US20040044038A1 - Polymorphic form XVI of fexofenadine hydrochloride - Google Patents

Polymorphic form XVI of fexofenadine hydrochloride Download PDF

Info

Publication number
US20040044038A1
US20040044038A1 US10/459,688 US45968803A US2004044038A1 US 20040044038 A1 US20040044038 A1 US 20040044038A1 US 45968803 A US45968803 A US 45968803A US 2004044038 A1 US2004044038 A1 US 2004044038A1
Authority
US
United States
Prior art keywords
fexofenadine hydrochloride
methanol
fexofenadine
solution
crystalline
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US10/459,688
Inventor
Barnaba Krochmal
Dov Diller
Ben-Zion Dolitzky
Judith Aronhime
Shlomit Wizel
Boaz Gome
Igor Lifshitz
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Teva Pharmaceutical Industries Ltd
Teva Pharmaceuticals USA Inc
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US10/459,688 priority Critical patent/US20040044038A1/en
Assigned to TEVA PHARMACEUTICALS USA, INC. reassignment TEVA PHARMACEUTICALS USA, INC. ASSIGNMENT OF RIGHTS IN BARBADOS Assignors: TEVA PHARMACEUTICAL INDUSTRIES LTD.
Assigned to TEVA PHARMACEUTICAL INDUSTRIES LTD. reassignment TEVA PHARMACEUTICAL INDUSTRIES LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WIZEL, SHLOMIT, ARONHIME, JUDITH, DOLITZKY, BEN-ZION, KROCHMAL, BARNADA, DILLER, DOV, GOME, BOAZ, LIFSHITZ, IGOR
Publication of US20040044038A1 publication Critical patent/US20040044038A1/en
Priority to US11/243,496 priority patent/US7671071B2/en
Priority to US12/208,768 priority patent/US20090054486A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/08Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms
    • C07D211/18Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with substituted hydrocarbon radicals attached to ring carbon atoms
    • C07D211/20Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with substituted hydrocarbon radicals attached to ring carbon atoms with hydrocarbon radicals, substituted by singly bound oxygen or sulphur atoms
    • C07D211/22Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hydrocarbon or substituted hydrocarbon radicals directly attached to ring carbon atoms with substituted hydrocarbon radicals attached to ring carbon atoms with hydrocarbon radicals, substituted by singly bound oxygen or sulphur atoms by oxygen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • the present invention relates to the solid state chemistry of fexofenadine hydrochloride and its use as an active pharmaceutical agent.
  • fexofenadine can be prepared starting from ethyl ⁇ , ⁇ -dimethylphenyl acetate and 4-chlorobutyroyl chloride, which are reacted under Freidel-Crafts conditions.
  • Chloride is displaced from the Freidel-Crafts product with ⁇ , ⁇ -diphenyl-4-piperidinemethanol to give 4-[4-[4-(hydroxydiphenylmethyl)-1-piperidinyl]-1-oxobutyl]- ⁇ , ⁇ -dimethylbenzeneacetate, which is isolated as its hydrochloride salt.
  • the ketone is then reduced with PtO/H 2 and the ester group is hydrolyzed to yield fexofenadine hydrochloride.
  • the present invention relates to the solid state physical properties, i.e., polymorphism, of fexofenadine hydrochloride. These properties may be influenced by controlling the conditions under which fexofenadine hydrochloride is obtained in solid form.
  • Solid state physical properties include, for example, the flowability of the milled solid. Flowability affects the ease with which the material is handled during processing into a pharmaceutical product. When particles of the powdered compound do not flow past each other easily, a formulation specialist must take that fact into account when developing a tablet or capsule formulation, which may necessitate the use of glidants such as colloidal silicon dioxide, talc, starch or tribasic calcium phosphate.
  • Another important solid state property of a pharmaceutical compound is its rate of dissolution in aqueous fluid.
  • the rate of dissolution of an active ingredient in a patient's stomach fluid may have therapeutic consequences because it imposes an upper limit on the rate at which an orally-administered active ingredient may reach the bloodstream.
  • the rate of dissolution is also a consideration in formulating syrups, elixirs and other liquid medicaments.
  • the solid state form of a compound may also affect its behavior on compaction and its storage stability.
  • polymorphic form may give rise to thermal behavior different from that of the amorphous material or another polymorphic form. Thermal behavior is measured in the laboratory by such techniques as capillary melting point, thermogravimetric analysis (TGA) and differential scanning calorimetry (DSC), and may be used to distinguish some polymorphic forms from others.
  • TGA thermogravimetric analysis
  • DSC differential scanning calorimetry
  • a particular polymorphic form may also give rise to distinct properties that may be detectable by powder X-ray diffraction, solid state 13C NMR spectrometry and infrared spectrometry.
  • Form II is reported to have a capillary melting point range of 100-105° C., a DSC endotherm with onset between 124126° C. and a PXRD pattern with d-spacings of 7.8, 6.4, 5.2, 4.9, 4.7, 4.4, 4.2, 4.1, 3.7, 3.6, 3.5 ⁇ .
  • Form III is reported to have a capillary melting point range of 166-171° C., a DSC endotherm with onset at 166° C. and a PXRD pattern with d-spacings of 8.95, 4.99, 4.88, 4.75, 4.57, 4.47, 4.46, 3.67, 3.65 ⁇ .
  • Form IV is reported to undergo decomposition at 115-116° C. In the general written description, a DSC endotherm with onset at 146° C. is reported.
  • Form IV is reported as having a PXRD pattern with d-spacings of 10.38, 6.97, 6.41, 5.55, 5.32, 5.23, 5.11, 4.98, 4.64, 4.32, 4.28, 4.12, 4.02, 3.83, 3.65, 3.51, 3.46 and 2.83 A.
  • the '872 patent discusses methods of interconverting Forms I-IV.
  • Aqueous recrystallization of Form I can be used to produce Form II.
  • Water-minimizing recrystallization or azeotropic distillation of either Form II or Form IV can yield Form I.
  • Form III is reported to be accessible by water minimizing recrystallization of Form II.
  • Crystal digestion of Form III can be used to obtain Form I.
  • Forms II and IV can be obtained directly by sodium borohydride reduction of 4-[4-[4-(hydroxydiphenylmethyl)-1-piperidinyl]-1-oxobutyl]- ⁇ , ⁇ -dimethylbenzeneacetate as described in Examples 1 and 2.
  • amorphous fexofenadine hydrochloride can be prepared by lyophilizing or spray drying a solution of fexofenadine hydrochloride.
  • the product is characterized by its IR spectrum and a featureless PXRD pattern.
  • Fexofenadine hydrochloride Forms V, VI, and VIII through XV are disclosed in US 20030021849 and US 20020177608 (WO02/080857), both of which are incorporated herein by reference.
  • the present invention provides a crystalline fexofenadine hydrochloride in the solid state characterized by data selected from the group consisting of a PXRD pattern with peaks at 10.1, 15.2, 18.6, 19.2, 20.1+0.2 degrees two theta; a DSC profile with two endothermic peaks at a temperature range of up to about 125° C. and an additional endotherm at a temperature of about 135° C.; and a TGA thermogram with a loss on drying (LOD) of about 6% to about 10% at a temperature range of up to about 145° C.
  • LOD loss on drying
  • the present invention provides pharmaceutical formulations of fexofenadine hydrochloride Form XVI and their methods of administration.
  • the present invention provides a process for preparing crystalline fexofenadine hydrochloride Form XVI comprising the steps of combining fexofenadine free base, HCl and methanol to obtain a solution, precipitating fexofenadine hydrochloride in the presence of methanol and recovering the fexofenadine hydrochloride.
  • the present invention provides a process for preparing crystalline fexofenadine hydrochloride Form XVI comprising the steps of combining fexofenadine base, HCl and methanol to obtain a solution, evaporating the methanol to obtain a residue, adding methanol and a C5 to C 12 hydrocarbon to the residue to precipitate fexofenadine hydrochloride and recovering the fexofenadine hydrochloride.
  • the present invention provides a process for preparing crystalline fexofenadine hydrochloride Form XVI comprising the steps of combining a solution of HCl in a mixture of methanol and isopropyl alcohol, with fexofenadine base, to obtain a solution, evaporating the methanol and the isopropyl alcohol to obtain a residue, adding a mixture of methanol and heptane to the residue to precipitate crystalline fexofenadine hydrochloride and recovering the fexofenadine hydrochloride.
  • the present invention provides a process for preparing crystalline fexofenadine hydrochloride Form XVI comprising the steps of combining fexofenadine free base, HCl and methanol to obtain a solution, removing the methanol to concentrate the solution, seeding the solution with fexofenadine hydrochloride Form XVI, stirring the solution, cooling the solution and recovering the fexofenadine hydrochloride.
  • the present invention provides a process for preparing fexofenadine hydrochloride Form XVI comprising the step of stirring a slurry of fexofenadine hydrochloride amorphous in methanol for a sufficient time to obtain fexofenadine hydrochloride Form XVI.
  • the present invention provides for a crystalline form of fexofenadine hydrochloride characterized by a PXRD pattern with peaks at 10.1, 15.2, 18.6, 19.2, 20.1 ⁇ 0.2, wherein the crystalline form has a water content of from about 6% to about 10%.
  • the present invention provides for a crystalline form of fexofenadine hydrochloride characterized by a PXRD pattern with peaks at 10.1, 15.2, 18.6, 19.2, 20.1 ⁇ 0.2, wherein the crystalline form with said PXRD peaks is substantially stable under storage at relative humidity of about 100% for at least about 1 week, and storage at about 40° C. and about a 75% relative humidity for at least about 6 months.
  • the present invention provides for a process for preparing a crystalline form of fexofenadine hydrochloride having a PXRD pattern with peaks at 10.1, 15.2, 18.6, 19.2, 20.1 ⁇ 0.2, comprising the steps of crystallizing the crystalline form with said PXRD peaks from a solution of fexofenadine hydrochloride in methanol and recovering the crystalline form.
  • FIG. 1 is a PXRD pattern for fexofenadine hydrochloride Form XVI.
  • FIG. 2 is DSC thermogram for fexofenadine hydrochloride Form XVI.
  • FIG. 3 is a TGA thermogram for fexofenadine hydrochloride Form XVI.
  • MTBE refers to methyl t-butyl ether (syn. t-butyl methyl ether).
  • Fexofenadine hydrochloride Form XVI is characterized by a PXRD pattern (FIG. 24) with peaks at 5.2, 10.1, 15.2, 15.5, 17.0, 17.3, 18.6, 19.2, 19.6, 20.1, 21.7, 22.5, 23.2, 24.0, 24.3, 25.6 ⁇ 0.2 degrees two theta. The most characteristic peaks are at 10.1, 15.2, 18.6, 19.2, 20.1+0.2 degrees two theta.
  • Fexofenadine hydrochloride Form XVI is also characterized by a DSC thermogram (FIG. 25) with two large endothermic peaks at a temperature range of up to about 125° C. and an additional small endotherm at a temperature of about 135° C.
  • the first endothermic peak ( ⁇ 7585J/g) is observed at a temperature of about 67° C.
  • the second endothermic peak is observed at a temperature of about 120° C.
  • the third endothermic peak ⁇ 0.3-2 J/g
  • the TGA thermogram of fexofenadine HCl Form XVI shows an LOD value of about 6% to about 10% in a temperature range of up to 145° C.
  • Fexofenadine hydrochloride Form XVI contains from about 6% to about 10% water by weight as measured by the Karl Fischer method. At the end of precipitation step, usually a Form XVI contains about 6% water by KF. But the form absorbs water, and its water content may increase to as much as 10% water by weight.
  • Fexofenadine hydrochloride Form XVI is substantially stable during storage.
  • Fexofenadine hydrochloride Form XVI is stable against transformation to other crystalline forms upon storage at relative humidity of up to about 100% for at least about 1 week, and storage at about 40° C. and about 75% relative humidity for at least about 6 months.
  • the conversion is preferably less than about 5%, more preferably less than about 2% by weight.
  • the present invention provides for processes for preparation of fexofenadine hydrochloride Form XVI, which allow preparing fexofenadine HCl form XVI substantially free of other polymorphic forms of fexofenadine HCl, including amorphous form.
  • substantially free refers to less than about 5% on a weight basis, preferably less than about 2%, weight of polymorphic forms other than Form XVI compared to the weight of all the polymorphic forms, including Form XVI.
  • a suitable method for determining the presence of other polymorphic forms is with X-Ray Powder diffraction.
  • the present invention provides for preparation of fexofenadine hydrochloride Form XVI by precipitation of the crystalline form from a methanol containing mixture.
  • a solution of fexofenadine hydrochloride in methanol is prepared.
  • fexofenadine hydrochloride Form XVI may be recovered in various manners, such as by precipitation from the solution (including concentration of the solution before precipitation); or removal of the methanol to obtain a residue, followed by precipitation from methanol, or precipitation from a mixture of methanol and an anti-solvent. Precipitation may be carried out from both a slurry and a solution. When a residue is added to methanol, with or without an anti-solvent, generally a slurry is obtained.
  • the present invention provides a process for preparing crystalline fexofenadine hydrochloride Form XVI comprising combining fexofenadine free base, HCl and methanol to obtain a solution, precipitating fexofenadine hydrochloride Form XVI from the solution, and recovering the fexofenadine hydrochloride Form XVI.
  • the present invention provides a process for preparing the crystalline fexofenadine hydrochloride Form XVI comprising the steps of combining fexofenadine free base with HCl to obtain a solution in methanol, removing the methanol to obtain a residue, adding methanol and a C 5 to a C 12 hydrocarbon to the residue to cause precipitation of fexofenadine hydrochloride and recovering the fexofenadine hydrochloride.
  • an anti-solvent such as a C 5 to a C 12 hydrocarbon is optional, i.e., the residue may only be taken up in methanol.
  • a solution of HCl in a mixture of methanol and a polar organic solvent is added to fexofenadine base, preferably at a temperature of about 0 to about 10° C.
  • An ice bath can be used to cool the solution.
  • the resulting solution can be filtered to remove impurities, including any material that does not go into solution.
  • Suitable polar organic solvents are protic and aprotic polar solvents such as alcohols, ketones, esters and ethers. Preferred solvents include acetone and isopropanol. Preferably, a small amount of the polar solvent relative to methanol is used.
  • the solvent is then removed to obtain a residue.
  • the solvent is removed by evaporation, more preferably under reduced pressure.
  • the temperature can be increased or the pressure reduced to accelerate the evaporation process.
  • the pressure is reduced by an oil pump to remove the solvent by evaporation.
  • Fexofenadine hydrochloride Form XVI is then crystallized from methanol, or a mixture of methanol and a suitable anti-solvent, such as a C 5 to a C 12 saturated or a monoaromatic hydrocarbon.
  • a suitable anti-solvent such as a C 5 to a C 12 saturated or a monoaromatic hydrocarbon.
  • hydrocarbons include heptane and hexane, with saturated hydrocarbons such as heptane being more preferred.
  • the ratio of methanol to the hydrocarbon is from about 1:3 to about 1:33 (v/v).
  • the resulting mixture is stirred.
  • the fexofenadine hydrochloride so recovered is then preferably dried at a temperature of about 50° C. to about 80° C., more preferably at a temperature of from about 60 to about 70° C., most preferable under reduced pressure. Both the wet and the dried samples are fexofenadine hydrochloride Form XVI.
  • fexofenadine hydrochloride Form XVI is prepared by concentrating the solution of fexofenadine hydrochloride in methanol before precipitation, preferably followed by seeding and cooling to precipitate Form XVI.
  • the solution is preferably concentrated to a level of about 2 to about 2.5 volumes of methanol in comparison to the weight of fexofenadine base (ml/g).
  • the fexofenadine HCl methanol solution may optionally be filtered in order to remove foreign particles.
  • the solution is stirred and cooled to enhance precipitation.
