 and a molecular weight of 427.54.
  • Ranolazine is an active pharmaceutical substance which has antianginal and anti-ischemic effects that do not depend upon reductions in heart rate or blood pressure. In the United States, ranolazine is marketed under the name RANEXATM, and is indicated for the treatment of chronic angina.
  • ranolazine The preparation of ranolazine is disclosed in U.S. Patent No. 4,567,264. More precisely, U.S. Patent No. 4,567,264 describes the preparation of ranolazine base by using a guaiacol derivative (i.e. 2-(2-methoxyphenoxy)-methyloxirane, compound III) as an intermediate compound (See Scheme 1).
  • a guaiacol derivative i.e. 2-(2-methoxyphenoxy)-methyloxirane, compound III
  • compound (III) is used as an intermediate for the synthesis of ranolazine, it is likely to be present as an impurity not only in the active pharmaceutical ingredient, but also in the finished product. Further, given that compound (III) is not classified as a toxic substance, to date its presence in both the ranolazine active pharmaceutical ingredient and in the corresponding finished drug is controlled in order to fulfill the general acceptance of not more than 0.05% established for ranolazine impurities.
  • the invention provides a method of determining the presence or amount of potentially geno toxic impurities in ( ⁇ )-l-[3-(2-methoxyphenoxy)-2-hydroxypropyl]-4-
  • the invention provides ranolazine free, or substantially free, of potential genotoxicity wherein compound (III) is present in an amount equal to or less than 750 ppb (w/w) relative to ranolazine, and provides a process for preparing the same.
  • Figure 1 is a GC chromatogram of a reference standard solution of compound (III) in toluene [1.5 ⁇ g/mL; which corresponds to 750 ppb of compound (III) relative to ranolazine (w/w)].
  • Figure 2 is the GC chromatogram of the sample solution obtained from ranolazine of Example 1.
  • the invention provides a method of determining the presence of potentially genotoxic impurities in ( ⁇ )-l-[3-(2-methoxyphenoxy)-2- hydroxypropyl]-4-[ ⁇ /-(2,6-dimethylphenyl)carbamoylmethyl]piperazine (i.e. ranolazine) or in a composition containing ranolazine, wherein 2-(2- methoxyphenoxy)methyloxirane is employed as a reference marker.
  • the invention refers to the use of compound (III) as a reference marker in the determination of potentially genotoxic impurities in a sample of a ranolazine batch.
  • reference marker refers to a compound that is employed in qualitative analysis to identify components of a mixture based on their position in a chromatogram, e.g. in a HPLC or GC chromatogram, or on a Thin Layer Chromatography (TLC) plate.
  • composition containing ranolazine refers to a chemical or pharmaceutical mixture containing ranolazine intended for pharmaceutical use.
  • the invention provides a method for determining the amount of potentially genotoxic impurities in ( ⁇ )-l-[3-(2- methoxyphenoxy)-2-hydroxypropyl]-4-[ ⁇ /-(2,6-dimethylphenyl)carbamoylmethyl]- piperazine (i.e. ranolazine) or in a composition containing ranolazine, wherein 2-(2- methoxyphenoxy)-methyloxirane is employed as a reference standard.
  • the invention refers to the use of compound (III) as a reference standard in the determination of potentially genotoxic impurities in a sample of a ranolazine batch, and for example refers to its use in order to quantify the amount of compound (III) in said sample of a ranolazine batch.
  • reference standard refers to a compound that may be used both for quantitative and qualitative analysis of ranolazine.
  • HPLC retention time of the reference standard allows a relative retention time with respect to ranolazine to be determined, thus making qualitative analysis possible.
  • concentration of compound (III) in a solution injected into an HPLC or GC column allows the areas under the HPLC or GC peaks to be compared, thus making quantitative analysis possible.
  • a reference marker is used only for qualitative analysis, while a reference standard may be used for quantitative or qualitative analysis, or both. Hence a reference marker is a subset of a reference standard, and is included within the definition of a reference standard.
  • the present inventors have found that the acceptable limit for compound (III) in both the ranolazine active substance and in the corresponding finished drug product needs to be significantly narrowed to a value much lower than the 0.05% general acceptance for impurities in ranolazine.
  • the applicants have found that the presence of compound (III) in ranolazine and in the corresponding finished drug product needs to be tightly controlled and restricted in order to fulfill the permitted daily dose for potential genotoxic impurities present in a pharmaceutical product.
  • a dose of 1.5 micrograms/day has been described as the acceptable level for genotoxic impurities in pharmaceuticals.
  • the present inventors have found that the compositions of ranolazine for use in pharmaceuticals require a maximum concentration of compound (III) of not more than 750 ppb (i.e. 0.750 ppm or 0.000075%) by weight.
  • control of compound (III) at such low limits represents a significant challenge, particularly from an analytical point of view.
  • the present inventors have found that the standard HPLC methods for analyzing the chemical purity of ranolazine are not suitable methods for determining the presence of compound (III) at such low limits, and have provided an improved method, e.g., GC method, which is suitable for this purpose.
  • the invention provides a standard solution for the determination of potentially genotoxic impurities in samples of a ranolazine batch, wherein said standard solution comprises a solution of compound (III) in an organic solvent having a concentration between 0.60 - 2.00 ⁇ g/mL, and preferably having a concentration of 0.60 ⁇ g/mL, 0.80 ⁇ g/mL, 1.00 ⁇ g/mL, 1.20 ⁇ g/mL, 1.50 ⁇ g/mL, or 2.00 ⁇ g/mL [i.e. which correspond to 300 ppb, 400 ppb, 500 ppb, 600 ppb, 750 ppb, and 1.000 ppb, of compound (III), respectively].
  • the said standard solutions are suitable standard solutions for determining the concentration of compound (III) in ranolazine according to the limit concentration of 750 ppb (w/w) found by the inventors.
  • the said standard solutions are used for preparing a linear calibration curve which is suitable for determining the concentration of compound (III) in ranolazine according to the limit concentration of 750 ppb (w/w) found by the inventors.
  • the organic solvent of the standard solution of the invention is preferably toluene.
  • the invention provides an analytical method for the determination of potentially genotoxic impurities in samples of a ranolazine batch, wherein said method can detect at least a concentration equal to or less than 750 ppb of compound (III) relative to ranolazine (w/w), said method comprising: (i) preparing a test sample by extracting the ranolazine from the sample of a ranolazine batch containing an unknown amount of compound (III), to obtain a solution in an organic solvent having an unknown amount of compound (III) and free, or substantially free, of ranolazine, preferably containing less than 0.5% of ranolazine, more preferably less than 0.1%, even more preferably less than 0.05%, and most preferably less than 0.01% of ranolazine,
  • step (ii) measuring by a suitable GC apparatus the test sample of step (i) to obtain a GC chromatogram, wherein measuring comprises comparing the height and/or area of the peak corresponding to the impurity, and
  • the preparing a test sample of step (i) comprises:
  • the determining the presence of compound (III) in the sample of the ranolazine of step (iii) of the analytical method above can be carried out by (iii) (a) comparing the height and/or area of the peak corresponding to the impurity in step (ii) to the height and/or area of the peak corresponding to the impurity in a standard solution preparation containing a known amount of compound (III) of between 0.60 - 2.00 ⁇ g/mL, and preferably containing a known amount of compound (III) of 0.60 ⁇ g/mL, 0.80 ⁇ g/mL, 1.00 ⁇ g/mL, 1.20 ⁇ g/mL, 1.50 ⁇ g/mL, or 2.00 ⁇ g/mL.
  • the determining the presence of compound (III) in the sample of the ranolazine of step (iii) of the analytical method above can be carried out automatically by (iii) (b) using a linear calibration curve previously prepared from at least two standard solution preparations, preferably from at least five standard solution preparations, and more preferably from six standard solution preparations, of compound (III) having a concentration between 0.60 - 2.00 ⁇ g/mL, preferably having a concentration of 0.60 ⁇ g/mL, 0.80 ⁇ g/mL, 1.00 ⁇ g/mL, 1.20 ⁇ g/mL, 1.50 ⁇ g/mL, or 2.00 ⁇ g/mL [i.e.
  • the analytical method of the invention above can detect down to 132 ppb of compound (III) relative to ranolazine in a sample of a ranolazine batch.
  • the analytical method of the invention shows a linear calibration curve using standard solution preparations of compound (III) having a concentration of 0.60 ⁇ g/mL, 0.80 ⁇ g/mL, 1.00 ⁇ g/mL, 1.20 ⁇ g/mL, 1.50 ⁇ g/mL, and 2.00 ⁇ g/mL [i.e. which correspond to 300 ppb, 400 ppb, 500 ppb, 600 ppb, 750 ppb, and 1.000 ppb, of compound (III), respectively].
  • the organic solvent suitable for the analytical method above may be any suitable organic solvent which is immiscible in water.
  • the organic solvent is a hydrocarbon, e.g., toluene.
  • a water non-miscible organic solvent is an organic solvent which shows a reduced solubility in water and therefore it separates as an upper or lower phase when the concentration of water is increased over its solubility limit.
  • Preferred water non-miscible organic solvents are those having water solubility values (w/w) of less than 50%, more preferably less than 10%, even more preferably less than 1%, and even more preferably less than 0.1%.
  • Non limiting examples of suitable water non-miscible organic solvents include pentyl acetate, anisole, benzaldehyde, benzene, bromobenzene, butyl acetate, butyl ether, chlorobenzene, chloroform, cyclohexane, cyclohexanone, cyclopentane, 1 ,2- dichlorobenzene, 1,2-dichloroethane, dichloromethane, diethoxymethane, diisobutyl ketone, dimethoxymethane, ethyl acetate, ethylbenzene, 1 ,2-diethoxyethane, ethyl ether, phosphoral, n-heptane, n-hexane, isobutyl acetate, isopropyl acetate, isopropyl ether, methyl acetate, methyl tert-butyl ether, methyl cyclohex
  • Particularly preferred organic solvents for the analytic method above are ethyl acetate, isopropyl acetate, methyl tert-butyl ether, methyl ethyl ketone, methyl isobutyl ketone, toluene, xylene, and mixtures thereof, and more particularly the more preferred organic solvent for the analytic method above is toluene.
  • the acidic compound of the step (i) (a) of the analytical method above is preferably an organic or an inorganic acid compound, more preferably is an organic acid compound, and more preferably is tartaric acid.
  • the suitable GC apparatus of step (ii) preferably comprises a column having a polysiloxane as stationary phase, and preferably comprises column having 5% phenyl 95% dimethyl polysiloxane as stationary phase.
  • a suitable GC apparatus is preferably an Agilent 6890N apparatus, wherein a suitable column is preferably a TRB-5A column (composition: 5% phenyl 95% dimethyl polysiloxane, 30 m length, 0.32 mm internal diameter, 0.5 ⁇ m film thickness), and a suitable detector is preferably a flame ionization detector (FID).
  • the invention provides a process for preparing ranolazine which is free, or substantially free, of potential genotoxicity, said process comprising:
  • the providing the ranolazine of step (i) of the process above preferably comprises:
  • the reducing the residual amount of compound (III) in the ranolazine of step (iii) of the process above preferably comprises crystallizing or digesting the ranolazine, and more preferably comprises digesting the ranolazine in a non- hydroxylic organic solvent.
  • the digesting the ranolazine in a non-hydro xylic organic solvent preferably comprises: (iii) (a) suspending the ranolazine in a non-hydroxylic organic solvent,
  • the non-hydroxylic organic solvent of the process of the invention above is preferably a ketone solvent, and more preferably is methyl ethyl ketone.
  • the process of the invention above further comprises an additional step of (iv) admixing the ranolazine with at least one pharmaceutically acceptable carrier, thereby obtaining a pharmaceutical composition comprising ranolazine which is free, or essentially free, of potential genotoxicity.
  • the at least one pharmaceutically acceptable carrier is preferably methacylic acid-ethyl acrylate copolymer, microcrystalline cellulose, hypromellose, sodium hydroxide, magnesium stearate, titanium dioxide, polyethylene glycol, polysorbate 80, glycerol triacetate, lactose monohydrate, or mixtures thereof.
  • the amount of undesired compound (III) in ranolazine or in a composition containing ranolazine can be reduced by methods such as recrystallization or digestion. Recrystallization or digestion of samples containing ranolazine can be performed in any suitable solvent.
  • the preferred solvent is any suitable non-hydroxylic organic solvent. More preferably, methyl ethyl ketone is the suitable non-hydroxylic organic solvent for reducing the amount of compound (III) in compositions containing ranolazine by means of digestion methods.
  • the amount of undesired compound (III) in compositions containing ranolazine can be reduced by adjusting the stoichiometric amount of compound (III) used in the preparation of ranolazine.
  • the present invention provides ranolazine which is free, or substantially free, of potential genotoxicity. More precisely, the said ranolazine which is free, or substantially free, of potential genotoxicity has a concentration of compound (III) of equal to or less than 750 ppb (w/w).
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising ranolazine which is free, or essentially free, of potential genotoxicity, and wherein said pharmaceutical composition has a concentration of compound (III) of equal to or less than 750 ppb (w/w), relative to ranolazine.
  • the GC analysis was performed on an Agilent 6890N. The following parameters were used: Gas carrier: He; Column head pressure: 10 psi (constant pressure); Split ratio: 3:0, Injector Temp.: 250 0 C; Detector Temp. (FID): 300 0 C;
  • Standard solutions of compound (III) A stock solution of 100 ⁇ g/mL of compound (III) in toluene was prepared by weighing 10 mg of compound (III) in a
  • Blank preparation 2g of tartaric acid were dissolved with 10 mL of water. Then, 2 mL of toluene and Ig of sodium chloride were added. The mixture was stirred, the aqueous phase was removed, and the organic phase was injected into the GC apparatus.
  • Preparation of a reference standard solution preparation 2g of tartaric acid were dissolved with 10 mL of water. Then, Ig of sodium chloride and 2 mL of a standard solution of compound (III) in toluene (i.e. solutions containing 0.60, 0.80, 1.0, 1.2, 1.5 and 2.0 ⁇ g/mL) were added. The mixture was stirred, the aqueous phase was removed, and the organic phase was injected into the GC apparatus. A linear calibration curve was determined using the six standard solution preparations [i.e.
  • Test sample preparation 2g of ranolazine were weighed with 2 g of tartaric acid and dissolved with 10 mL of water. Then, 2 mL of toluene and Ig of sodium chloride were added. The mixture was stirred, the aqueous phase was removed, and the organic phase was injected into the GC apparatus.
  • a standard preparation prepared from a standard solution in toluene of compound (III) of 1.5 ⁇ g/mL was used as standard solution preparation for calculating the content of compound (III) in the samples of ranolazine.
  • other standard preparations of compound (III) prepared from standard solutions having a concentration of (III) between 0.60 - 2.0 ⁇ g/mL can be used as standard solution preparation for calculating the content of compound (III) in the samples of ranolazine.
  • ri c s i nnn ppb of compound of formula (III) — x X lUUU rs w
  • ri Area peak response of compound (III) in the Test sample preparation.
  • rs Area peak response of compound (III) in the standard solution preparation.
  • c s Concentration ( ⁇ g/mL) of compound (III) in the standard solution of compound (III).
  • the content of compound (III) in the sample of ranolazine can be calculated automatically from the linear calibration curve determined above from the six standard solution preparations [i.e. 0.6, 0.80, 1.0,
  • the chromatographic separation was carried out in a Symmetry C 18, 5 ⁇ m, 250 x 4.6 mm LD column; at 37 0 C.
  • the mobile phase B was acetonitrile.
  • the chromatograph was equipped with a 230 nm detector and the flow rate was 1.0 mL per minute.
  • the chromatograph was programmed as follows: Initial 0-2 min. isocratic 100% mobile phase A, 2-16 min. linear gradient to 93% mobile phase A, 16-
  • Test samples were prepared by dissolving the appropriate amount of sample to obtain 5 mg per mL in mobile phase, and 20 ⁇ L were injected.
  • Example 1 Preparation of Ranolazine free of potential genotoxicity. 30.56 g of ranolazine [24.07 g of estimated dry mass; HPLC purity:

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Urology & Nephrology (AREA)
  • Cardiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Vascular Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Heart & Thoracic Surgery (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)
  • Investigating Or Analyzing Non-Biological Materials By The Use Of Chemical Means (AREA)

Abstract

Disclosed is ranolazine, or a composition containing ranolazine, which is free, or substantially free, of potential genotoxicity wherein 2-(2-methoxyphenoxy)- methyloxirane is present in an amount equal to or less than 750 ppb (w/w) relative to ranolazine, and provides a process for preparing the same. Also disclosed is a method for determining the purity of ranolazine, wherein 2-(2-methoxyphenoxy)- methyloxirane is employed as a reference marker and/or standard.

Description

A PIPERAZINE DERIVATIVE FREE, OR ESSENTIALLY FREE, OF POTENTIAL GENOTOXICITY, AND A PROCESS FOR PREPARING THE SAME
This application claims the benefit of the U.S. Provisional Patent
Application Ser. No 61/181607 filed on May 27, 2009.
BACKGROUND OF THE INVENTION
Ranolazine (Compound I) is the international common accepted name for (±)- 1 -[3-(2-methoxyphenoxy)-2-hydroxypropyl]-4-[Λ/-(2,6-dimethylphenyl)- carbamoyl-methyljpiperazine, and has an empirical formula of C24H33N3O4 and a molecular weight of 427.54.
Figure imgf000002_0001
Ranolazine is an active pharmaceutical substance which has antianginal and anti-ischemic effects that do not depend upon reductions in heart rate or blood pressure. In the United States, ranolazine is marketed under the name RANEXA™, and is indicated for the treatment of chronic angina.
The preparation of ranolazine is disclosed in U.S. Patent No. 4,567,264. More precisely, U.S. Patent No. 4,567,264 describes the preparation of ranolazine base by using a guaiacol derivative (i.e. 2-(2-methoxyphenoxy)-methyloxirane, compound III) as an intermediate compound (See Scheme 1).
Figure imgf000003_0001
Figure imgf000003_0002
Scheme 1
Since compound (III) is used as an intermediate for the synthesis of ranolazine, it is likely to be present as an impurity not only in the active pharmaceutical ingredient, but also in the finished product. Further, given that compound (III) is not classified as a toxic substance, to date its presence in both the ranolazine active pharmaceutical ingredient and in the corresponding finished drug is controlled in order to fulfill the general acceptance of not more than 0.05% established for ranolazine impurities. BRIEF SUMMARY OF THE INVENTION
The invention provides a method of determining the presence or amount of potentially geno toxic impurities in (±)-l-[3-(2-methoxyphenoxy)-2-hydroxypropyl]-4-
[Λ/-(2,6-dimethylphenyl)carbamoylmethyl]piperazine (i.e. ranolazine) or in a composition containing ranolazine, wherein 2-(2-methoxyphenoxy)-methyloxirane is employed as a reference marker or reference standard.
Additionally, the invention provides ranolazine free, or substantially free, of potential genotoxicity wherein compound (III) is present in an amount equal to or less than 750 ppb (w/w) relative to ranolazine, and provides a process for preparing the same. BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a GC chromatogram of a reference standard solution of compound (III) in toluene [1.5 μg/mL; which corresponds to 750 ppb of compound (III) relative to ranolazine (w/w)].
Figure 2 is the GC chromatogram of the sample solution obtained from ranolazine of Example 1.
DETAILED DESCRIPTION OF THE INVENTION
The present inventors have found that 2-(2-methoxyphenoxy)- methyloxirane (compound III),
Figure imgf000004_0001
shows potential genotoxic properties when tested in the Ames bacterial mutagenicity test, which is an indication that the material may have mutagenic and carcinogenic potential.
In an embodiment, the invention provides a method of determining the presence of potentially genotoxic impurities in (±)-l-[3-(2-methoxyphenoxy)-2- hydroxypropyl]-4-[Λ/-(2,6-dimethylphenyl)carbamoylmethyl]piperazine (i.e. ranolazine) or in a composition containing ranolazine, wherein 2-(2- methoxyphenoxy)methyloxirane is employed as a reference marker. Thus, the invention refers to the use of compound (III) as a reference marker in the determination of potentially genotoxic impurities in a sample of a ranolazine batch.
The term "reference marker", as used herein, refers to a compound that is employed in qualitative analysis to identify components of a mixture based on their position in a chromatogram, e.g. in a HPLC or GC chromatogram, or on a Thin Layer Chromatography (TLC) plate. The term "composition containing ranolazine", as used herein, refers to a chemical or pharmaceutical mixture containing ranolazine intended for pharmaceutical use.
In another embodiment, the invention provides a method for determining the amount of potentially genotoxic impurities in (±)-l-[3-(2- methoxyphenoxy)-2-hydroxypropyl]-4-[Λ/-(2,6-dimethylphenyl)carbamoylmethyl]- piperazine (i.e. ranolazine) or in a composition containing ranolazine, wherein 2-(2- methoxyphenoxy)-methyloxirane is employed as a reference standard. Thus, the invention refers to the use of compound (III) as a reference standard in the determination of potentially genotoxic impurities in a sample of a ranolazine batch, and for example refers to its use in order to quantify the amount of compound (III) in said sample of a ranolazine batch.
The term "reference standard", as used herein, refers to a compound that may be used both for quantitative and qualitative analysis of ranolazine. For example, the HPLC retention time of the reference standard allows a relative retention time with respect to ranolazine to be determined, thus making qualitative analysis possible. Furthermore, the concentration of compound (III) in a solution injected into an HPLC or GC column allows the areas under the HPLC or GC peaks to be compared, thus making quantitative analysis possible. A reference marker is used only for qualitative analysis, while a reference standard may be used for quantitative or qualitative analysis, or both. Hence a reference marker is a subset of a reference standard, and is included within the definition of a reference standard.