  • the resulting suspension may optionally be stirred, preferably at a low temperature (about minus 15 to about 10° C.) for a sufficient amount of time, preferably for at least about 20 minutes, to increase the yield.
  • the fexofenadine hydrochloride so recovered is then preferably dried at a temperature of about 50° C. to about 80° C., more preferably at a temperature of from about 60 to about 70° C., most preferable under reduced pressure. Both the wet and the dried samples are fexofenadine hydrochloride Form XVI.
  • the present invention provides for preparation fexofenadine hydrochloride Form XVI through stirring a slurry of amorphous fexofenadine hydrochloride in methanol.
  • An anti-solvent may optionally be added to the methanol.
  • the anti-solvent is a C 5 to C 12 hydrocarbon, more preferably a saturated hydrocarbon and most preferably heptane.
  • a small amount of methanol compared to heptane is used, more preferably from about 3% to about 26% volume of methanol compared to volume of heptane.
  • the slurry process is carried out for a sufficient time to obtain fexofenadine hydrochloride Form XVI.
  • the slurry process is carried out for at least about 5 hours, more preferably from at least about 10 hours and most preferably for at least about 15 hours.
  • the polymorphs of the present invention can be selectively obtained from fexofenadine hydrochloride generally through crystallization with different recrystallization solvent systems.
  • the starting material can be anhydrous fexofenadine hydrochloride or any fexofenadine hydrochloride hydrate or lower alcohol solvate.
  • the use of other solvates, such as the ethyl acetate solvate of the present invention, is not believed to interfere with the effectiveness of the process.
  • the starting fexofenadine hydrochloride can also be in an amorphous or any crystalline crystal form.
  • the process can be used as a purification method by using the desired form in an unacceptably pure state as starting material.
  • the conditions can also be changed to induce precipitation.
  • a preferred way of inducing precipitation is to reduce the solubility of the solvent.
  • the solubility of the solvent can be reduced, for example, by cooling the solvent.
  • an anti-solvent is added to a solution to decrease its solubility for a particular compound, thus resulting in precipitation.
  • an anti-solvent is added to an oily residue or a gummy material, wherein the low solubility of the anti-solvent for a particular compound results in precipitation of that compound.
  • Another manner to accelerate crystallization is by seeding with a crystal of the product or scratching the inner surface of the crystallization vessel with a glass rod. Other times, crystallization can occur spontaneously without any inducement.
  • the present invention encompasses both embodiments where precipitation happens spontaneously or is induced, unless if such inducement is critical for obtaining a particular polymorphic form of fexofenadine hydrochloride.
  • fexofenadine As an antihistamine, fexofenadine is effective at relieving symptoms caused by airborne and contact inducers of histamine release. Such substances include pollen, spores, animal dander, cockroach dander, industrial chemicals, dust and dust mites. Symptoms that can be alleviated by fexofenadine include bronchial spasms, sneezing, rhinorrhia, nasal congestion, lacrimation, redness, rash, urticaria and itch.
  • compositions of the present invention contain fexofenadine hydrochloride Form XVI, optionally in a mixture with other forms or amorphous fexofenadine and/or active ingredients such as pseudoephedrine. They can also be optionally mixed with pseudoephedrine.
  • active ingredients such as pseudoephedrine.
  • the pharmaceutical compositions of the present invention can contain one or more excipients. Excipients are added to the composition for a variety of purposes.
  • Diluents increase the bulk of a solid pharmaceutical composition and can make a pharmaceutical dosage form containing the composition easier for the patient and care giver to handle.
  • Diluents for solid compositions include, for example, microcrystalline cellulose (e.g. Avicel'), microfine cellulose, lactose, starch, pregelitinized starch, calcium carbonate, calcium sulfate, sugar, dextrates, dextrin, dextrose, dibasic calcium phosphate dihydrate, tribasic calcium phosphate, kaolin, magnesium carbonate, magnesium oxide, maltodextrin, mannitol, polymethacrylates (e.g. Eudragit®), potassium chloride, powdered cellulose, sodium chloride, sorbitol and talc.
  • microcrystalline cellulose e.g. Avicel'
  • microfine cellulose lactose
  • starch pregelitinized starch
  • calcium carbonate calcium sulfate
  • sugar dextrates
  • Solid pharmaceutical compositions that are compacted into a dosage form like a tablet can include excipients whose functions include helping to bind the active ingredient and other excipients together after compression.
  • Binders for solid pharmaceutical compositions include acacia, alginic acid, carbomer (e.g. carbopol), carboxymethylcellulose sodium, dextrin, ethyl cellulose, gelatin, guar gum, hydrogenated vegetable oil, hydroxyethyl cellulose, hydroxypropyl cellulose (e.g. Klucel®), hydroxypropyl methyl cellulose (e.g. Methocel®), liquid glucose, magnesium aluminum silicate, maltodextrin, methylcellulose, polymethacrylates, povidone (e.g. Kollidon®, Plasdone®), pregelatinized starch, sodium alginate and starch.
  • carbomer e.g. carbopol
  • carboxymethylcellulose sodium, dextrin ethyl cellulose
  • gelatin guar
  • the dissolution rate of a compacted solid pharmaceutical composition in the patient's stomach can be increased by the addition of a disintegrant to the composition.
  • Disintegrants include alginic acid, carboxymethylcellulose calcium, carboxymethylcellulose sodium (e.g. Ac-DiSol®, Primellose®), colloidal silicon dioxide, croscarmellose sodium, crospovidone (e.g. Kollidon®, Polyplasdone®), guar gum, magnesium aluminum silicate, methyl cellulose, microcrystalline cellulose, polacrilin potassium, powdered cellulose, pregelatinized starch, sodium alginate, sodium starch glycolate (e.g. Explotab®) and starch.
  • alginic acid include alginic acid, carboxymethylcellulose calcium, carboxymethylcellulose sodium (e.g. Ac-DiSol®, Primellose®), colloidal silicon dioxide, croscarmellose sodium, crospovidone (e.g. Kollidon®, Polyplasdone®),
  • Glidants can be added to improve the flowability of non-compacted solid composition and improve the accuracy of dosing.
  • Excipients that can function as glidants include colloidal silicon dixoide, magnesium trisilicate, powdered cellulose, starch, talc and tribasic calcium phosphate.
  • a dosage form such as a tablet is made by compaction of a powdered composition
  • the composition is subjected to pressure from a punch and dye.
  • Some excipients and active ingredients have a tendency to adhere to the surfaces of the punch and dye, which can cause the product to have pitting and other surface irregularities.
  • a lubricant can be added to the composition to reduce adhesion and ease release of the product form the dye.
  • Lubricants include magnesium stearate, calcium stearate, glyceryl monostearate, glyceryl palmitostearate, hydrogenated castor oil, hydrogenated vegetable oil, mineral oil, polyethylene glycol, sodium benzoate, sodium lauryl sulfate, sodium stearyl fumarate, stearic acid, talc and zinc stearate.
  • Flavoring agents and flavor enhancers make the dosage form more palatable to the patient.
  • Common flavoring agents and flavor enhancers for pharmaceutical products that can be included in the composition of the present invention include maltol, vanillin, ethyl vanillin, menthol, citric acid, fumaric acid, ethyl maltol, and tartaric acid.
  • Solid and liquid compositions can also be dyed using any pharmaceutically acceptable colorant to improve their appearance and/or facilitate patient identification of the product and unit dosage level.
  • liquid pharmaceutical compositions of the present invention fexofenadine hydrochloride Form XVI and any other solid excipients are dissolved or suspended in a liquid carrier such as water, vegetable oil, alcohol, polyethylene glycol, propylene glycol or glycerin.
  • a liquid carrier such as water, vegetable oil, alcohol, polyethylene glycol, propylene glycol or glycerin.
  • Liquid pharmaceutical compositions can contain emulsifying agents to disperse uniformly throughout the composition an active ingredient or other excipient that is not soluble in the liquid carrier.
  • Emulsifying agents that can be useful in liquid compositions of the present invention include, for example, gelatin, egg yolk, casein, cholesterol, acacia, tragacanth, chondrus, pectin, methyl cellulose, carbomer, cetostearyl alcohol and cetyl alcohol.
  • Liquid pharmaceutical compositions of the present invention can also contain a viscosity enhancing agent to improve the mouth-feel of the product and/or coat the lining of the gastrointestinal tract.
  • a viscosity enhancing agent include acacia, alginic acid bentonite, carbomer, carboxymethylcellulose calcium or sodium, cetostearyl alcohol, methyl cellulose, ethylcellulose, gelatin guar gum, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, maltodextrin, polyvinyl alcohol, povidone, propylene carbonate, propylene glycol alginate, sodium alginate, sodium starch glycolate, starch tragacanth and xanthan gum.
  • Sweetening agents such as sorbitol, saccharin, sodium saccharin, sucrose, aspartame, fructose, mannitol and invert sugar can be added to improve the taste.
  • Preservatives and chelating agents such as alcohol, sodium benzoate, butylated hydroxy toluene, butylated hydroxyanisole and ethylenediamine tetraacetic acid can be added at levels safe for ingestion to improve storage stability.
  • a liquid composition according to the present invention can also contain a buffer such as guconic acid, lactic acid, citric acid or acetic acid, sodium guconate, sodium lactate, sodium citrate or sodium acetate.
  • a buffer such as guconic acid, lactic acid, citric acid or acetic acid, sodium guconate, sodium lactate, sodium citrate or sodium acetate.
  • the solid compositions of the present invention include powders, granulates, aggregates and compacted compositions.
  • the dosages include dosages suitable for oral, buccal, rectal, parenteral (including subcutaneous, intramuscular, and intravenous), inhalant and ophthalmic administration. Although the most suitable route in any given case will depend on the nature and severity of the condition being treated, the most preferred route of the present invention is oral.
  • the dosages can be conveniently presented in unit dosage form and prepared by any of the methods well-known in the pharmaceutical arts.
  • Dosage forms include solid dosage forms like tablets, powders, capsules, suppositories, sachets, troches and losenges as well as liquid syrups, suspensions and elixirs.
  • a dosage form of the present invention is a capsule containing the composition, preferably a powdered or granulated solid composition of the invention, within either a hard or soft shell.
  • the shell can be made from gelatin and optionally contain a plasticizer such as glycerin and sorbitol, and an opacifying agent or colorant.
  • compositions and dosage forms can be formulated into compositions and dosage forms according to methods known in the art.
  • a composition for tableting or capsule filing can be prepared by wet granulation.
  • wet granulation some or all of the active ingredients and excipients in powder form are blended and then further mixed in the presence of a liquid, typically water, which causes the powders to clump up into granules.
  • the granulate is screened and/or milled, dried and then screened and/or milled to the desired particle size.
  • the granulate can then be tableted or other excipients can be added prior to tableting, such as a glidant and/or a lubricant.
  • a tableting composition can be prepared conventionally by diy blending.
  • the blended composition of the actives and excipients can be compacted into a slug or a sheet and then comminuted into compacted granules.
  • the compacted granules can be compressed subsequently into a tablet.
  • a blended composition can be compressed directly into a compacted dosage form using direct compression techniques.
  • Direct compression produces a more uniform tablet without granules.
  • Excipients that are particularly well-suited to direct compression tableting include microcrystalline cellulose, spray dried lactose, dicalcium phosphate dihydrate and colloidal silica. The proper use of these and other excipients in direct compression tableting is known to those in the art with experience and skill in particular formulation challenges of direct compression tableting.
  • a capsule filling of the present invention can comprise any of the aforementioned blends and granulates that were described with reference to tableting, only they are not subjected to a final tableting step.
  • Capsules, tablets and lozenges and other unit dosage forms preferably contain a dosage level of about 30 to about 180 mg of fexofenadine hydrochloride. Other dosages may also be administered depending on the need.
  • a round standard aluminum sample holder with a round zero background quartz plate was used. Scans were performed over a range of 2 to 40 degrees two-theta, continuously, with a scan rate of 3 degrees/min.
  • the DSC thermogram was obtained using a DSC Mettler 821 Star.
  • the temperature range of scans was 30-350° C. at a rate of 10° C./min.
  • the weight of the sample was 2-5 mg.
  • the sample was purged with nitrogen gas at a flow rate of 40 mL/min. Standard 40 ⁇ l aluminum crucibles having lids with three small holes were used.
  • TGA thermogram for fexofenadine hydrochloride Form XVI was performed on Mettler TG50 using standard allumina pan and a sample weight: 7-15 mg.
  • HCl/IPA (1.6 ml) (6.05-6.24 N) was added to methanol (20 ml) and was cooled in an ice water bath. This solution was added to fexofenadine free base (5 grams) in a round bottom flask with a magnetic stirrer in an ice bath. The fexofenadine base dissolved immediately. The solution was filtered thru a glass fiber filter (GF/F), and the solvent evaporated off in a water bath at a temperature of 25° C. using a water aspirator, followed by a diaphragm pump, which was followed by an oil pump. Heptane (15 ml) was added.
  • GF/F glass fiber filter
  • Example 1 was repeated, except 4 ml of methanol was used in the crystallization step instead of 5 ml.
  • PXRD analysis confirmed presence of Form XVI of fexofenadine hydrochloride.
  • Example 1 was repeated, except 3 ml of methanol was used in the crystallization step instead of 5 ml.
  • PXRD analysis confirmed presence of Form XVI of fexofenadine hydrochloride.
  • Example 1 was repeated, except 2.5 ml of methanol was used in the crystallization step instead of 5 ml.
  • PXRD analysis confirmed presence of Form XVI of fexofenadine hydrochloride.
  • Fexofenadine free base (20 grams) was crushed and put into a 250 ml round bottom flask in an ice bath with a magnetic stirrer. HCVIPA (6.5 ml) was added to 80 ml methanol and cooled in an ice bath, and then added to the flask with mixing. After 15 minutes, the flask was filtered, and the filtrate evaporated off at room temperature first with a water aspirator then with a diaphragm pump and finally with an oil pump. The remaining material (5 grams) was stirred as a slurry overnight with a mixture of heptane (15 ml) and methanol (1.5 ml), filtered and dried for 1 hour at 65° C. under vacuum. PXRD analysis confirmed presence of Form XVI of fexofenadine hydrochloride.
  • Step 1 Preparation of HCl Gas Solution in Methanol
  • HCl gas was dissolved in cold methanol (T ⁇ 10° C.), until about 5% w/w concentration was achieved.
  • Step 2 Dilution of HCl/Methanol Solution
  • Step 3 Titration of Fexofenadine-Base With Diluted HCl/Methanol Solution
  • the solution was seeded with fexofenadine HCl Form XVI crystals, and then stirred for an additional 30 to 90 minutes.
  • the seeded solution was cooled and kept at a temperature of 0 to 10° C. for at least 4 hours, and the slurry was stirred for an additional 30 to 90 minutes.
  • the solid was separated from the mother liquor by filtration under reduced pressure.
  • the wet product was dried under reduce pressure at a temperature of 50-65° C.
  • Amorphous fexofenadine HCl (5 gr) was stirred in a mixture of heptane (15 ml) and methanol (1.5 mil) at room temperature. After stirring overnight, a solid was filtered and dried at 65° C. The PXRD pattern of the solid confirmed that the product was fexofenadine hydrochloride Form XVI.