In addition, the present inventors have found that the acceptable limit for compound (III) in both the ranolazine active substance and in the corresponding finished drug product needs to be significantly narrowed to a value much lower than the 0.05% general acceptance for impurities in ranolazine.
Namely, the applicants have found that the presence of compound (III) in ranolazine and in the corresponding finished drug product needs to be tightly controlled and restricted in order to fulfill the permitted daily dose for potential genotoxic impurities present in a pharmaceutical product. In this regard, a dose of 1.5 micrograms/day has been described as the acceptable level for genotoxic impurities in pharmaceuticals. Thus, the concentration of a potential genotoxic impurity considered acceptable in a pharmaceutical product can be calculated according to the following formula: maximum concentration of the potential genotoxic impurity (ppm) = 1.5 μg/active ingredient daily dose (in g). Since the maximum recommended dose of ranolazine is of Ig twice daily (i.e. 2 g/day), the present inventors have found that the compositions of ranolazine for use in pharmaceuticals require a maximum concentration of compound (III) of not more than 750 ppb (i.e. 0.750 ppm or 0.000075%) by weight. Thus, control of compound (III) at such low limits represents a significant challenge, particularly from an analytical point of view. In this regard, the present inventors have found that the standard HPLC methods for analyzing the chemical purity of ranolazine are not suitable methods for determining the presence of compound (III) at such low limits, and have provided an improved method, e.g., GC method, which is suitable for this purpose.
In another aspect, the invention provides a standard solution for the determination of potentially genotoxic impurities in samples of a ranolazine batch, wherein said standard solution comprises a solution of compound (III) in an organic solvent having a concentration between 0.60 - 2.00 μg/mL, and preferably having a concentration of 0.60 μg/mL, 0.80 μg/mL, 1.00 μg/mL, 1.20 μg/mL, 1.50 μg/mL, or 2.00 μg/mL [i.e. which correspond to 300 ppb, 400 ppb, 500 ppb, 600 ppb, 750 ppb, and 1.000 ppb, of compound (III), respectively]. The said standard solutions are suitable standard solutions for determining the concentration of compound (III) in ranolazine according to the limit concentration of 750 ppb (w/w) found by the inventors. In an aspect, the said standard solutions are used for preparing a linear calibration curve which is suitable for determining the concentration of compound (III) in ranolazine according to the limit concentration of 750 ppb (w/w) found by the inventors.
The organic solvent of the standard solution of the invention is preferably toluene. In an embodiment, the invention provides an analytical method for the determination of potentially genotoxic impurities in samples of a ranolazine batch, wherein said method can detect at least a concentration equal to or less than 750 ppb of compound (III) relative to ranolazine (w/w), said method comprising: (i) preparing a test sample by extracting the ranolazine from the sample of a ranolazine batch containing an unknown amount of compound (III), to obtain a solution in an organic solvent having an unknown amount of compound (III) and free, or substantially free, of ranolazine, preferably containing less than 0.5% of ranolazine, more preferably less than 0.1%, even more preferably less than 0.05%, and most preferably less than 0.01% of ranolazine,
(ii) measuring by a suitable GC apparatus the test sample of step (i) to obtain a GC chromatogram, wherein measuring comprises comparing the height and/or area of the peak corresponding to the impurity, and
(iii) determining the presence of compound (III) in the sample of the ranolazine.
Preferably, the preparing a test sample of step (i) comprises:
(i) (a) treating the sample of a ranolazine batch containing an unknown amount of compound (III) with a suitable amount of an acidic compound in the presence of water and a water non-miscible organic solvent, to obtain an aqueous / organic biphasic system comprising an aqueous phase containing the acid addition salt of ranolazine and an organic phase having an unknown amount of compound (III) and free, or essentially free, of ranolazine, preferably containing less than 0.5% of ranolazine, more preferably less than 0.1%, even more preferably less than 0.05%, and most preferably less than 0.01% of ranolazine, and (i) (b) removing the aqueous phase from the mixture, thereby obtaining a test sample solution in a water non-miscible organic solvent having an unknown amount of compound (III) and free, or essentially free, of ranolazine, preferably containing less than 0.5% of ranolazine, more preferably less than 0.1%, even more preferably less than 0.05%, and most preferably less than 0.01% of ranolazine. The determining the presence of compound (III) in the sample of the ranolazine of step (iii) of the analytical method above can be carried out by (iii) (a) comparing the height and/or area of the peak corresponding to the impurity in step (ii) to the height and/or area of the peak corresponding to the impurity in a standard solution preparation containing a known amount of compound (III) of between 0.60 - 2.00 μg/mL, and preferably containing a known amount of compound (III) of 0.60 μg/mL, 0.80 μg/mL, 1.00 μg/mL, 1.20 μg/mL, 1.50 μg/mL, or 2.00 μg/mL. Alternatively, the determining the presence of compound (III) in the sample of the ranolazine of step (iii) of the analytical method above can be carried out automatically by (iii) (b) using a linear calibration curve previously prepared from at least two standard solution preparations, preferably from at least five standard solution preparations, and more preferably from six standard solution preparations, of compound (III) having a concentration between 0.60 - 2.00 μg/mL, preferably having a concentration of 0.60 μg/mL, 0.80 μg/mL, 1.00 μg/mL, 1.20 μg/mL, 1.50 μg/mL, or 2.00 μg/mL [i.e. which correspond to 300 ppb, 400 ppb, 500 ppb, 600 ppb, 750 ppb, and 1.000 ppb, of compound (III), respectively], and more preferably by using a linear calibration curve previously prepared from six standard solution preparations of compound (III) having a concentration of 0.60 μg/mL, 0.80 μg/mL, 1.00 μg/mL, 1.20 μg/mL, 1.50 μg/mL, and 2.00 μg/mL, respectively. In keeping with the invention, the analytical method of the invention above can detect down to 132 ppb of compound (III) relative to ranolazine in a sample of a ranolazine batch. Further, the analytical method of the invention shows a linear calibration curve using standard solution preparations of compound (III) having a concentration of 0.60 μg/mL, 0.80 μg/mL, 1.00 μg/mL, 1.20 μg/mL, 1.50 μg/mL, and 2.00 μg/mL [i.e. which correspond to 300 ppb, 400 ppb, 500 ppb, 600 ppb, 750 ppb, and 1.000 ppb, of compound (III), respectively].