Abstract

Provided is a crystalline (polymorphic) form of fexofenadine hydrochloride, denominated fexofenadine hydrochloride Form XVI.

Description

    CROSS REFERENCE TO RELATED APPLICATION
  • This application claims the benefit under 35 U.S.C. § 119(e) of provisional application Serial No. 60/387,972, filed Jun. 10, 2002, which is incorporated herein by reference.[0001]
  • FIELD OF THE INVENTION
  • The present invention relates to the solid state chemistry of fexofenadine hydrochloride and its use as an active pharmaceutical agent. [0002]
  • BACKGROUND OF THE INVENTION
  • 4-[4-[4-(hydroxydiphenylmethyl)-1-piperidinyl]-1-hydroxybutyl]-α,α-dimethylbenzeneacetic acid of formula (I) (fexofenadine) is an H[0003] 1 receptor antagonist and a useful antihistaminic drug. It has low permeability into central nervous system tissues and weak antimuscarinic activity, causing it to have few systemic side effects.
    Figure US20040044038A1-20040304-C00001
  • The antihistamic activity of fexofenadine is disclosed in U.S. Pat. No. 4,254,129, incorporated herein by reference. According to the '129 patent, fexofenadine can be prepared starting from ethyl α,α-dimethylphenyl acetate and 4-chlorobutyroyl chloride, which are reacted under Freidel-Crafts conditions. Chloride is displaced from the Freidel-Crafts product with α,α-diphenyl-4-piperidinemethanol to give 4-[4-[4-(hydroxydiphenylmethyl)-1-piperidinyl]-1-oxobutyl]-α,α-dimethylbenzeneacetate, which is isolated as its hydrochloride salt. The ketone is then reduced with PtO/H[0004] 2 and the ester group is hydrolyzed to yield fexofenadine hydrochloride.
  • Other methods of preparing fexofenadine are discussed in U.S. Pat. Nos. 5,578,610, 5,589,487, 5,581,011, 5,663,412, 5,750,703, 5,994,549, 5,618,940, 5,631375, 5,644,061, 5,650,516, 5,652,370, 5,654,433, 5,663,353, 5,675,009, 5,375,693 and 6,147,216. [0005]
  • The present invention relates to the solid state physical properties, i.e., polymorphism, of fexofenadine hydrochloride. These properties may be influenced by controlling the conditions under which fexofenadine hydrochloride is obtained in solid form. Solid state physical properties include, for example, the flowability of the milled solid. Flowability affects the ease with which the material is handled during processing into a pharmaceutical product. When particles of the powdered compound do not flow past each other easily, a formulation specialist must take that fact into account when developing a tablet or capsule formulation, which may necessitate the use of glidants such as colloidal silicon dioxide, talc, starch or tribasic calcium phosphate. [0006]
  • Another important solid state property of a pharmaceutical compound is its rate of dissolution in aqueous fluid. The rate of dissolution of an active ingredient in a patient's stomach fluid may have therapeutic consequences because it imposes an upper limit on the rate at which an orally-administered active ingredient may reach the bloodstream. The rate of dissolution is also a consideration in formulating syrups, elixirs and other liquid medicaments. The solid state form of a compound may also affect its behavior on compaction and its storage stability. [0007]
  • These practical physical characteristics are influenced by the conformation and orientation of molecules in the unit cell, which defines a particular polymorphic form of a substance. The polymorphic form may give rise to thermal behavior different from that of the amorphous material or another polymorphic form. Thermal behavior is measured in the laboratory by such techniques as capillary melting point, thermogravimetric analysis (TGA) and differential scanning calorimetry (DSC), and may be used to distinguish some polymorphic forms from others. A particular polymorphic form may also give rise to distinct properties that may be detectable by powder X-ray diffraction, solid state 13C NMR spectrometry and infrared spectrometry. [0008]
  • U.S. Pat. Nos. 5,738,872, 5,932,247 and 5,855,912, incorporated herein by reference, describe four crystal forms of fexofenadine hydrochloride which are designated Forms I-IV. According to the '872 and related patents, Forms II and IV are hydrates and Forms I and III are anhydrates. Each form is characterized by its melting point, onset of endotherm in the DSC profile, and PXRD. Form I is reported to have a capillary melting point range of 196-201° C., a DSC endotherm with onset between 195-199° C. and a powder X-ray diffraction (“PXRD”) pattern with d-spacings of 14.89, 11.85, 7.30, 6.28, 5.91, 5.55, 5.05, 4.96, 4.85, 4.57, 4.45, 3.94, 3.89, 3.84, 3.78, 3.72, 3.63, 3.07, 3.04, 2.45 Å. Form II is reported to have a capillary melting point range of 100-105° C., a DSC endotherm with onset between 124126° C. and a PXRD pattern with d-spacings of 7.8, 6.4, 5.2, 4.9, 4.7, 4.4, 4.2, 4.1, 3.7, 3.6, 3.5 Å. Form III is reported to have a capillary melting point range of 166-171° C., a DSC endotherm with onset at 166° C. and a PXRD pattern with d-spacings of 8.95, 4.99, 4.88, 4.75, 4.57, 4.47, 4.46, 3.67, 3.65 Å. In Example 2, Form IV is reported to undergo decomposition at 115-116° C. In the general written description, a DSC endotherm with onset at 146° C. is reported. Form IV is reported as having a PXRD pattern with d-spacings of 10.38, 6.97, 6.41, 5.55, 5.32, 5.23, 5.11, 4.98, 4.64, 4.32, 4.28, 4.12, 4.02, 3.83, 3.65, 3.51, 3.46 and 2.83 A. [0009]
  • The '872 patent discusses methods of interconverting Forms I-IV. Aqueous recrystallization of Form I can be used to produce Form II. Water-minimizing recrystallization or azeotropic distillation of either Form II or Form IV can yield Form I. Form III is reported to be accessible by water minimizing recrystallization of Form II. Crystal digestion of Form III can be used to obtain Form I. Forms II and IV can be obtained directly by sodium borohydride reduction of 4-[4-[4-(hydroxydiphenylmethyl)-1-piperidinyl]-1-oxobutyl]-α,α-dimethylbenzeneacetate as described in Examples 1 and 2. [0010]
  • International Publication No. WO 00/71124 A1, discloses that amorphous fexofenadine hydrochloride can be prepared by lyophilizing or spray drying a solution of fexofenadine hydrochloride. The product is characterized by its IR spectrum and a featureless PXRD pattern. [0011]
  • International Publication Nos. WO 01/94313 and WO 02/066429 are also directed to polymorphic forms of fexofenadine hydrochloride. [0012]
  • Fexofenadine hydrochloride Forms V, VI, and VIII through XV are disclosed in US 20030021849 and US 20020177608 (WO02/080857), both of which are incorporated herein by reference. [0013]
  • There is a need in the art for additional polymorphic forms of fexofenadine hydrochloride. [0014]
  • SUMMARY OF THE INVENTION
  • In one aspect, the present invention provides a crystalline fexofenadine hydrochloride in the solid state characterized by data selected from the group consisting of a PXRD pattern with peaks at 10.1, 15.2, 18.6, 19.2, 20.1+0.2 degrees two theta; a DSC profile with two endothermic peaks at a temperature range of up to about 125° C. and an additional endotherm at a temperature of about 135° C.; and a TGA thermogram with a loss on drying (LOD) of about 6% to about 10% at a temperature range of up to about 145° C. [0015]
  • In another aspect, the present invention provides pharmaceutical formulations of fexofenadine hydrochloride Form XVI and their methods of administration. [0016]
  • In another aspect, the present invention provides a process for preparing crystalline fexofenadine hydrochloride Form XVI comprising the steps of combining fexofenadine free base, HCl and methanol to obtain a solution, precipitating fexofenadine hydrochloride in the presence of methanol and recovering the fexofenadine hydrochloride. [0017]
  • In another aspect, the present invention provides a process for preparing crystalline fexofenadine hydrochloride Form XVI comprising the steps of combining fexofenadine base, HCl and methanol to obtain a solution, evaporating the methanol to obtain a residue, adding methanol and a C5 to C[0018] 12 hydrocarbon to the residue to precipitate fexofenadine hydrochloride and recovering the fexofenadine hydrochloride.
  • In another aspect, the present invention provides a process for preparing crystalline fexofenadine hydrochloride Form XVI comprising the steps of combining a solution of HCl in a mixture of methanol and isopropyl alcohol, with fexofenadine base, to obtain a solution, evaporating the methanol and the isopropyl alcohol to obtain a residue, adding a mixture of methanol and heptane to the residue to precipitate crystalline fexofenadine hydrochloride and recovering the fexofenadine hydrochloride. [0019]
  • In another aspect, the present invention provides a process for preparing crystalline fexofenadine hydrochloride Form XVI comprising the steps of combining fexofenadine free base, HCl and methanol to obtain a solution, removing the methanol to concentrate the solution, seeding the solution with fexofenadine hydrochloride Form XVI, stirring the solution, cooling the solution and recovering the fexofenadine hydrochloride. [0020]
  • In another aspect, the present invention provides a process for preparing fexofenadine hydrochloride Form XVI comprising the step of stirring a slurry of fexofenadine hydrochloride amorphous in methanol for a sufficient time to obtain fexofenadine hydrochloride Form XVI. [0021]
  • In another aspect, the present invention provides for a crystalline form of fexofenadine hydrochloride characterized by a PXRD pattern with peaks at 10.1, 15.2, 18.6, 19.2, 20.1±0.2, wherein the crystalline form has a water content of from about 6% to about 10%. [0022]
  • In another aspect, the present invention provides for a crystalline form of fexofenadine hydrochloride characterized by a PXRD pattern with peaks at 10.1, 15.2, 18.6, 19.2, 20.1±0.2, wherein the crystalline form with said PXRD peaks is substantially stable under storage at relative humidity of about 100% for at least about 1 week, and storage at about 40° C. and about a 75% relative humidity for at least about 6 months. [0023]
  • In another aspect, the present invention provides for a process for preparing a crystalline form of fexofenadine hydrochloride having a PXRD pattern with peaks at 10.1, 15.2, 18.6, 19.2, 20.1±0.2, comprising the steps of crystallizing the crystalline form with said PXRD peaks from a solution of fexofenadine hydrochloride in methanol and recovering the crystalline form.[0024]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 is a PXRD pattern for fexofenadine hydrochloride Form XVI. [0025]
  • FIG. 2 is DSC thermogram for fexofenadine hydrochloride Form XVI. [0026]
  • FIG. 3 is a TGA thermogram for fexofenadine hydrochloride Form XVI.[0027]
  • DETAILED DESCRIPTION OF THE INVENTION
  • As used herein, “MTBE” refers to methyl t-butyl ether (syn. t-butyl methyl ether). [0028]
  • In one aspect the present invention provides for fexofenadine hydrochloride Form XVI. Fexofenadine hydrochloride Form XVI is characterized by a PXRD pattern (FIG. 24) with peaks at 5.2, 10.1, 15.2, 15.5, 17.0, 17.3, 18.6, 19.2, 19.6, 20.1, 21.7, 22.5, 23.2, 24.0, 24.3, 25.6±0.2 degrees two theta. The most characteristic peaks are at 10.1, 15.2, 18.6, 19.2, 20.1+0.2 degrees two theta. [0029]
  • Fexofenadine hydrochloride Form XVI is also characterized by a DSC thermogram (FIG. 25) with two large endothermic peaks at a temperature range of up to about 125° C. and an additional small endotherm at a temperature of about 135° C. The first endothermic peak (≈7585J/g) is observed at a temperature of about 67° C., while the second endothermic peak (≈60J/g) is observed at a temperature of about 120° C., and the third endothermic peak (≈0.3-2 J/g) is observed at a temperature of about 135° C. [0030]
  • The TGA thermogram of fexofenadine HCl Form XVI shows an LOD value of about 6% to about 10% in a temperature range of up to 145° C. [0031]
  • Karl Fischer and elemental analysis of fexofenadine hydrochloride Form XVI point to a water content higher than an anhydrate. Fexofenadine hydrochloride Form XVI contains from about 6% to about 10% water by weight as measured by the Karl Fischer method. At the end of precipitation step, usually a Form XVI contains about 6% water by KF. But the form absorbs water, and its water content may increase to as much as 10% water by weight. [0032]
  • Fexofenadine hydrochloride Form XVI is substantially stable during storage. Fexofenadine hydrochloride Form XVI is stable against transformation to other crystalline forms upon storage at relative humidity of up to about 100% for at least about 1 week, and storage at about 40° C. and about 75% relative humidity for at least about 6 months. The conversion is preferably less than about 5%, more preferably less than about 2% by weight. [0033]
  • In another aspect, the present invention provides for processes for preparation of fexofenadine hydrochloride Form XVI, which allow preparing fexofenadine HCl form XVI substantially free of other polymorphic forms of fexofenadine HCl, including amorphous form. As used herein, “substantially free” refers to less than about 5% on a weight basis, preferably less than about 2%, weight of polymorphic forms other than Form XVI compared to the weight of all the polymorphic forms, including Form XVI. A suitable method for determining the presence of other polymorphic forms is with X-Ray Powder diffraction. [0034]