The organic solvent suitable for the analytical method above may be any suitable organic solvent which is immiscible in water. Preferably, the organic solvent is a hydrocarbon, e.g., toluene. A water non-miscible organic solvent is an organic solvent which shows a reduced solubility in water and therefore it separates as an upper or lower phase when the concentration of water is increased over its solubility limit. Preferred water non-miscible organic solvents are those having water solubility values (w/w) of less than 50%, more preferably less than 10%, even more preferably less than 1%, and even more preferably less than 0.1%. Non limiting examples of suitable water non-miscible organic solvents include pentyl acetate, anisole, benzaldehyde, benzene, bromobenzene, butyl acetate, butyl ether, chlorobenzene, chloroform, cyclohexane, cyclohexanone, cyclopentane, 1 ,2- dichlorobenzene, 1,2-dichloroethane, dichloromethane, diethoxymethane, diisobutyl ketone, dimethoxymethane, ethyl acetate, ethylbenzene, 1 ,2-diethoxyethane, ethyl ether, phosphoral, n-heptane, n-hexane, isobutyl acetate, isopropyl acetate, isopropyl ether, methyl acetate, methyl tert-butyl ether, methyl cyclohexane, methyl ethyl ketone, methyl formate, methyl isobutyl ketone, nitrobenzene, n-pentane, 3- pentanone, propyl acetate, propylene carbonate, l-methoxy-2-propanol acetate, propylene oxide, tetrachloroethylene, toluene, 1,1,1-trichloroethane, trichloroethylene, xylene, and mixtures thereof. Particularly preferred organic solvents for the analytic method above are ethyl acetate, isopropyl acetate, methyl tert-butyl ether, methyl ethyl ketone, methyl isobutyl ketone, toluene, xylene, and mixtures thereof, and more particularly the more preferred organic solvent for the analytic method above is toluene.
The acidic compound of the step (i) (a) of the analytical method above is preferably an organic or an inorganic acid compound, more preferably is an organic acid compound, and more preferably is tartaric acid.
The suitable GC apparatus of step (ii) preferably comprises a column having a polysiloxane as stationary phase, and preferably comprises column having 5% phenyl 95% dimethyl polysiloxane as stationary phase. For example, a suitable GC apparatus is preferably an Agilent 6890N apparatus, wherein a suitable column is preferably a TRB-5A column (composition: 5% phenyl 95% dimethyl polysiloxane, 30 m length, 0.32 mm internal diameter, 0.5 μm film thickness), and a suitable detector is preferably a flame ionization detector (FID). In yet another embodiment, the invention provides a process for preparing ranolazine which is free, or substantially free, of potential genotoxicity, said process comprising:
(i) providing ranolazine, (ii) determining the concentration of compound (III) in the ranolazine, and preferably by the chromatographic method of the invention, and
(iii) reducing the residual amount of compound (III) in the ranolazine if the concentration of compound (III) in the ranolazine is higher than 750 ppb (w/w).
The providing the ranolazine of step (i) of the process above, preferably comprises:
(i)(a) synthesizing ranolazine directly, or
(i)(b) isolating ranolazine from a pharmaceutical composition comprising ranolazine.
The reducing the residual amount of compound (III) in the ranolazine of step (iii) of the process above preferably comprises crystallizing or digesting the ranolazine, and more preferably comprises digesting the ranolazine in a non- hydroxylic organic solvent.
The digesting the ranolazine in a non-hydro xylic organic solvent preferably comprises: (iii) (a) suspending the ranolazine in a non-hydroxylic organic solvent,
(iii) (b) optionally, heating the suspension, and
(iii) (c) removing the solvent from the suspension, preferably by filtering the suspension.
The non-hydroxylic organic solvent of the process of the invention above is preferably a ketone solvent, and more preferably is methyl ethyl ketone.
In another aspect, the process of the invention above further comprises an additional step of (iv) admixing the ranolazine with at least one pharmaceutically acceptable carrier, thereby obtaining a pharmaceutical composition comprising ranolazine which is free, or essentially free, of potential genotoxicity.
The at least one pharmaceutically acceptable carrier is preferably methacylic acid-ethyl acrylate copolymer, microcrystalline cellulose, hypromellose, sodium hydroxide, magnesium stearate, titanium dioxide, polyethylene glycol, polysorbate 80, glycerol triacetate, lactose monohydrate, or mixtures thereof.
As said above, the amount of undesired compound (III) in ranolazine or in a composition containing ranolazine can be reduced by methods such as recrystallization or digestion. Recrystallization or digestion of samples containing ranolazine can be performed in any suitable solvent. For digestion methods, the preferred solvent is any suitable non-hydroxylic organic solvent. More preferably, methyl ethyl ketone is the suitable non-hydroxylic organic solvent for reducing the amount of compound (III) in compositions containing ranolazine by means of digestion methods. Also, the amount of undesired compound (III) in compositions containing ranolazine can be reduced by adjusting the stoichiometric amount of compound (III) used in the preparation of ranolazine.
In other embodiments, the present invention provides ranolazine which is free, or substantially free, of potential genotoxicity. More precisely, the said ranolazine which is free, or substantially free, of potential genotoxicity has a concentration of compound (III) of equal to or less than 750 ppb (w/w).