  • In another aspect, the present invention provides for preparation of fexofenadine hydrochloride Form XVI by precipitation of the crystalline form from a methanol containing mixture. In the first step, a solution of fexofenadine hydrochloride in methanol is prepared. Subsequently, fexofenadine hydrochloride Form XVI may be recovered in various manners, such as by precipitation from the solution (including concentration of the solution before precipitation); or removal of the methanol to obtain a residue, followed by precipitation from methanol, or precipitation from a mixture of methanol and an anti-solvent. Precipitation may be carried out from both a slurry and a solution. When a residue is added to methanol, with or without an anti-solvent, generally a slurry is obtained. [0035]
  • In one embodiment, the present invention provides a process for preparing crystalline fexofenadine hydrochloride Form XVI comprising combining fexofenadine free base, HCl and methanol to obtain a solution, precipitating fexofenadine hydrochloride Form XVI from the solution, and recovering the fexofenadine hydrochloride Form XVI. [0036]
  • In another embodiment, the present invention provides a process for preparing the crystalline fexofenadine hydrochloride Form XVI comprising the steps of combining fexofenadine free base with HCl to obtain a solution in methanol, removing the methanol to obtain a residue, adding methanol and a C[0037] 5 to a C12 hydrocarbon to the residue to cause precipitation of fexofenadine hydrochloride and recovering the fexofenadine hydrochloride. The addition of an anti-solvent such as a C5 to a C12 hydrocarbon is optional, i.e., the residue may only be taken up in methanol.
  • In one embodiment, a solution of HCl in a mixture of methanol and a polar organic solvent is added to fexofenadine base, preferably at a temperature of about 0 to about 10° C. An ice bath can be used to cool the solution. The resulting solution can be filtered to remove impurities, including any material that does not go into solution. [0038]
  • Suitable polar organic solvents are protic and aprotic polar solvents such as alcohols, ketones, esters and ethers. Preferred solvents include acetone and isopropanol. Preferably, a small amount of the polar solvent relative to methanol is used. [0039]
  • The solvent is then removed to obtain a residue. Preferably, the solvent is removed by evaporation, more preferably under reduced pressure. The temperature can be increased or the pressure reduced to accelerate the evaporation process. Preferably the pressure is reduced by an oil pump to remove the solvent by evaporation. [0040]
  • Fexofenadine hydrochloride Form XVI is then crystallized from methanol, or a mixture of methanol and a suitable anti-solvent, such as a C[0041] 5 to a C12 saturated or a monoaromatic hydrocarbon. Examples of such hydrocarbons include heptane and hexane, with saturated hydrocarbons such as heptane being more preferred. Preferably the ratio of methanol to the hydrocarbon is from about 1:3 to about 1:33 (v/v). Preferably, the resulting mixture is stirred.
  • The fexofenadine hydrochloride so recovered is then preferably dried at a temperature of about 50° C. to about 80° C., more preferably at a temperature of from about 60 to about 70° C., most preferable under reduced pressure. Both the wet and the dried samples are fexofenadine hydrochloride Form XVI. [0042]
  • In another embodiment, fexofenadine hydrochloride Form XVI is prepared by concentrating the solution of fexofenadine hydrochloride in methanol before precipitation, preferably followed by seeding and cooling to precipitate Form XVI. In this embodiment, the solution is preferably concentrated to a level of about 2 to about 2.5 volumes of methanol in comparison to the weight of fexofenadine base (ml/g). The fexofenadine HCl methanol solution may optionally be filtered in order to remove foreign particles. [0043]
  • In a preferred embodiment, after the seeding step, the solution is stirred and cooled to enhance precipitation. After precipitation, the resulting suspension may optionally be stirred, preferably at a low temperature (about minus 15 to about 10° C.) for a sufficient amount of time, preferably for at least about 20 minutes, to increase the yield. [0044]
  • The fexofenadine hydrochloride so recovered is then preferably dried at a temperature of about 50° C. to about 80° C., more preferably at a temperature of from about 60 to about 70° C., most preferable under reduced pressure. Both the wet and the dried samples are fexofenadine hydrochloride Form XVI. [0045]
  • In another embodiment, the present invention provides for preparation fexofenadine hydrochloride Form XVI through stirring a slurry of amorphous fexofenadine hydrochloride in methanol. An anti-solvent may optionally be added to the methanol. [0046]
  • In one embodiment, the anti-solvent is a C[0047] 5 to C12 hydrocarbon, more preferably a saturated hydrocarbon and most preferably heptane. Preferably a small amount of methanol compared to heptane is used, more preferably from about 3% to about 26% volume of methanol compared to volume of heptane. The slurry process is carried out for a sufficient time to obtain fexofenadine hydrochloride Form XVI. Preferably the slurry process is carried out for at least about 5 hours, more preferably from at least about 10 hours and most preferably for at least about 15 hours.
  • One skilled in the art would appreciate that the polymorphs of the present invention can be selectively obtained from fexofenadine hydrochloride generally through crystallization with different recrystallization solvent systems. The starting material can be anhydrous fexofenadine hydrochloride or any fexofenadine hydrochloride hydrate or lower alcohol solvate. The use of other solvates, such as the ethyl acetate solvate of the present invention, is not believed to interfere with the effectiveness of the process. The starting fexofenadine hydrochloride can also be in an amorphous or any crystalline crystal form. The process can be used as a purification method by using the desired form in an unacceptably pure state as starting material. The processes of the present invention can also be practiced as the last step in the methods discussed in U.S. Pat. Nos. 5,578,610, 5,589,487, 5,581,011, 5,663,412, 5,750,703, 5,994,549, 5,618,940, 5,631375, 5,644,061, 5,650,516, 5,652,370, 5,654,433, 5,663,353, 5,675,009, 5,375,693 and 6,147,216 to prepare a novel polymorph of the present invention. [0048]
  • Many processes of the present invention involve crystallization out of a particular solvent. One skilled in the art would appreciate that the conditions concerning crystallization can be modified without affecting the form of the polymorph obtained. For example, when mixing fexofenadine hydrochloride or free base in a solvent to form a solution, warming of the mixture can be necessary to completely dissolve the starting material. If warming does not clarify the mixture, the mixture can be diluted or filtered. To filter, the hot mixture can be passed through paper, glass fiber or other membrane material, or a clarifying agent such as celite. Depending upon the equipment used and the concentration and temperature of the solution, the filtration apparatus may need to be preheated to avoid premature crystallization. [0049]
  • The conditions can also be changed to induce precipitation. A preferred way of inducing precipitation is to reduce the solubility of the solvent. The solubility of the solvent can be reduced, for example, by cooling the solvent. [0050]
  • In one embodiment, an anti-solvent is added to a solution to decrease its solubility for a particular compound, thus resulting in precipitation. In another embodiment, an anti-solvent is added to an oily residue or a gummy material, wherein the low solubility of the anti-solvent for a particular compound results in precipitation of that compound. [0051]
  • Another manner to accelerate crystallization is by seeding with a crystal of the product or scratching the inner surface of the crystallization vessel with a glass rod. Other times, crystallization can occur spontaneously without any inducement. The present invention encompasses both embodiments where precipitation happens spontaneously or is induced, unless if such inducement is critical for obtaining a particular polymorphic form of fexofenadine hydrochloride. [0052]
  • As an antihistamine, fexofenadine is effective at relieving symptoms caused by airborne and contact inducers of histamine release. Such substances include pollen, spores, animal dander, cockroach dander, industrial chemicals, dust and dust mites. Symptoms that can be alleviated by fexofenadine include bronchial spasms, sneezing, rhinorrhia, nasal congestion, lacrimation, redness, rash, urticaria and itch. [0053]
  • Fexofenadine hydrochloride Forms XVI useful for delivering fexofenadine to the gastrointestinal tract, mucus membranes, bloodstream and inflamed tissues of a patient suffering from inflammation caused by a histamine. They can be formulated into a variety of compositions for administration to humans and animals. [0054]
  • Pharmaceutical compositions of the present invention contain fexofenadine hydrochloride Form XVI, optionally in a mixture with other forms or amorphous fexofenadine and/or active ingredients such as pseudoephedrine. They can also be optionally mixed with pseudoephedrine. In addition to the active ingredient(s), the pharmaceutical compositions of the present invention can contain one or more excipients. Excipients are added to the composition for a variety of purposes. [0055]
  • Diluents increase the bulk of a solid pharmaceutical composition and can make a pharmaceutical dosage form containing the composition easier for the patient and care giver to handle. Diluents for solid compositions include, for example, microcrystalline cellulose (e.g. Avicel'), microfine cellulose, lactose, starch, pregelitinized starch, calcium carbonate, calcium sulfate, sugar, dextrates, dextrin, dextrose, dibasic calcium phosphate dihydrate, tribasic calcium phosphate, kaolin, magnesium carbonate, magnesium oxide, maltodextrin, mannitol, polymethacrylates (e.g. Eudragit®), potassium chloride, powdered cellulose, sodium chloride, sorbitol and talc. [0056]
  • Solid pharmaceutical compositions that are compacted into a dosage form like a tablet can include excipients whose functions include helping to bind the active ingredient and other excipients together after compression. Binders for solid pharmaceutical compositions include acacia, alginic acid, carbomer (e.g. carbopol), carboxymethylcellulose sodium, dextrin, ethyl cellulose, gelatin, guar gum, hydrogenated vegetable oil, hydroxyethyl cellulose, hydroxypropyl cellulose (e.g. Klucel®), hydroxypropyl methyl cellulose (e.g. Methocel®), liquid glucose, magnesium aluminum silicate, maltodextrin, methylcellulose, polymethacrylates, povidone (e.g. Kollidon®, Plasdone®), pregelatinized starch, sodium alginate and starch. [0057]
  • The dissolution rate of a compacted solid pharmaceutical composition in the patient's stomach can be increased by the addition of a disintegrant to the composition. Disintegrants include alginic acid, carboxymethylcellulose calcium, carboxymethylcellulose sodium (e.g. Ac-DiSol®, Primellose®), colloidal silicon dioxide, croscarmellose sodium, crospovidone (e.g. Kollidon®, Polyplasdone®), guar gum, magnesium aluminum silicate, methyl cellulose, microcrystalline cellulose, polacrilin potassium, powdered cellulose, pregelatinized starch, sodium alginate, sodium starch glycolate (e.g. Explotab®) and starch. [0058]
  • Glidants can be added to improve the flowability of non-compacted solid composition and improve the accuracy of dosing. Excipients that can function as glidants include colloidal silicon dixoide, magnesium trisilicate, powdered cellulose, starch, talc and tribasic calcium phosphate. [0059]
  • When a dosage form such as a tablet is made by compaction of a powdered composition, the composition is subjected to pressure from a punch and dye. Some excipients and active ingredients have a tendency to adhere to the surfaces of the punch and dye, which can cause the product to have pitting and other surface irregularities. A lubricant can be added to the composition to reduce adhesion and ease release of the product form the dye. Lubricants include magnesium stearate, calcium stearate, glyceryl monostearate, glyceryl palmitostearate, hydrogenated castor oil, hydrogenated vegetable oil, mineral oil, polyethylene glycol, sodium benzoate, sodium lauryl sulfate, sodium stearyl fumarate, stearic acid, talc and zinc stearate. [0060]
  • Flavoring agents and flavor enhancers make the dosage form more palatable to the patient. Common flavoring agents and flavor enhancers for pharmaceutical products that can be included in the composition of the present invention include maltol, vanillin, ethyl vanillin, menthol, citric acid, fumaric acid, ethyl maltol, and tartaric acid. [0061]