In another further aspect, the present invention relates to a pharmaceutical composition comprising ranolazine which is free, or essentially free, of potential genotoxicity, and wherein said pharmaceutical composition has a concentration of compound (III) of equal to or less than 750 ppb (w/w), relative to ranolazine.
The following examples further illustrate the invention but, of course, should not be construed as in any way limiting its scope. Examples
General Experimental Conditions
GC Method:
The GC analysis was performed on an Agilent 6890N. The following parameters were used: Gas carrier: He; Column head pressure: 10 psi (constant pressure); Split ratio: 3:0, Injector Temp.: 250 0C; Detector Temp. (FID): 300 0C;
Column: TRB-5A (5% phenyl 95% dimethyl polysiloxane), 30 m length, 0.32 mm internal diameter, 0.5 μm film thickness.
The following temperature program was used: Initial Temp.: 170 0C; Initial time: 10 min; Rate: 10 °C/min; Final Temp.: 280 0C; Final time: 10 min. Injection volume: 4 μL (Agilent 7683 autosampler).
Standard solutions of compound (III): A stock solution of 100 μg/mL of compound (III) in toluene was prepared by weighing 10 mg of compound (III) in a
100 mL volumetric flask and dissolving with toluene. The stock solution of 100 μg/mL of compound (III) was diluted quantitatively with toluene to obtain solutions containing 0.6, 0.8, 1.0, 1.2, 1.5 and 2.0 μg/mL of compound (III).
Blank preparation: 2g of tartaric acid were dissolved with 10 mL of water. Then, 2 mL of toluene and Ig of sodium chloride were added. The mixture was stirred, the aqueous phase was removed, and the organic phase was injected into the GC apparatus.
Preparation of a reference standard solution preparation: 2g of tartaric acid were dissolved with 10 mL of water. Then, Ig of sodium chloride and 2 mL of a standard solution of compound (III) in toluene (i.e. solutions containing 0.60, 0.80, 1.0, 1.2, 1.5 and 2.0 μg/mL) were added. The mixture was stirred, the aqueous phase was removed, and the organic phase was injected into the GC apparatus. A linear calibration curve was determined using the six standard solution preparations [i.e. 0.6, 0.80, 1.0, 1.2, 1.5 and 2.0 μg/mL; which correspond to 300 ppb, 400 ppb, 500 ppb, 600 ppb, 750 ppb, and 1.000 ppb, of compound (III), respectively]. The obtained GC chromatogram for the preparation prepared using the standard solution containing 1.5 μg/mL of compound (III) is shown in Figure 1.
Test sample preparation: 2g of ranolazine were weighed with 2 g of tartaric acid and dissolved with 10 mL of water. Then, 2 mL of toluene and Ig of sodium chloride were added. The mixture was stirred, the aqueous phase was removed, and the organic phase was injected into the GC apparatus.
System suitability: a standard preparation prepared from a standard solution in toluene of compound (III) of 1.5 μg/mL was used as standard solution preparation for calculating the content of compound (III) in the samples of ranolazine. Alternatively, other standard preparations of compound (III) prepared from standard solutions having a concentration of (III) between 0.60 - 2.0 μg/mL can be used as standard solution preparation for calculating the content of compound (III) in the samples of ranolazine.
Approximate GC Retention Time for 2-(2-methoxyphenoxy)- methyloxirane (compound III): 7.2 minutes.
Calculation: The calculation of the content of compound (III) in the sample of ranolazine can be calculated using the equation below:
ri c s i nnn ppb of compound of formula (III) = — x X lUUU rs w where: ri = Area peak response of compound (III) in the Test sample preparation. rs = Area peak response of compound (III) in the standard solution preparation. cs = Concentration (μg/mL) of compound (III) in the standard solution of compound (III). w = Weight (g) of ranolazine in the Test sample preparation. 1000 = convention factor to convert the result to ppb. The limit of detection (LOD): 132 ppb of compound (III). Alternatively to the equation above, the content of compound (III) in the sample of ranolazine can be calculated automatically from the linear calibration curve determined above from the six standard solution preparations [i.e. 0.6, 0.80, 1.0,
1.2, 1.5 and 2.0 μg/mL; which correspond to 300 ppb, 400 ppb, 500 ppb, 600 ppb, 750 ppb, and 1.000 ppb, of compound (III), respectively].
HPLC Method:
The chromatographic separation was carried out in a Symmetry C 18, 5 μm, 250 x 4.6 mm LD column; at 37 0C.
The mobile phase A was prepared by mixing 297.5 g of acetonitrile with 1620 g of pH= 4.4 buffer, which was prepared from 1.87 g of ammonium acetate in 1620 mL of water adjusting the pH to 4.4 with glacial acetic acid. This mobile phase was mixed and filtered through 0.22 μm nylon filter under vacuum.
The mobile phase B was acetonitrile.
The chromatograph was equipped with a 230 nm detector and the flow rate was 1.0 mL per minute.
The chromatograph was programmed as follows: Initial 0-2 min. isocratic 100% mobile phase A, 2-16 min. linear gradient to 93% mobile phase A, 16-
24 min. isocratic 93% mobile phase A, 24-45 min. linear gradient to 80% mobile phase A, 45-62 min. isocratic 80% mobile phase A, 62-82 min. linear gradient to 100% mobile phase A and 82-95 min. equilibration with 100% mobile phase A.
Test samples were prepared by dissolving the appropriate amount of sample to obtain 5 mg per mL in mobile phase, and 20 μL were injected.
Approximate HPLC Retention Times:
Figure imgf000014_0001
Example 1: Preparation of Ranolazine free of potential genotoxicity. 30.56 g of ranolazine [24.07 g of estimated dry mass; HPLC purity:
99.8%; Compound III (GC Method): higher than 900 ppb (i.e. about 2.5 ppm)] was suspended in 103 mL of methyl ethyl ketone and the suspension was heated to 80 0C. The solution was cooled to 20-25 0C and kept at that temperature for 1 hour. The suspension was filtered, washed with 8.05 g (10 mL) of methyl ethyl ketone and dried in a vacuum oven at 50-60 0C till constant weight. 22.73 g of ranolazine was obtained [purification yield: 94.4%; HPLC purity: 99.91%; Compound III (GC method): 378 ppb (See Figure 2). Not detected by HPLC].
All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.
Preferred embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.

Claims

Claims:
1. A process for preparing ranolazine, compound of formula I,
Figure imgf000016_0001
which is free, or essentially free, of potential genotoxicity, said process comprising:
(i) providing ranolazine,
(ii) determining the concentration of compound (III),
Figure imgf000016_0002
in the ranolazine, and (iii) if the concentration of compound (III) in the ranolazine is higher than
750 ppb, reducing the residual amount of compound (III) in the ranolazine.
2. A process according to claim 1, wherein the providing ranolazine of step (i) comprises: (i)(a) synthesizing ranolazine directly, or
(i)(b) isolating ranolazine from a pharmaceutical composition comprising ranolazine.
3. A process according to any of claims 1 and 2, wherein the reducing the residual amount of compound (III) in the ranolazine of step (iii) comprises crystallizing or digesting the ranolazine.
4. A process according to claim 3, wherein the reducing the residual amount of compound (III) in the ranolazine of step (iii) comprises digesting the ranolazine in a non-hydroxylic organic solvent.
5. A process according to claim 4, wherein the digesting the ranolazine in a non- hydroxylic organic solvent comprises:
(iii) (a) suspending the ranolazine in a non-hydroxylic organic solvent, (iii) (b) optionally, heating the suspension, and
(iii) (c) removing the solvent from the suspension, preferably by filtering the suspension.
6. A process according to any of claims 4 and 5, wherein the non-hydroxylic organic solvent is a ketone solvent, and preferably is methyl ethyl ketone.
7. A process according to any of claims 1 to 6, said process further comprising an additional step of (iv) admixing the ranolazine with at least one pharmaceutically acceptable carrier, thereby obtaining a pharmaceutical composition comprising ranolazine which is free, or essentially free, of potential genotoxicity.
8. A process according to claim 7, wherein the at least one pharmaceutically acceptable carrier is methacylic acid-ethyl acrylate copolymer, microcrystalline cellulose, hypromellose, sodium hydroxide, magnesium stearate, titanium dioxide, polyethylene glycol, polysorbate 80, glycerol triacetate, lactose monohydrate, or mixtures thereof.
9. Ranolazine which is free, or essentially free, of potential genotoxicity, and wherein said ranolazine has a concentration of compound (III) of equal to or less than 750 ppb (w/w).
10. A pharmaceutical composition comprising ranolazine which is free, or essentially free, of potential genotoxicity, and wherein said pharmaceutical composition has a concentration of compound (III) of equal to or less than 750 ppb (w/w), relative to ranolazine.
11. A standard solution for the determination of potentially genotoxic impurities in samples of a ranolazine batch, wherein said standard solution comprises a solution of compound (III) in an organic solvent having a concentration between 0.60 - 2.00 μg/mL, and preferably having a concentration of 0.60 μg/mL, 0.80 μg/mL, 1.00 μg/mL, 1.20 μg/mL, 1.50 μg/mL, or 2.00 μg/mL.
12. The standard solution of claim 11 , wherein the organic solvent is toluene.
13. Use of compound (III) as a reference standard and/or marker in the determination of potentially genotoxic impurities in a sample of a ranolazine batch.
14. An analytical method for the determination of potentially genotoxic impurities in samples of a ranolazine batch, characterized in that said method can detect at least a concentration equal to or less than 750 ppb of compound (III) relative to ranolazine (w/w), said method comprising: (i) preparing a test sample by extracting the ranolazine from the sample of a ranolazine batch containing an unknown amount of compound (III), to obtain a solution in an organic solvent having an unknown amount of compound (III) and free, or substantially free, of ranolazine, (ii) measuring by a suitable GC apparatus the test sample of step (i) to obtain a GC chromatogram, wherein measuring comprises comparing the height and/or area of the peak corresponding to the impurity, and
(iii) determining the concentration of compound (III) in the sample of the ranolazine batch.
15. An analytical method according to claim 14, wherein the preparing a test sample of step (i) further comprises:
(i) (a) treating the sample of a ranolazine batch containing an unknown amount of compound (III) with a suitable amount of an acidic compound in the presence of water and a water non-miscible organic solvent, to obtain an aqueous / organic biphasic system comprising an aqueous phase containing the acid addition salt of ranolazine and an organic phase having an unknown amount of compound (III) and free, or essentially free, of ranolazine, and (i) (b) removing the aqueous phase from the mixture, thereby obtaining a test sample solution in a water non-miscible organic solvent having an unknown amount of compound (III) and free, or essentially free, of ranolazine.
16. An analytical method according to claim 15, wherein the water non-miscible organic solvent is at least one of the group consisting of pentyl acetate, anisole, benzaldehyde, benzene, bromobenzene, butyl acetate, butyl ether, chlorobenzene, chloroform, cyclohexane, cyclohexanone, cyclopentane, 1 ,2-dichlorobenzene, 1,2- dichloroethane, dichloromethane, diethoxymethane, diisobutyl ketone, dimethoxymethane, ethyl acetate, ethylbenzene, 1 ,2-diethoxyethane, ethyl ether, phosphoral, n-heptane, n-hexane, isobutyl acetate, isopropyl acetate, isopropyl ether, methyl acetate, methyl tert-butyl ether, methyl cyclohexane, methyl ethyl ketone, methyl formate, methyl isobutyl ketone, nitrobenzene, n-pentane, 3-pentanone, propyl acetate, propylene carbonate, l-methoxy-2-propanol acetate, propylene oxide, tetrachloroethylene, toluene, 1,1,1-trichloroethane, trichloroethylene, xylene, and mixtures thereof, preferably is at least one of the group consisting of ethyl acetate, isopropyl acetate, methyl tert-butyl ether, methyl ethyl ketone, methyl isobutyl ketone, toluene, xylene, and mixtures thereof, and more preferably is toluene.
17. An analytical method according to any of claims 15 and 16, wherein the acidic compound is an organic or an inorganic acid compound, more preferably is an organic acid compound, and more preferably is tartaric acid.
18. An analytical method according to any of claims 14 to 17, wherein the suitable GC apparatus of step (ii) comprises a column having a polysiloxane as stationary phase, and preferably comprises column having 5% phenyl 95% dimethyl polysiloxane as stationary phase.
19. A process according to any of claims 1 to 8, wherein the determining the concentration of compound (III) in ranolazine of step (ii) comprises carrying out an analytical method as defined in any of claims 14 to 18.
PCT/EP2010/057315 2009-05-27 2010-05-27 A piperazine derivative free, or essentially free, of potential genotoxicity, and a process for preparing the same WO2010136522A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US18160709P 2009-05-27 2009-05-27
US61/181,607 2009-05-27

Publications (2)

Publication Number Publication Date
WO2010136522A2 true WO2010136522A2 (en) 2010-12-02
WO2010136522A3 WO2010136522A3 (en) 2011-02-03

Family

ID=43216709

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2010/057315 WO2010136522A2 (en) 2009-05-27 2010-05-27 A piperazine derivative free, or essentially free, of potential genotoxicity, and a process for preparing the same

Country Status (1)

Country Link
WO (1) WO2010136522A2 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106568862A (en) * 2016-11-04 2017-04-19 万全万特制药江苏有限公司 Method for separation and determination of vortioxetine hydrobromide intermediate related substances by liquid chromatography
CN113466371A (en) * 2021-06-30 2021-10-01 黔西县黔希煤化工投资有限责任公司 Method for measuring trace diisopropyl ether in phenol water
CN113941003A (en) * 2021-10-25 2022-01-18 江苏集萃新型药物制剂技术研究所有限公司 Multimeric compositions, pharmaceutical formulations, compositions and methods of preparation
CN114315632A (en) * 2021-01-05 2022-04-12 海南鑫开源医药科技有限公司 Preparation method of ranolazine intermediate impurity

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008139492A2 (en) * 2007-05-15 2008-11-20 Natco Pharma Limited A process for the preparation of highly pure ranolazine base
WO2010025370A2 (en) * 2008-08-28 2010-03-04 Dr. Reddy's Laboratories Ltd. Preparation of ranolazine
WO2010023687A2 (en) * 2008-08-28 2010-03-04 Shodhana Laboratories Limited Preparation of ranolazine, its salts and intermediates thereof

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008139492A2 (en) * 2007-05-15 2008-11-20 Natco Pharma Limited A process for the preparation of highly pure ranolazine base
WO2010025370A2 (en) * 2008-08-28 2010-03-04 Dr. Reddy's Laboratories Ltd. Preparation of ranolazine
WO2010023687A2 (en) * 2008-08-28 2010-03-04 Shodhana Laboratories Limited Preparation of ranolazine, its salts and intermediates thereof

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106568862A (en) * 2016-11-04 2017-04-19 万全万特制药江苏有限公司 Method for separation and determination of vortioxetine hydrobromide intermediate related substances by liquid chromatography
CN114315632A (en) * 2021-01-05 2022-04-12 海南鑫开源医药科技有限公司 Preparation method of ranolazine intermediate impurity
CN113466371A (en) * 2021-06-30 2021-10-01 黔西县黔希煤化工投资有限责任公司 Method for measuring trace diisopropyl ether in phenol water
CN113466371B (en) * 2021-06-30 2023-09-08 黔西县黔希煤化工投资有限责任公司 Method for determining trace diisopropyl ether in phenolic water
CN113941003A (en) * 2021-10-25 2022-01-18 江苏集萃新型药物制剂技术研究所有限公司 Multimeric compositions, pharmaceutical formulations, compositions and methods of preparation

Also Published As

Publication number Publication date
WO2010136522A3 (en) 2011-02-03

Similar Documents

Publication Publication Date Title
US7294735B2 (en) Purification of cinacalcet
EP1761478B1 (en) An isolated atomoxetine impurity, processes for the preparation of atomoxetine impurities and their use as reference standards
CA2692866A1 (en) A method for analysing memantine, or a pharmaceutically acceptable salt thereof, and its impurities using gas chromatography
Jastrzębska et al. A new derivatization reagent for determination of biogenic amines in wines
WO2011012659A2 (en) Diethyl 4-(4-fluorobenzylamino)-1,2-phenylenedicarbamate, and salts thereof
WO2010136522A2 (en) A piperazine derivative free, or essentially free, of potential genotoxicity, and a process for preparing the same
EP3147282B1 (en) Optical resolution method of lenalidomide
AU2022203369B2 (en) Analogs of deutetrabenazine, their preparation and use
Chen et al. High-performance liquid chromatographic analysis of zolmitriptan in human plasma using fluorescence detection
CN104133010B (en) High performance liquid chromatography separation analysis Asimadoline intermediate and optical isomer
US10725057B2 (en) Method for pretreatment and method for analysis of lenalidomide in biological sample
Bakheit et al. Bisoprolol: A comprehensive profile
JP2012507497A (en) HPLC analysis methods for bosentan and related substances, and use of these substances as reference standards or markers
Djordjevic et al. Liquid chromatographic-mass spectrometric method for the determination of fluoxetine and norfluoxetine in human plasma: application to clinical study
WO2011064574A1 (en) Hplc method for detecting lenalidomide
RU2609807C2 (en) 5,6,7,8-tetrahydro-6-[n,n-bis[(2-thienyl)ethyl]]amino-1-naphthol and preparation method and use thereof
CN109521102A (en) Method of separating and assaying of the hydrobromic acid Vortioxetine finished product in relation to substance
Li et al. Determination of phenazopyridine in human plasma by GC—MS and its pharmacokinetics
Liu et al. Cleaning verification assays for highly potent compounds by high performance liquid chromatography mass spectrometry: Strategy, validation, and long-term performance
Asiri et al. Donepezil
CN114814060B (en) Detection method of valsartan amlodipine tablet related substances
Maibaum Indirect high-performance liquid chromatographic resolution of racemic tertiary amines as their diastereomeric urea derivatives after N-dealkylation
Yasui-Furukori et al. Determination of fluvoxamine and its metabolite fluvoxamino acid by liquid–liquid extraction and column-switching high-performance liquid chromatography
Kumar et al. Fexofenadine hydrochloride
Lindner et al. Enantioselective drug monitoring of (R)-and (S)-propranolol in human plasma via derivatization with optically active (R, R)-O, O-diacetyl tartaric acid anhydride

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: 10720785

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase in:

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2010720785

Country of ref document: EP

122 Ep: pct application non-entry in european phase

Ref document number: 10720785

Country of ref document: EP

Kind code of ref document: A2