  • Solid and liquid compositions can also be dyed using any pharmaceutically acceptable colorant to improve their appearance and/or facilitate patient identification of the product and unit dosage level. [0062]
  • In liquid pharmaceutical compositions of the present invention, fexofenadine hydrochloride Form XVI and any other solid excipients are dissolved or suspended in a liquid carrier such as water, vegetable oil, alcohol, polyethylene glycol, propylene glycol or glycerin. [0063]
  • Liquid pharmaceutical compositions can contain emulsifying agents to disperse uniformly throughout the composition an active ingredient or other excipient that is not soluble in the liquid carrier. Emulsifying agents that can be useful in liquid compositions of the present invention include, for example, gelatin, egg yolk, casein, cholesterol, acacia, tragacanth, chondrus, pectin, methyl cellulose, carbomer, cetostearyl alcohol and cetyl alcohol. [0064]
  • Liquid pharmaceutical compositions of the present invention can also contain a viscosity enhancing agent to improve the mouth-feel of the product and/or coat the lining of the gastrointestinal tract. Such agents include acacia, alginic acid bentonite, carbomer, carboxymethylcellulose calcium or sodium, cetostearyl alcohol, methyl cellulose, ethylcellulose, gelatin guar gum, hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, maltodextrin, polyvinyl alcohol, povidone, propylene carbonate, propylene glycol alginate, sodium alginate, sodium starch glycolate, starch tragacanth and xanthan gum. [0065]
  • Sweetening agents such as sorbitol, saccharin, sodium saccharin, sucrose, aspartame, fructose, mannitol and invert sugar can be added to improve the taste. [0066]
  • Preservatives and chelating agents such as alcohol, sodium benzoate, butylated hydroxy toluene, butylated hydroxyanisole and ethylenediamine tetraacetic acid can be added at levels safe for ingestion to improve storage stability. [0067]
  • A liquid composition according to the present invention can also contain a buffer such as guconic acid, lactic acid, citric acid or acetic acid, sodium guconate, sodium lactate, sodium citrate or sodium acetate. [0068]
  • Selection of excipients and the amounts to use can be readily determined by the formulation scientist based upon experience and consideration of standard procedures and reference works in the field. [0069]
  • The solid compositions of the present invention include powders, granulates, aggregates and compacted compositions. The dosages include dosages suitable for oral, buccal, rectal, parenteral (including subcutaneous, intramuscular, and intravenous), inhalant and ophthalmic administration. Although the most suitable route in any given case will depend on the nature and severity of the condition being treated, the most preferred route of the present invention is oral. The dosages can be conveniently presented in unit dosage form and prepared by any of the methods well-known in the pharmaceutical arts. [0070]
  • Dosage forms include solid dosage forms like tablets, powders, capsules, suppositories, sachets, troches and losenges as well as liquid syrups, suspensions and elixirs. [0071]
  • A dosage form of the present invention is a capsule containing the composition, preferably a powdered or granulated solid composition of the invention, within either a hard or soft shell. The shell can be made from gelatin and optionally contain a plasticizer such as glycerin and sorbitol, and an opacifying agent or colorant. [0072]
  • The active ingredient and excipients can be formulated into compositions and dosage forms according to methods known in the art. [0073]
  • A composition for tableting or capsule filing can be prepared by wet granulation. In wet granulation some or all of the active ingredients and excipients in powder form are blended and then further mixed in the presence of a liquid, typically water, which causes the powders to clump up into granules. The granulate is screened and/or milled, dried and then screened and/or milled to the desired particle size. The granulate can then be tableted or other excipients can be added prior to tableting, such as a glidant and/or a lubricant. [0074]
  • A tableting composition can be prepared conventionally by diy blending. For instance, the blended composition of the actives and excipients can be compacted into a slug or a sheet and then comminuted into compacted granules. The compacted granules can be compressed subsequently into a tablet. [0075]
  • As an alternative to dry granulation, a blended composition can be compressed directly into a compacted dosage form using direct compression techniques. Direct compression produces a more uniform tablet without granules. Excipients that are particularly well-suited to direct compression tableting include microcrystalline cellulose, spray dried lactose, dicalcium phosphate dihydrate and colloidal silica. The proper use of these and other excipients in direct compression tableting is known to those in the art with experience and skill in particular formulation challenges of direct compression tableting. [0076]
  • A capsule filling of the present invention can comprise any of the aforementioned blends and granulates that were described with reference to tableting, only they are not subjected to a final tableting step. [0077]
  • Capsules, tablets and lozenges and other unit dosage forms preferably contain a dosage level of about 30 to about 180 mg of fexofenadine hydrochloride. Other dosages may also be administered depending on the need. [0078]
  • The following describes the instrumentation used by the present invention to characterize the new polymorphs. The PXRD patterns (such as that for fexofenadine HCl Form XVI) were obtained by methods known in the art using a Scintag X-ray powder diffractometer, a variable goniometer, an X-Ray tube with Cu target anode (Cu radiation λ=1.5418 Å) and a solid state detector. A round standard aluminum sample holder with a round zero background quartz plate was used. Scans were performed over a range of 2 to 40 degrees two-theta, continuously, with a scan rate of 3 degrees/min. [0079]
  • The DSC thermogram was obtained using a DSC Mettler 821 Star. The temperature range of scans was 30-350° C. at a rate of 10° C./min. The weight of the sample was 2-5 mg. The sample was purged with nitrogen gas at a flow rate of 40 mL/min. [0080] Standard 40 μl aluminum crucibles having lids with three small holes were used.
  • The TGA thermogram for fexofenadine hydrochloride Form XVI was performed on Mettler TG50 using standard allumina pan and a sample weight: 7-15 mg. [0081]
  • EXAMPLES
  • Example 1 [0082]
  • Preparation of Fexofenadine Hydrochloride Form XVI [0083]
  • HCl/IPA (1.6 ml) (6.05-6.24 N) was added to methanol (20 ml) and was cooled in an ice water bath. This solution was added to fexofenadine free base (5 grams) in a round bottom flask with a magnetic stirrer in an ice bath. The fexofenadine base dissolved immediately. The solution was filtered thru a glass fiber filter (GF/F), and the solvent evaporated off in a water bath at a temperature of 25° C. using a water aspirator, followed by a diaphragm pump, which was followed by an oil pump. Heptane (15 ml) was added. The stirrer was turned on, 5 ml of methanol was added and the slurry was stirred overnight. The next day it was filtered and dried in the vacuum oven for 2 hours at 65° C. PXRD analysis confirmed presence of Form XVI of fexofenadine hydrochloride. [0084]
  • KF=6.685% [0085]
  • Elemental analysis: C, 66.28%; H, 7-89%; Cl, 5.65% [0086]
  • Example 2
  • Preparation of Fexofenadine Hydrochloride Form XVI [0087]
  • Example 1 was repeated, except 4 ml of methanol was used in the crystallization step instead of 5 ml. PXRD analysis confirmed presence of Form XVI of fexofenadine hydrochloride. [0088]
  • KF=6.507% [0089]
  • Elemental analysis: C, 66.80%; H, 7.91%; Cl, 6.23% [0090]
  • Example 3
  • Preparation of Fexofenadine Hydrochloride Form XVI [0091]
  • Example 1 was repeated, except 3 ml of methanol was used in the crystallization step instead of 5 ml. PXRD analysis confirmed presence of Form XVI of fexofenadine hydrochloride. [0092]
  • KF=6.221% [0093]
  • Elemental analysis: C, 67.18%; H, 7.74% Cl, 6.35% [0094]
  • Example 4
  • Preparation of Fexofenadine Hydrochloride Form XVI Example 1 was repeated, except 2 ml of methanol was used in the crystallization step instead of 5 ml. PXRD analysis confirmed presence of Form XVI of fexofenadine hydrochloride. [0095]
  • KF=7.314% [0096]
  • Elemental analysis: C, 65.95%; H, 7.77%; Cl, 6.34% [0097]
  • Example 5
  • Preparation of Fexofenadine Hydrochloride Form XVI [0098]
  • Example 1 was repeated, except 2.5 ml of methanol was used in the crystallization step instead of 5 ml. PXRD analysis confirmed presence of Form XVI of fexofenadine hydrochloride. [0099]
  • KF=6.250% [0100]
  • Elemental analysis: C, 66.70%; H, 7.64%; Cl, 6.40% [0101]
  • Example 6
  • Preparation of Fexofenadine Hydrochloride Form XVI [0102]
  • Fexofenadine free base (20 grams) was crushed and put into a 250 ml round bottom flask in an ice bath with a magnetic stirrer. HCVIPA (6.5 ml) was added to 80 ml methanol and cooled in an ice bath, and then added to the flask with mixing. After 15 minutes, the flask was filtered, and the filtrate evaporated off at room temperature first with a water aspirator then with a diaphragm pump and finally with an oil pump. The remaining material (5 grams) was stirred as a slurry overnight with a mixture of heptane (15 ml) and methanol (1.5 ml), filtered and dried for 1 hour at 65° C. under vacuum. PXRD analysis confirmed presence of Form XVI of fexofenadine hydrochloride. [0103]
  • Example 7
  • Process for Preparation of Fexofenadine-HCl Form XVI by Crystallization from Methanol [0104]
  • Step 1: Preparation of HCl Gas Solution in Methanol [0105]
  • HCl gas was dissolved in cold methanol (T<10° C.), until about 5% w/w concentration was achieved. [0106]
  • Step 2: Dilution of HCl/Methanol Solution [0107]
  • Methanol/HCl solution (79.9 grams) (4.5 w/w) was diluted with 121.8 grams of methanol to obtain diluted HCl/methanol solution. [0108]
  • Step 3: Titration of Fexofenadine-Base With Diluted HCl/Methanol Solution [0109]
  • The diluted HCl/methanol solution was cooled (T<10° C.). Fexofendine base (50 grams) (1.88% H[0110] 2O) was reacted with the diluted HCl/methanol solution, to form a fexofenadine-HCl solution. The molar ratio between fexofenadine-base and HCl was 1:1.
  • Step 4: Removal of Particulate Matter [0111]
  • The fexofenadine-HCl solution was filtered under reduced pressure to remove particulate matter (foreign particles). [0112]
  • Alternative A—Step 5: Isolation of the Product [0113]
  • The clear solution was distilled under reduced pressure at a jacket temperature of not more than 40° C. until the ratio of the residual solvent in the reactor was 2 to 2.5 volumes vs. the weight of fexofenadine base (ml/g). [0114]
  • After the final solvent volume was reached, the solution was seeded with fexofenadine HCl Form XVI crystals, and then stirred for an additional 30 to 90 minutes. The seeded solution was cooled and kept at a temperature of 0 to 10° C. for at least 4 hours, and the slurry was stirred for an additional 30 to 90 minutes. The solid was separated from the mother liquor by filtration under reduced pressure. The wet product was dried under reduce pressure at a temperature of 50-65° C. [0115]
  • Alternative B—Step 5: Isolation of the Product [0116]
  • The clear solution was distilled under reduced pressure at a jacket temperature not more than 40° C. until there was no more distillate. Methanol in the ratio of 2 to 2.5 volumes vs. the weight of fexofenadine base was added to the reactor (ml/g), and the fexofenadine HCl in methanol mixture was heated to dissolution. After the final solvent volume was reached, the solution was seeded with fexofenadine HCl Form XVI crystals, and then stirred for an additional 30 to 90 minutes. The seeded solution was cooled and kept at a temperature of 0 to 10° C. for at least 4 hours, and the slurry was stirred for an additional 30 to 90 minutes. The solid was separated from the mother liquor by filtration under reduced pressure. The wet product was dried under reduce pressure at a temperature of 50-65° C. [0117]
  • Example 8
  • Preparation of Fexofenadine Hydrochloride Form XVI from Amorphous form [0118]
  • Amorphous fexofenadine HCl (5 gr) was stirred in a mixture of heptane (15 ml) and methanol (1.5 mil) at room temperature. After stirring overnight, a solid was filtered and dried at 65° C. The PXRD pattern of the solid confirmed that the product was fexofenadine hydrochloride Form XVI. [0119]
  • Having thus described the invention with reference to particular preferred embodiments and illustrated it with examples, those in the art will appreciate modifications to the invention as described and illustrated that do not depart from the spirit and scope of the invention as disclosed in the specification. All the references cited herein are incorporated by reference in their entirety. [0120]

Claims (54)

What is claimed is:
1. A crystalline fexofenadine hydrochloride in the solid state characterized by data selected from the group consisting of a PXRD pattern with peaks at 10.1, 15.2, 18.6, 19.2, 20.1+0.2 degrees two theta; a DSC profile with two endothermic peaks at a temperature range of up to about 125° C. and an additional endotherm at a temperature of about 135° C.; and a TGA thermogram with a loss on drying (LOD) of about 6% to about 10% at a temperature range of up to about 145° C.
2. The crystalline fexofenadine hydrochloride of claim 1, characterized by a DSC profile with two endothermic peaks at a temperature range of up to about 125° C. and an additional endotherm at a temperature of about 135° C.
3. The crystalline fexofenadine hydrochloride of claim 2, wherein one of the two endothermic peaks is at a temperature of about 67° C. and the other at a temperature of about 120° C.
4. The crystalline fexofenadine hydrochloride of claim 3, wherein the crystalline form is characterized by a DSC thermogram as substantially depicted in FIG. 2.
5. The crystalline fexofenadine hydrochloride of claim 1, characterized with a PXRD pattern with peaks at 10.1, 15.2, 18.6, 19.2, 20.1±0.2 degrees two theta.
6. The crystalline fexofenadine hydrochloride of claim 5, further characterized by a PXRD pattern with peaks at 5.2, 15.5, 17.0, 17.3, 19.6, 21.7, 22.5, 23.2, 24.0, 24.3, 25.6+0.2 degrees two theta.
7. The crystalline fexofenadine hydrochloride of claim 6, further characterized by a PXRD pattern as substantially depicted in FIG. 1
8. The crystalline form of claim 5, wherein the crystalline form is substantially free of other polymorphic forms of fexofenadine hydrochloride.
9. The crystalline form of claim 8, wherein the crystalline form contains less than about 2% by weight of other polymorphic forms of fexofenadine hydrochloride.
10. A pharmaceutical formulation comprising an effective amount of fexofenadine hydrochloride of claim 1 and a pharmaceutically acceptable excipient.
11. The pharmaceutical formulation of claim 10, further comprising pseudoephedrine hydrochloride as an adjuvant.
12. A method of inhibiting binding between an H1 receptor and histamine in a patient suffering from contraction of the bronchi, vasodilation, itching or other inflammation response to histamine comprising administering to the patient the pharmaceutical composition of claim 10.
13. A process for preparing the crystalline fexofenadine hydrochloride of claim 1 comprising the steps of
a) combining fexofenadine free base, HCl and methanol to obtain a solution;
b) precipitating fexofenadine hydrochloride of claim 1 in the presence of methanol; and
c) recovering the fexofenadine hydrochloride of claim 1.
14. The process of claim 13, further comprising a step of drying the fexofenadine hydrochloride recovered in step (c).
15. The process of claim 14, wherein the fexofenadine hydrochloride is recovered substantially free of other polymorphic forms of fexofenadine hydrochloride.
16. The process of claim 15, wherein the fexofenadine hydrochloride recovered contains less than about 2% of other polymorphic forms of fexofenadine hydrochloride.
17. The process of claim 13, wherein the crystalline form recovered has a PXRD pattern with peaks at 10.1, 15.2, 18.6, 19.2, 20.1±0.2 degrees two theta.
18. The process of claim 13, further comprising the step of seeding the solution with fexofenadine hydrochloride Form XVI.
19. The process of claim 13, wherein the solution further comprises a polar organic solvent.
20. The process of claim 19, wherein the polar organic solvent is selected from the group consisting of an ether, an alcohol, an ester, a ketone and mixtures thereof.
21. The process of claim 20, wherein the polar organic solvent is an alcohol.
22. The process of claim 21, wherein the alcohol is iso-propyl alcohol.
23. The process of claim 13, further comprising introducing an anti-solvent in step (b).
24. The process of claim 23, wherein the anti-solvent is a hydrocarbon.
25. The process of claim 24, wherein the hydrocarbon is saturated.
26. The process of claim 25, wherein the hydrocarbon is selected from the group consisting of heptane and hexane.
27. The process of claim 13, further comprising the step of reducing the solution of step (a) to a residue by removing the methanol and adding methanol to the residue.
28. The process of claim 27, further comprising adding an anti-solvent to the residue.
29. The process of claim 28, wherein the anti-solvent is a hydrocarbon.
30. The process of claim 29, wherein the hydrocarbon is saturated.
31. The process of claim 30, wherein the hydrocarbon is selected from the group consisting of heptane and hexane.
32. The process of claim 27, wherein the methanol is removed by evaporation.
33. The process of claim 13, further comprising the step of concentrating the solution before precipitation.
34. The process of claim 33, wherein the solution is concentrated to a level of about 2 to about 2.5 volumes of methanol in comparison to the weight of fexofenadine base (ml/g).
35. The process of claim 34, wherein the concentrating is carried out by evaporation.
36. The process of claim 33, further comprising the step of seeding the solution with fexofenadine hydrochloride Form XVI.
37. The process of claim 33, further comprising the step of stirring and cooling during step (b).
38. The process of claim 13, wherein the fexofenadine hydrochloride recovered is substantially free of other polymorphic forms of fexofenadine hydrochloride.
39. The process of claim 38, wherein the fexofenadine hydrochloride recovered contains less than about 2% of other polymorphic forms of fexofenadine hydrochloride.
40. The process of claim 13, further comprising the step of stirring the precipitate before the recovery step at a temperature of from about minus 15° C. to about 10° C. for a sufficient time to increase the yield.
41. A process for preparing crystalline fexofenadine hydrochloride of claim 1 comprising the steps of
a) combining fexofenadine base, HCl and methanol to obtain a solution,
b) evaporating the methanol to obtain a residue;
c) adding methanol and a C5 to C12 hydrocarbon to the residue to precipitate fexofenadine hydrochloride of claim 1; and
d) recovering the fexofenadine hydrochloride of claim 1.
42. The process of claim 41, wherein the hydrocarbon is heptane.
43. A process for preparing crystalline fexofenadine hydrochloride of claim 1 comprising the steps of:
a) combining a solution of HCl in a mixture of methanol and isopropyl alcohol, with fexofenadine base, to obtain a solution;
b) evaporating the methanol and the isopropyl alcohol to obtain a residue;
c) adding a mixture of methanol and heptane to the residue to precipitate crystalline fexofenadine hydrochloride of claim 1; and
d) recovering the fexofenadine hydrochloride of claim 1.
44. A process for preparing crystalline fexofenadine hydrochloride of claim 1 comprising the steps of
a) combining fexofenadine free base, HCl and methanol to obtain a solution;
b) removing the methanol to concentrate the solution;
c) seeding the solution with fexofenadine hydrochloride Form XVI;
d) stirring the solution;
e) cooling the solution; and
d) recovering the fexofenadine hydrochloride.
45. A process for preparing fexofenadine hydrochloride of claim 1, comprising the step of stirring a slurry of fexofenadine hydrochloride amorphous in methanol for a sufficient time to obtain fexofenadine hydrochloride of claim 1.
46. The process of claim 45, further comprising an anti-solvent in mixture with methanol.
47. The process of claim 46, wherein the antisolvent is a hydrocarbon.
48. The process of claim 47, wherein the hydrocarbon is a saturated hydrocarbon.
49. The process of claim 48, wherein the hydrocarbon is heptane.
50. The process of claim 47, wherein the ratio of methanol to the hydrocarbon is from about 3% to about 26% volume of methanol compared to volume of the hydrocarbon.
51. A crystalline form of fexofenadine hydrochloride characterized by a PXRD pattern with peaks at 10.1, 15.2, 18.6, 19.2, 20.1+0.2, wherein the crystalline form has a water content of from about 6% to about 10% by weight.
52. The crystalline form of claim 51, wherein the crystalline form has about 10% water by weight.
53. A crystalline form of fexofenadine hydrochloride characterized by a PXRD pattern with peaks at 10.1, 15.2, 18.6, 19.2, 20.1+0.2, wherein the crystalline form with said PXRD peaks is substantially stable under storage at relative humidity of about 100% for at least about 1 week, and storage at about 40° C. and about a 75% relative humidity for at least about 6 months.
54. A process for preparing a crystalline form of fexofenadine hydrochloride having a PXRD pattern with peaks at 10.1, 15.2, 18.6, 19.2, 20.1+0.2, comprising the steps of crystallizing the crystalline form with said PXRD peaks from a solution of fexofenadine hydrochloride in methanol and recovering the crystalline form.
US10/459,688 2002-06-10 2003-06-10 Polymorphic form XVI of fexofenadine hydrochloride Abandoned US20040044038A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US10/459,688 US20040044038A1 (en) 2002-06-10 2003-06-10 Polymorphic form XVI of fexofenadine hydrochloride
US11/243,496 US7671071B2 (en) 2002-06-10 2005-10-03 Polymorphic Form XVI of fexofenadine hydrochloride
US12/208,768 US20090054486A1 (en) 2002-06-10 2008-09-11 Polymorphic form xvi of fexofenadine hydrochloride

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US38797202P 2002-06-10 2002-06-10
US10/459,688 US20040044038A1 (en) 2002-06-10 2003-06-10 Polymorphic form XVI of fexofenadine hydrochloride

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/243,496 Continuation US7671071B2 (en) 2002-06-10 2005-10-03 Polymorphic Form XVI of fexofenadine hydrochloride

Publications (1)

Publication Number Publication Date
US20040044038A1 true US20040044038A1 (en) 2004-03-04

Family

ID=29736394

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/459,688 Abandoned US20040044038A1 (en) 2002-06-10 2003-06-10 Polymorphic form XVI of fexofenadine hydrochloride
US11/243,496 Expired - Fee Related US7671071B2 (en) 2002-06-10 2005-10-03 Polymorphic Form XVI of fexofenadine hydrochloride
US12/208,768 Abandoned US20090054486A1 (en) 2002-06-10 2008-09-11 Polymorphic form xvi of fexofenadine hydrochloride

Family Applications After (2)

Application Number Title Priority Date Filing Date
US11/243,496 Expired - Fee Related US7671071B2 (en) 2002-06-10 2005-10-03 Polymorphic Form XVI of fexofenadine hydrochloride
US12/208,768 Abandoned US20090054486A1 (en) 2002-06-10 2008-09-11 Polymorphic form xvi of fexofenadine hydrochloride

Country Status (8)

Country Link
US (3) US20040044038A1 (en)
EP (1) EP1474392A1 (en)
JP (1) JP2005532356A (en)
AU (1) AU2003248657A1 (en)
DE (1) DE03757471T1 (en)
ES (1) ES2239554T1 (en)
IL (1) IL165689A0 (en)
WO (1) WO2003104197A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020177608A1 (en) * 2001-04-09 2002-11-28 Ben-Zion Dolitzky Polymorphs of fexofenadine hydrochloride
US20030158227A1 (en) * 2001-11-08 2003-08-21 Barnaba Krochmal Polymorphs of fexofenadine base
US20050256163A1 (en) * 2004-04-26 2005-11-17 Ilan Kor Crystalline forms of fexofenadine hydrochloride and processes for their preparation
US20080095843A1 (en) * 2006-07-11 2008-04-24 Nutalapati Siva R K Controlled-release formulations
US20090012301A1 (en) * 2004-09-28 2009-01-08 Teva Pharmaceuticals Usa, Inc. Fexofenadine crystal form and processes for its preparation thereof
US20100183717A1 (en) * 2009-01-16 2010-07-22 Kristin Arnold Controlled-release formulations

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0319935D0 (en) 2003-08-26 2003-09-24 Cipla Ltd Polymorphs
ITMI20041143A1 (en) * 2004-06-08 2004-09-08 Dipharma Spa FEXOFENADINA POLYMORPHS AND PROCEDURE FOR THEIR PREPARATION
US20090306135A1 (en) 2008-03-24 2009-12-10 Mukesh Kumar Sharma Stable amorphous fexofenadine hydrochloride
CA2930128A1 (en) 2013-11-15 2015-05-21 Akebia Therapeutics, Inc. Solid forms of {[5-(3-chlorophenyl)-3-hydroxypyridine-2-carbonyl]amino}acetic acid, compositions, and uses thereof
CN104072402B (en) * 2014-07-16 2016-08-17 昆山龙灯瑞迪制药有限公司 A kind of fexofenadine hydrochloride compound of new crystalline form and preparation method thereof

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4254129A (en) * 1979-04-10 1981-03-03 Richardson-Merrell Inc. Piperidine derivatives
US4636499A (en) * 1984-06-13 1987-01-13 Aktiebolaget Hassle Sulphenamides
US4659716A (en) * 1984-02-15 1987-04-21 Schering Corporation Antihistaminic 8-(halo)-substituted 6,11-dihydro-11-(4-piperidylidene)-5H-benzo[5,6]cyclohepta[1,2-b]pyridines
US4929605A (en) * 1987-10-07 1990-05-29 Merrell Dow Pharmaceuticals Inc. Pharmaceutical composition for piperidinoalkanol derivatives
US5375693A (en) * 1992-08-03 1994-12-27 Sepracor, Inc. Methods and compositions for treating allergic disorders and other disorders metabolic derivatives of terfenadine
US5578610A (en) * 1993-06-24 1996-11-26 Albany Molecular Research, Inc. Piperidine derivatives
US5618940A (en) * 1992-04-10 1997-04-08 Merrell Pharmaceuticals Inc. Process for piperidine derivatives
US5654433A (en) * 1993-01-26 1997-08-05 Merrell Pharmaceuticals Inc. Process for piperidine derivatives
US5738872A (en) * 1995-02-28 1998-04-14 Hoechst Marion Roussel, Inc. Pharmaceutical composition for piperidinoalkanol compounds
US5925761A (en) * 1997-02-04 1999-07-20 Sepracor Inc. Synthesis of terfenadine and derivatives
US5990127A (en) * 1997-03-11 1999-11-23 Hoechst Marion Roussel Deutschland Gmbh Process for the preparation of 4-(4-(4-(hydroxybiphenyl)-1-piperidinyl)-1-hydroxybutyl)-α,α -dimethylphenylacetic acid and phosphorylated derivatives
US6037353A (en) * 1992-05-11 2000-03-14 Hoechst Marion Roussel, Inc. Method of providing an antihistaminic effect in a hepatically impaired patient
US6039974A (en) * 1997-08-26 2000-03-21 Hoechst Marion Roussel, Inc. Pharmaceutical composition for combination of piperidinoalkanol-decongestant
US6147216A (en) * 1993-06-25 2000-11-14 Merrell Pharmaceuticals Inc. Intermediates useful for the preparation of antihistaminic piperidine derivatives
US6153754A (en) * 1995-12-21 2000-11-28 Albany Molecular Research, Inc. Process for production of piperidine derivatives
US6201124B1 (en) * 1995-12-21 2001-03-13 Albany Molecular Research, Inc. Process for production of piperidine derivatives
US6242606B1 (en) * 1993-06-25 2001-06-05 Merrell Pharmaceuticals Inc. Intermediates useful for the preparation of antihistaminic piperidine derivatives
US20010012896A1 (en) * 1994-05-18 2001-08-09 Henton Daniel R. Processes for preparing anhydrous and hydrate forms of antihistaminic piperidine derivatives, polymorphs and pseudomorphs thereof
US6451815B1 (en) * 1997-08-14 2002-09-17 Aventis Pharmaceuticals Inc. Method of enhancing bioavailability of fexofenadine and its derivatives
US20020177608A1 (en) * 2001-04-09 2002-11-28 Ben-Zion Dolitzky Polymorphs of fexofenadine hydrochloride
US20030021849A1 (en) * 2001-04-09 2003-01-30 Ben-Zion Dolitzky Polymorphs of fexofenadine hydrochloride

Family Cites Families (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2176201T3 (en) 1992-04-10 2002-12-01 Merrell Pharma Inc DERIVATIVES OF 4-DIFENYLMETIL PIPERIDINE AND PREPARATION PROCEDURE.
US20020007068A1 (en) * 1999-07-16 2002-01-17 D'ambra Thomas E. Piperidine derivatives and process for their production
US20030045722A1 (en) * 1994-05-18 2003-03-06 Henton Daniel R. Processes for preparing anhydrous and hydrate forms of antihistaminic piperidine derivatives, polymorphs and pseudomorphs thereof
US6700012B2 (en) * 1998-07-02 2004-03-02 Aventis Pharmaceuticals Inc. Antihistaminic piperidine derivatives and intermediates for the preparation thereof
DE69942876D1 (en) 1998-07-02 2010-12-02 Aventisub Ii Inc ANTIHISTAMINIC PIPERIDINE DERIVATIVES AND INTERMEDIATE PRODUCTS FOR THEIR PREPARATION
US6683094B2 (en) * 1998-07-02 2004-01-27 Aventis Pharmaceuticals Inc. Antihistaminic piperidine derivatives and intermediates for the preparation thereof
IN191492B (en) 1999-05-25 2003-12-06 Ranbaxy Lab Ltd
US6613906B1 (en) * 2000-06-06 2003-09-02 Geneva Pharmaceuticals, Inc. Crystal modification
CH695216A5 (en) * 2001-02-23 2006-01-31 Cilag Ag A method for manufacturing a non-hydrated salt of a piperidine derivative and a novel crystalline form thus obtainable of such a salt.
ITRM20010260A1 (en) * 2001-05-17 2002-11-18 Dinamite Dipharma S P A In For PROCESS FOR THE PREPARATION OF PHEXOPHENADINE OR ACID 4 1-HYDROXY-4-4- (HYDROXYDIDENYLMETHYL) -1-PIPERIDINYL | -BUTYL | -ALPHA, ALPHA-DIMETHYLBENZ
WO2002102777A2 (en) * 2001-06-18 2002-12-27 Dr. Reddy's Laboratories Ltd. NOVEL CRYSTALLINE FORMS OF 4-[4-[4-(HYDROXYDIPHENYLMETHYL)-1-PIPERINDINYL]-1-HYDROXYBUTYL]-α, α-DIMETHYLBENZENE ACETIC ACID AND ITS HYDROCHLORIDE
US7700779B2 (en) * 2001-06-18 2010-04-20 Dr. Reddy's Laboratories Limited Crystalline forms of fexofenadine and its hydrochloride
SI21232A (en) * 2001-06-25 2003-12-31 Aurobindo Pharma Limited Process for the preparation of a highly pure pharmaceutical intermediate, 4-(cyclo-propylcarbonyl)-alpha,alpha-dimethylphenylacetic acid
PT1414453E (en) * 2001-07-31 2008-05-29 Texcontor Ets Fexofenadine hydrochloride polymorph
US20030158227A1 (en) * 2001-11-08 2003-08-21 Barnaba Krochmal Polymorphs of fexofenadine base
EP1490034B1 (en) 2002-04-04 2007-06-27 Dr. Reddy's Laboratories Ltd Novel pharmaceutical compositions for antihistaminic-decongestant combination and method of making such compositions
CA2514610A1 (en) 2003-01-31 2004-08-12 Ranbaxy Laboratories Limited Process for the preparation of fexofenadine
GB0319935D0 (en) 2003-08-26 2003-09-24 Cipla Ltd Polymorphs
CA2560882A1 (en) * 2004-04-26 2005-11-03 Teva Pharmaceutical Industries Ltd. Crystalline forms of fexofenadine hydrochloride and processes for their preparation
ITMI20041143A1 (en) * 2004-06-08 2004-09-08 Dipharma Spa FEXOFENADINA POLYMORPHS AND PROCEDURE FOR THEIR PREPARATION
EP1616861A3 (en) * 2004-06-15 2006-02-08 Dipharma S.p.A. A process for the preparation of keto compounds
JP2008514641A (en) * 2004-09-28 2008-05-08 テバ ファーマシューティカル インダストリーズ リミティド Crystalline fexofenadine and method for its preparation
WO2007007347A1 (en) 2005-07-07 2007-01-18 Wockhardt Limited Industrial process of fexofenadine hydrochloride with controlled side products
WO2007049303A2 (en) 2005-10-28 2007-05-03 Ind-Swift Laboratories Limited An improved process for the preparation of highly pure fexofenadine
WO2007052310A2 (en) 2005-11-03 2007-05-10 Morepen Laboratories Limited Polymorphs of fexofenadine hydrochloride and process for their preparation
WO2007135693A2 (en) 2006-05-18 2007-11-29 Ind-Swift Laboratories Limited Intermediates useful for the preparation of antihistaminic piperidine derivative

Patent Citations (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4254129A (en) * 1979-04-10 1981-03-03 Richardson-Merrell Inc. Piperidine derivatives
US4659716A (en) * 1984-02-15 1987-04-21 Schering Corporation Antihistaminic 8-(halo)-substituted 6,11-dihydro-11-(4-piperidylidene)-5H-benzo[5,6]cyclohepta[1,2-b]pyridines
US4636499A (en) * 1984-06-13 1987-01-13 Aktiebolaget Hassle Sulphenamides
US4929605A (en) * 1987-10-07 1990-05-29 Merrell Dow Pharmaceuticals Inc. Pharmaceutical composition for piperidinoalkanol derivatives
US5675009A (en) * 1992-04-10 1997-10-07 Merrell Pharmaceuticals Inc. Process for piperidine derivatives
US5618940A (en) * 1992-04-10 1997-04-08 Merrell Pharmaceuticals Inc. Process for piperidine derivatives
US5631375A (en) * 1992-04-10 1997-05-20 Merrell Pharmaceuticals, Inc. Process for piperidine derivatives
US5644061A (en) * 1992-04-10 1997-07-01 Merrell Pharmaceuticals Inc. Process for piperidine derivatives
US5650516A (en) * 1992-04-10 1997-07-22 Merrell Pharmaceuticals Inc. Process for piperidine derivatives
US5652370A (en) * 1992-04-10 1997-07-29 Merrell Pharmaceuticals Inc. Process for piperidine derivatives
US5663353A (en) * 1992-04-10 1997-09-02 Merrell Pharmaceuticals Inc. Process for piperidine derivatives
US6399632B1 (en) * 1992-05-11 2002-06-04 Merrell Pharmaceuticals Inc. Method of providing an antihistaminic effect in a hepatically impaired patient
US6187791B1 (en) * 1992-05-11 2001-02-13 Merrell Pharmaceuticals Inc. Method of providing an antihistaminic effect in a hepatically impaired patient
US6037353A (en) * 1992-05-11 2000-03-14 Hoechst Marion Roussel, Inc. Method of providing an antihistaminic effect in a hepatically impaired patient
US5375693A (en) * 1992-08-03 1994-12-27 Sepracor, Inc. Methods and compositions for treating allergic disorders and other disorders metabolic derivatives of terfenadine
US5654433A (en) * 1993-01-26 1997-08-05 Merrell Pharmaceuticals Inc. Process for piperidine derivatives
US5994549A (en) * 1993-06-24 1999-11-30 Albany Molecular Research, Inc. Piperidine derivatives and process for their production
US5663412A (en) * 1993-06-24 1997-09-02 Albany Molecular Research, Inc. Aromatic ketones
US5750703A (en) * 1993-06-24 1998-05-12 Albany Molecular Research, Inc. Piperidine derivatives and process for their production
US5578610A (en) * 1993-06-24 1996-11-26 Albany Molecular Research, Inc. Piperidine derivatives
US5581011A (en) * 1993-06-24 1996-12-03 Albany Molecular Research, Inc. Aromatic ketones and processes for their preparation
US5589487A (en) * 1993-06-24 1996-12-31 Albany Molecular Research, Inc. Piperidine derivatives and process for their production
US6242606B1 (en) * 1993-06-25 2001-06-05 Merrell Pharmaceuticals Inc. Intermediates useful for the preparation of antihistaminic piperidine derivatives
US6348597B2 (en) * 1993-06-25 2002-02-19 Merrell Pharmaceuticals, Inc. Intermediates useful for the preparation of antihistaminic piperidine derivatives
US6147216A (en) * 1993-06-25 2000-11-14 Merrell Pharmaceuticals Inc. Intermediates useful for the preparation of antihistaminic piperidine derivatives
US6340761B1 (en) * 1993-06-25 2002-01-22 Merrell Pharmaceuticals Inc. Intermediates useful for the preparation of antihistaminic piperidine derivatives
US20010025106A1 (en) * 1994-05-18 2001-09-27 Henton Daniel R. Processes for preparing anhydrous and hydrate forms of antihistaminic piperidine derivatives, polymorphs and pseudomorphs thereof
US20010012896A1 (en) * 1994-05-18 2001-08-09 Henton Daniel R. Processes for preparing anhydrous and hydrate forms of antihistaminic piperidine derivatives, polymorphs and pseudomorphs thereof
US20010014741A1 (en) * 1994-05-18 2001-08-16 Henton Daniel R. Processes for preparing anhydrous and hydrate forms of antihistaminic piperidine derivatives, polymorphs and pseudomorphs thereof
US20020193603A1 (en) * 1994-05-18 2002-12-19 Henton Daniel R. Processes for preparing anhydrous and hydrate forms of antihistaminic piperidine derivatives, polymorphs and pseudomorphs thereof
US5855912A (en) * 1995-02-28 1999-01-05 Hoechst Marion Roussel, Inc. Pharmaceutical compositions for piperidinalkanol compounds
US6113942A (en) * 1995-02-28 2000-09-05 Aventis Pharmaceuticals Inc. Pharmaceutical composition for piperidinoalkanol compounds
US5932247A (en) * 1995-02-28 1999-08-03 Hoechst Marion Roussel, Inc. Pharmaceutical composition for piperidinoalkanol compounds
US5738872A (en) * 1995-02-28 1998-04-14 Hoechst Marion Roussel, Inc. Pharmaceutical composition for piperidinoalkanol compounds
US6201124B1 (en) * 1995-12-21 2001-03-13 Albany Molecular Research, Inc. Process for production of piperidine derivatives
US6153754A (en) * 1995-12-21 2000-11-28 Albany Molecular Research, Inc. Process for production of piperidine derivatives
US5925761A (en) * 1997-02-04 1999-07-20 Sepracor Inc. Synthesis of terfenadine and derivatives
US5990127A (en) * 1997-03-11 1999-11-23 Hoechst Marion Roussel Deutschland Gmbh Process for the preparation of 4-(4-(4-(hydroxybiphenyl)-1-piperidinyl)-1-hydroxybutyl)-α,α -dimethylphenylacetic acid and phosphorylated derivatives
US6451815B1 (en) * 1997-08-14 2002-09-17 Aventis Pharmaceuticals Inc. Method of enhancing bioavailability of fexofenadine and its derivatives
US6039974A (en) * 1997-08-26 2000-03-21 Hoechst Marion Roussel, Inc. Pharmaceutical composition for combination of piperidinoalkanol-decongestant
US20020177608A1 (en) * 2001-04-09 2002-11-28 Ben-Zion Dolitzky Polymorphs of fexofenadine hydrochloride
US20030021849A1 (en) * 2001-04-09 2003-01-30 Ben-Zion Dolitzky Polymorphs of fexofenadine hydrochloride

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020177608A1 (en) * 2001-04-09 2002-11-28 Ben-Zion Dolitzky Polymorphs of fexofenadine hydrochloride
US20030158227A1 (en) * 2001-11-08 2003-08-21 Barnaba Krochmal Polymorphs of fexofenadine base
US20050256163A1 (en) * 2004-04-26 2005-11-17 Ilan Kor Crystalline forms of fexofenadine hydrochloride and processes for their preparation
US20090082398A1 (en) * 2004-04-26 2009-03-26 Teva Pharmaceutical Industries Ltd. Crystalline forms of fexofenadine hydrochloride and processes for their preparation
US20090012301A1 (en) * 2004-09-28 2009-01-08 Teva Pharmaceuticals Usa, Inc. Fexofenadine crystal form and processes for its preparation thereof
US20080095843A1 (en) * 2006-07-11 2008-04-24 Nutalapati Siva R K Controlled-release formulations
US20100183717A1 (en) * 2009-01-16 2010-07-22 Kristin Arnold Controlled-release formulations

Also Published As

Publication number Publication date
WO2003104197A1 (en) 2003-12-18
IL165689A0 (en) 2006-01-15
US20060217557A1 (en) 2006-09-28
US7671071B2 (en) 2010-03-02
AU2003248657A1 (en) 2003-12-22
JP2005532356A (en) 2005-10-27
US20090054486A1 (en) 2009-02-26
ES2239554T1 (en) 2005-10-01
WO2003104197A9 (en) 2004-05-27
DE03757471T1 (en) 2005-09-01
EP1474392A1 (en) 2004-11-10

Similar Documents

Publication Publication Date Title
US7671071B2 (en) Polymorphic Form XVI of fexofenadine hydrochloride
US20090149497A1 (en) Polymorphs of fexofenadine hydrochloride
US7105557B2 (en) Polymorphs of valsartan
US7439373B2 (en) Crystalline mycophenolate sodium
US20050256163A1 (en) Crystalline forms of fexofenadine hydrochloride and processes for their preparation
US20090012121A1 (en) Polymorphs of fexofenadine hydrochloride
US7417165B2 (en) Crystalline forms of pregabalin
EP1507531B1 (en) Stable pharmaceutical compositions of desloratadine
US20090012301A1 (en) Fexofenadine crystal form and processes for its preparation thereof
WO2003039482A2 (en) Polymorphs of fexofenadine base
US20030158227A1 (en) Polymorphs of fexofenadine base
EP1950204A1 (en) Amorphous form of valsartan
US20070093549A1 (en) Methods for preparation of ladostigil tartrate crystalline form A1
EP1768969B1 (en) Crystalline mycophenolate sodium
JP2005537218A (en) Polymorphs of fexofenadine base

Legal Events

Date Code Title Description
AS Assignment

Owner name: TEVA PHARMACEUTICAL INDUSTRIES LTD., ISRAEL

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KROCHMAL, BARNADA;DILLER, DOV;DOLITZKY, BEN-ZION;AND OTHERS;REEL/FRAME:014577/0356;SIGNING DATES FROM 20030806 TO 20030913

Owner name: TEVA PHARMACEUTICALS USA, INC., PENNSYLVANIA

Free format text: ASSIGNMENT OF RIGHTS IN BARBADOS;ASSIGNOR:TEVA PHARMACEUTICAL INDUSTRIES LTD.;REEL/FRAME:014586/0502

Effective date: 20030910

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION