US20100041687A1 - Hydrobromide salt of an anti-hiv compound - Google Patents

Hydrobromide salt of an anti-hiv compound Download PDF

Info

Publication number
US20100041687A1
US20100041687A1 US12/517,665 US51766507A US2010041687A1 US 20100041687 A1 US20100041687 A1 US 20100041687A1 US 51766507 A US51766507 A US 51766507A US 2010041687 A1 US2010041687 A1 US 2010041687A1
Authority
US
United States
Prior art keywords
compound
formula
hiv
spray
solid dispersion
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
US12/517,665
Inventor
Daniel Joseph Christiaan Thoné
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.)
Janssen Sciences Ireland ULC
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
Publication of US20100041687A1 publication Critical patent/US20100041687A1/en
Assigned to JANSSEN PHARMACEUTICA NV reassignment JANSSEN PHARMACEUTICA NV ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: THONE, DANIEL JOSEPH CHRISTIAAN
Assigned to TIBOTEC PHARMACEUTICALS reassignment TIBOTEC PHARMACEUTICALS CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: TIBOTEC PHARMACEUTICALS LTD.
Assigned to JANSSEN R&D IRELAND reassignment JANSSEN R&D IRELAND CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: TIBOTEC PHARMACEUTICALS
Assigned to TIBOTEC PHARMACEUTICALS LTD. reassignment TIBOTEC PHARMACEUTICALS LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JANSSEN PHARMACEUTICA NV
Assigned to JANSSEN SCIENCES IRELAND UC reassignment JANSSEN SCIENCES IRELAND UC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JANSSEN R & D IRELAND
Assigned to Janssen Sciences Ireland Unlimited Company reassignment Janssen Sciences Ireland Unlimited Company CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: JANSSEN SCIENCES IRELAND UC
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1635Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1641Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1652Polysaccharides, e.g. alginate, cellulose derivatives; Cyclodextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/141Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers
    • A61K9/145Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers with organic compounds

Definitions

  • This invention concerns the hydrobromide salt of a diarylpyrimidine derivative that is useful as an anti-HIV agent. It further concerns solid dispersions as well as pharmaceutical dosage forms comprising this salt.
  • HIV human immunodeficiency virus
  • AIDS acquired immunodeficiency syndrome
  • PIs HIV protease inhibitors
  • HIV entry inhibitors One class of anti-HIV pharmaceutical agents comprises the non-nucleoside reverse transcriptase inhibitors (NNRTIs).
  • WO 00/27825 discloses a series of pyrimidine derivatives that function as NNRTIs.
  • a particularly successful type of NNRTI disclosed in WO 00/27825 is 4-[[4-amino-5-bromo-6-(4-cyano-2,6-dimethylphenyloxy)-2-pyrimidinyl]amino]benzonitrile, also known as etravirine or TMC125, and which is represented by the formula (A):
  • the compound of formula (A) has high intrinsic activity not only against wild-type HIV but also against HIV strains harboring resistance-inducing mutations. Consequently, the compound of formula (A) is very useful in the treatment of individuals infected by HIV. Yet its proper functioning is impeded by its very poor solubility in water and concomitant low bioavailability.
  • a pharmaceutical agent having low bioavailability is that the amount (or dose) administered to a patient needs to be higher than it would be if the bioavailability were higher.
  • An increased dose however not only results in an increased risk of side effects and intolerance to the pharmaceutical agent but also leads to an increase in the size and/or the number of dosage forms required.
  • pill burden The number or volume of dosage forms that needs to be administered is commonly referred to as the “pill burden”.
  • a high pill burden is undesirable for many reasons, such as the patient having to spend more time taking each dose and the patient having to store and/or transport a large number or volume of pills.
  • a high pill burden also increases the risk that patients will not take their entire dose, thereby failing to comply with the prescribed dosage regimen. As well as reducing the effectiveness of the treatment, this may also lead to the disease-causing organism or virus becoming resistant to the pharmaceutical agent.
  • a patient must take a combination of a number of different types of pharmaceutical agents.
  • One example of such a group of patients is that being treated for HIV.
  • Anti-HIV treatment generally involves administration of a combination of a number of different pharmaceutical agents.
  • a high pill burden is a particular disadvantage.
  • Having a high pill burden also means that patients infected with HIV may be less able to discreetly take the required doses of pharmaceutical agent(s). This may be a particular problem for those who do not wish their condition to become widely known.
  • the degree of crystallinity of a poorly water-soluble pharmaceutical agent is inversely proportional to its bioavailability. As such, the more crystalline the pharmaceutical agent is, the lower its bioavailability. Increased bioavailability may be achieved by providing the pharmaceutical agent in an amorphous form.
  • One way of achieving this is by incorporating the active ingredient into a solid dispersion wherein the active ingredient is dispersed in amorphous form throughout a polymeric matrix, which typically is a water-soluble pharmaceutically acceptable polymeric material.
  • Solid dispersions can be obtained in several ways, e.g. by dissolving the active ingredient and the matrix polymer, optionally in the presence of other ingredients, and allowing the solvent to evaporate.
  • Melt extrusion offers another method to obtain solid dispersions.
  • the spray drying technique offers still another way of obtaining solid dispersions of active ingredients.
  • a solution of the active ingredient and a polymer, optionally together with other ingredients, is sprayed thereby allowing the solvent to evaporate thus obtaining finely dispersed particles.
  • the compound of formula (A) suffers from poor bioavailability thereby impeding its proper functioning. Therefore there is need to improve the bioavailability of this active ingredient so that its beneficial properties can have full play. A higher bioavailability moreover helps to reduce the size and number of dosage forms to be administered and this in turn contributes to an even further reduced pill burden. It now has been found that the hydrobromic acid-addition salt form of the compound of formula (A) has improved bioavailability and is particularly attractive when used in the form of solid dispersions, in particular in the form of solid dispersions prepared by the spray-drying technique.
  • the said solid dispersion is a spray-dried solid dispersion.
  • a compound of formula (I) for preparing a solid dispersion of the compound of formula (I) in a water-soluble polymer.
  • said use is for preparing a solid dispersion of said compound of formula (I) by spray-drying.
  • a method of preparing a solid dispersion of the compound of formula (I) in a solid polymeric matrix comprising dispersing the compound of formula (I) into a solid polymeric matrix.
  • said method is by spray-drying.
  • a pharmaceutical formulation comprising a solid dispersion of a compound of formula (I) in a solid polymeric matrix, and a carrier.
  • said formulation comprises a spray-dried solid dispersion of a compound of formula (I).
  • the compound of formula (I), i.e. 4-[[4-amino-5-bromo-6-(4-cyano-2,6-dimethyl-phenyloxy)-2-pyrimidinyl]amino]benzonitrile hydrobromide, which is the hydrobromide salt of the compound of formula (A), is deemed novel. Therefore, in a further aspect, the present invention concerns said compound of formula (I). In one embodiment, the compound of formula (I) is in amorphous form. In a further aspect of the invention there are provided pharmaceutical formulations comprising the compound of formula (I) and a carrier. In these formulations, the compound of formula (I) is present in an effective amount, i.e. an amount that is effective to inhibit HIV.
  • the compound of formula (I) is a stable salt.
  • the maleate and methane sulfonate salts of compound (A) have been found to be unstable, because of their partial or complete decomposition to compound (A) and the free acid.
  • the compound of formula (A) may be prepared according to the method described in WO 00/27825.
  • the compound of formula (I) may be prepared by treating the compound of formula (A) with hydrobromic acid.
  • the compound of formula (I) is useful in the treatment of individuals infected by HIV. It can be used to inhibit HIV and additionally to treat conditions associated with HIV infection.
  • HIV infection include AIDS as well as secondary conditions associated with HIV infection such as thrombocytopaenia, Kaposi's sarcoma, and infection of the central nervous system characterized by progressive demyelination, resulting in dementia and symptoms such as progressive dysarthria, ataxia, and disorientation.
  • Other conditions associated with HIV infection comprise peripheral neuropathy, progressive generalized lymphadenopathy (PGL), and AIDS-related complex (ARC).
  • the compound of formula (I) is preferably highly amorphous, that is, it has a low level of crystallinity.
  • amorphous is meant that the active agent is in a non-crystalline state. This again advantageously increases the bioavailability of the compound of formula (I) thereby decreasing the amount of compound that needs to be administered to a patient resulting in a reduced pill burden.
  • the degree of crystallinity of the pharmaceutical agent in the solid pharmaceutical composition is about 20% or less, about 15% or less, about 10% or less, about 9% or less, about 8% or less, about 7% or less, about 6% or less, about 5% or less, about 4% or less, about 3% or less, about 2% or less, about 1% or less, about 0.5% or less, or about 0.1% or less.
  • XRPD X-ray powder diffraction
  • the compound of formula (I) may be formulated into a dosage form for administration to a patient.
  • Typical dosage forms include dosage forms for administration orally, rectally, vaginally, percutaneously or by parenteral injection.
  • Suitable dosage forms for oral administration include tablets, capsules, powders, pills, suspensions, and solutions.
  • Preferred dosage forms for oral administration are tablets and capsules.
  • Suitable dosage forms for rectal or vaginal administration include gels, suppositories, or pastes.
  • the compound of formula (I) may be combined with any suitable pharmaceutically acceptable excipient appropriate for the particular dosage form being prepared.
  • suitable excipients include water, glycols, oils, alcohols, starches, sugars, kaolin, diluents, lubricants, binders, and disintegrating agents.
  • polyethylene glycol is preferably used as the vehicle wherein the compound of formula (I) is dissolved. More preferably the polyethylene glycol is PEG 400.
  • the compound of formula (I) is particularly useful to formulate into a solid dispersion comprising the compound of formula (I), a pharmaceutically acceptable water-soluble polymer, and optionally one or more excipients.
  • a solid dispersion comprising the compound of formula (I), a pharmaceutically acceptable water-soluble polymer, and optionally one or more excipients.
  • the bioavailability of the compound of formula (I) is improved.
  • Different types of solid dispersions exist. In a first type of solid dispersion the pharmaceutical agent is molecularly dispersed, substantially homogeneously, throughout the polymer. This is generally described as a “solid solution”. A second type of solid dispersion is where there are islands of crystalline or semi-crystalline pharmaceutical agent dispersed throughout the polymer. A third type of solid dispersion is where there are islands or clusters of pharmaceutical agent in amorphous form dispersed throughout the polymer.
  • Spray-drying offers an attractive technique for preparing solid dispersions because it is a continuous process that can be used at large scale.
  • the amount of the compound of formula (I) in the solution introduced to the spray-drying chamber is typically about 10% to about 60%, about 15% to about 50%, about 20% to about 40%, or about 25% to about 35% by weight compared to the total weight of the compound of formula (I), water-soluble polymer and optional excipients.
  • the amount of compound dissolved is determined by its solubility in the solvent(s) chosen.
  • Polymers suitable for use in the solid dispersions of the compound of formula (I) comprise any that are pharmaceutically acceptable, water-soluble and substantially unreactive towards the pharmaceutical agent.
  • Suitable polymers include cellulosic polymers, such as methyl cellulose, ethyl cellulose, hydroxymethyl cellulose, hydroxy-ethyl cellulose, hydroxypropyl cellulose, hydroxybutyl cellulose, hydroxyethylmethyl cellulose, hydroxypropylmethyl cellulose (HPMC), e.g. HPMC 2910, carboxymethyl cellulose, hydroxypropylmethyl cellulose phthalate (HPMCP), e.g.
  • HP 50 hydroxy-propylmethyl cellulose acetate succinate (HPMCAS), cellulose acetate trimellitate (CAT), hydroxypropylcellulose acetate phthalate (HPCAP), hydroxypropylmethyl cellulose acetate phthalate (HPMCAP), methyl cellulose acetate phthalate (MCAP), and mixtures thereof such as a mixture of hydroxypropyl cellulose and ethyl cellulose.
  • Suitable polymers also include polyvinyl pyrrolidone, copolyvidone, which is polyvinyl pyrrolidone copolymerized with vinyl acetate, and aminoalkyl methacrylate copolymers, such as Eudragit E® 100 (Röhm GmbH, Germany).
  • the polymer is hydroxypropylmethyl cellulose (HPMC), polyvinyl pyrrolidone or copolyvidone.
  • HPMC hydroxypropylmethyl cellulose
  • polyvinyl pyrrolidones are PVP K12, PVP K30, or PVP K90 and a particularly preferred copolyvidone is PVP-co-VA64.
  • the amount of water-soluble polymer in the spray dried product may be in the range from about 30% to about 75%, in particular about 40% to about 75%, or about 50% to about 75% or about 60% to about 70%, by weight relative to the total weight of the spray dried product comprising compound (I), i.e. TMC125.HBr, water-soluble polymer and optional excipients.
  • the amount of water-soluble polymer in the feed mixture can be calculated based on these percentages and on the amount of solvent used.
  • the weight:weight ratio of water-soluble polymer to the compound of formula (I) may be in the range from about 10:1 to about 1:10, in particular from about 10:1 to about 1:1, more in particular from about 5:1 to about 1:1, or from about 4:1 to about 1:1; or from about 3:1 to about 1:1, e.g. a ratio of about 3:1, of about 3.5:1, or of about 3.6:1.
  • the ratio of water-soluble polymer to pharmaceutical agent is believed to affect the degree of crystallinity of the pharmaceutical agent in the resulting solid pharmaceutical composition. However, it is also desirable to reduce the amount of polymer in relation to the pharmaceutical agent in order to maximize the amount of pharmaceutical agent in the resulting pharmaceutical composition.
  • the solvent used in the spraying process may be any solvent that is inert with respect to the compound of formula (I) and that is able to dissolve the compound of formula (I) and the water-soluble polymer. Mixtures of solvents may be used. Suitable solvents include acetone, tetrahydrofuran (THF), dichloromethane, ethanol (anhydrous or aqueous), methanol, and combinations thereof. Where the polymer is HPMC, the solvent is preferably a mixture of dichloromethane and ethanol, more preferably a mixture of dichloromethane and ethanol, the latter in particular being anhydrous ethanol, in a 9:1 ratio by weight.
  • the solvent is preferably acetone.
  • the amount of solvent present in the feed mixture will be such that TMC125 and the water-soluble polymer are dissolved and that the feed mixture has sufficient low viscosity for it to be sprayed.
  • the amount of solvent in the feed mixture will be at least 80%, in particular at least 90%, preferably at least 95%, the percentages expressing the weight amount of solvent to the total weight of the feed mixture.
  • the feed mixture may contain further excipients such as to improve the properties of the feed mixture or the resulting solid pharmaceutical composition, for example to improve the handling or processing properties.
  • the addition of excipients obviously results in these being incorporated in the solid dispersion. Regardless of whether or not excipients are added to the feed mixture, these may also be mixed with the resulting solid pharmaceutical composition during formulation into a desired dosage form.
  • microcrystalline cellulose (“MCC”).
  • MCC microcrystalline cellulose
  • the microcrystalline cellulose (MCC) that can be used has an average particle size that is selected such that when mixed into the solution of pharmaceutical agent and water-soluble polymer, the resulting feed mixture is able to pass through the atomizing means into the spray-drying chamber without clogging or blocking the atomizer.
  • the size of the MCC is limited by the size of the atomizing means provided on the spray-drying chamber.
  • the atomizing means is a nozzle
  • the average particle size of the MCC may be in the range of from 5 ⁇ m to 50 ⁇ m, in particular from 10 ⁇ m to 30 ⁇ m, e.g. about 20 ⁇ m.
  • Microcrystalline cellulose that can be used comprises the AvicelTM series of products available from FMC BioPolymer, in particular Avicel PH 105® (20 ⁇ m), Avicel PH 101® (50 ⁇ m), Avicel PH 301® (50 ⁇ m);
  • microcrystalline cellulose products available from JRS Pharma in particular Vivapur® 105 (20 ⁇ m), Vivapur® 101 (50 ⁇ m), Emcocel® SP 15 (15 ⁇ m), Emcocel® 50 M 105 (50 ⁇ m), Prosolv® SMCC 50 (50 ⁇ m); the microcrystalline cellulose products available from DMV, in particular Pharmacel® 105 (20 ⁇ m), Pharmacel® 101 (50 ⁇ m); the microcrystalline cellulose products available from Blanver, in particular Tabulose (Microcel)® 101 (50 ⁇ m), Tabulose (Microcel)® 103 (50 ⁇ m); the microcrystalline cellulose products available from Asahi Kasei Corporation, such as Ceolus® PH-F20JP (20 ⁇ m), Ceolus® PH-101 (50 ⁇ m), Ceolus® PH-301 (50 ⁇ m), Ceolus® KG-802 (50 ⁇ m).
  • a particularly preferred microcrystalline cellulose is Avicel PH 105® (20 ⁇ m).
  • the amount of MCC in the spray dried product may be in the range from about 5% to about 25%, in particular about 7.5% to about 20%, or about 10% to about 15% or about 10% to about 12.5%, by weight relative to the total weight of the spray dried product comprising TMC125, water-soluble polymer, MCC and optional excipients.
  • the weight ratio of the amounts of MCC to TMC125 in the spray dried product can be calculated based on these percentages and in particular may be in the range of from about 2:1 to about 1:5, in particular from about 1:1 to 1:7, preferably about 1:2.
  • the amount of MCC in the feed mixture can be calculated based on these percentages and on the amount of solvent used. In view of the desirability of keeping the concentration of pharmaceutical agent in the resulting solid pharmaceutical composition as high as possible, the concentration of MCC is preferably kept as low as possible.
  • excipients suitable for inclusion in the feed mixture and/or in the resulting pharmaceutical composition include surfactants, solubilizers, disintegrants, pigments, flavorings, fillers, lubricants, glidants, preservatives, thickening agents, buffering agents and pH modifiers.
  • surfactants include sodium lauryl sulphate, Cremophor RH 40, Vitamin E TPGS, and polysorbates, such as Tween 20®.
  • Typical pH modifiers are acids, such as citric acid or succinic acid, bases, or buffers.
  • the solid pharmaceutical composition produced by the process of the invention typically comprises particles having an average effective particle size in the range of from about 10 ⁇ m to about 150 ⁇ m, or about 15 ⁇ m to about 100 ⁇ m, particularly about 20 ⁇ m to about 80 ⁇ m, or 30 ⁇ m to about 50 ⁇ m, preferably about 40 ⁇ m.
  • the term average effective particle size has its conventional meaning as known to the person skilled in the art and can be measured by art-known particle size measuring techniques such as, for example, sedimentation field flow fractionation, photon correlation spectroscopy, laser diffraction or disk centrifugation.
  • the average effective particle sizes mentioned herein may be related to weight distributions of the particles.
  • an average effective particle size of about 150 ⁇ m it is meant that at least 50% of the total weight of the particles is composed of particles having a particle size of less than the effective average of 150 ⁇ m, and the same applies to the other effective particle sizes mentioned.
  • the average effective particle sizes may be related to volume distributions of the particles but usually this will result in the same or about the same value for the average effective particle size.
  • the so-called “span” of the spray-dried particles may be lower than about 3, in particular lower than about 2.5, e.g. the span is about 2. Usually the span will not be lower than about 1.
  • the term “span” is defined by the formula (D 90 ⁇ D 10 )/D 50 wherein D 90 is the particle diameter corresponding to the diameter of particles that make up 90% of the total weight of all particles of equal or smaller diameter and wherein D 50 and D 10 are the diameters for 50 respectively 10% of the total weight of all particles.
  • the spray-dried particles may be formulated into a pharmaceutical formulation.
  • the latter comprises the spray-dried particles and a carrier, which may comprise one or more pharmaceutically acceptable ingredients such as, for example, a lubricant, a binder, thickener, and the like ingredients.
  • a carrier which may comprise one or more pharmaceutically acceptable ingredients such as, for example, a lubricant, a binder, thickener, and the like ingredients.
  • the spray-dried particles may be added to a suitable liquid, such as water.
  • the pharmaceutical formulations in turn may be converted into an appropriate dosage form.
  • Typical dosage forms include dosage forms for oral administration, such as tablets, capsules, suspensions and pastilles, and dosage forms for rectal or vaginal administration, such as gels, suppositories or pastes.
  • the spray-dried particles may be subjected to further processing steps depending on the nature of the final dosage form.
  • the pharmaceutical composition may be subjected to a post-drying process, or may undergo tabletting or encapsulating processes.
  • further processing steps may be required, such as the incorporation of a disintegrant for immediate release products or the coating of the powder with an enteric layer for controlled release products.
  • Suitable disintegrants include microcrystalline cellulose, starch, sodium starch glycolate, crosslinked carboxy-methylcellulose sodium, and crosslinked PVP.
  • the effective daily amount of the compound of formula (I) is from about 1 mg/kg to about 20 mg/kg body weight, or from about 2 mg/kg to about 10 mg/kg body weight, or from about 5 mg/kg to about 8 mg/kg body weight, or from about 5 mg/kg to about 6 mg/kg body weight.
  • the effective daily amounts per patient can be obtained by multiplying the before-mentioned amounts by 70 or by the actual weight of the patient.
  • a person skilled in the art will be able to determine the exact dose and frequency of administration required, which will depend on a number of factors, such as the severity of the condition being treated, the age and weight of the individual being treated and whether that individual is taking any other medication.
  • the effective daily amount of the compound of formula (I) may be administered as a single dose or as two, three, four or more sub-doses at appropriate intervals throughout the day.
  • the dose or sub-doses of the compound of formula (I) may be formulated as unit dosage forms containing a specified amount of the compound, typically from about 10 to about 1000 mg, from about 50 to about 800 mg, from about 100 to about 500 mg, from about 100 mg to about 300 mg, e.g. about 200 mg or about 240 mg of the compound of formula (I).
  • the compound of formula (A) that is, 4-[[4-amino-5-bromo-6-(4-cyano-2,6-dimethyl-phenyloxy)-2-pyrimidinyl]amino]benzonitrile, was dissolved in dichloromethane and aqueous hydrobromic acid was added. The mixture was evaporated to dry yielding the hydrobromide salt of (A), i.e. the compound of formula (I).
  • Other addition salts of the compound of formula (A) can be obtained similarly and are shown in the following Table 1.
  • Capsules were filled with the various salts of the compound of formula (I) in the amounts shown in the following Table 2.
  • Salt of compound (I) Amount of salt per capsule in mg Hydrobromide 118.6 Hydrochloride 108.4 Maleate 63.3 Methane sulfonate 122.1 Fumarate 63.3 Sulfate 122.5
  • the animals were randomized over the test groups by a computer-generated random algorithm according to body weight, with all animals within ⁇ 20% of the sex mean. A health inspection was performed prior to commencement of treatment to ensure that the animals were in a good state of health.
  • the dogs were subjected to an acclimatization period of at least 5 days before the start of treatment under laboratory conditions. A controlled environment was maintained in the room.
  • the animals were dosed once orally with the formulations of example 1, and blood was collected from all animals under light ether anaesthesia at regular intervals for a period of 12, 24 or 48 hours after dosing.
  • the mean pharmacokinetic parameters relating to the mean plasma concentration with time of each test formulation are shown in Table 3. The standard deviation, where calculated, is shown between brackets.
  • Analysis of the peak plasma concentration (C max ) and bioavailability in terms of area under the curve (AUC 0-last ) shows that the hydrobromide salt of compound (I), that is the compound of formula (I) has advantageously improved bioavailability in dogs compared with other salts of compound (I).
  • t max is the time taken to reach the peak plasma concentration.
  • t last is the last time at which the peak plasma concentration was determined.
  • Formulation G Compound of formula (I) 59.3 mg HPMC 5 m Pa ⁇ s 177.9 mg
  • formulation G the compound of formula (I) was dissolved in a first beaker in methanol/dichloromethane 50/50 v/v % at a concentration of 6.5 g per 750 ml of methanol/dichloromethane.
  • HPMC 2910 5 mPa ⁇ s was dissolved in methanol/dichloro-methane 50/50 v/v % in a second beaker.
  • the concentration of HPMC 2910 5 mPa ⁇ s in the solution was 6.5 g per 100 ml methanol/dichloromethane.
  • the spray dried powder was dried under reduced pressure at 40° C. for 5 days.
  • the resulting formulation was filled into Swedish-orange capsules.
  • formulation L 118.6 mg of the compound of formula (I) was dissolved in a first beaker in methanol/dichloromethane 50/50 v/v % at a concentration of 6.5 g per 750 ml of methanol/dichloromethane
  • a second beaker 234.8 mg HPMC 2910 5 mPa ⁇ s was dissolved in methanol/dichloromethane 50/50 v/v %.
  • the concentration of HPMC 2910 5 mPa ⁇ s in the solution was 6.5 g per 100 ml methanol/dichloromethane.
  • the two solutions were subsequently mixed and then sprayed onto 121 mg of HMPC 5 mPa ⁇ s.
  • the resulting powder was filled into Swedish-orange capsules.
  • One capsule of formulation L contained 118.6 mg of the compound of formula (I), which is equivalent to 100 mg of compound (A).
  • a randomized human bioequivalence trial was carried out to monitor bioavailability in humans of formulation L.
  • formulation L nine subjects were given 800 mg of formulation L twice daily for 13 days, with a single dose on day 14. A 12 h pharmacokinetic profile was determined on day 1 and on day 14. Pre-dose concentrations were determined every other day. The results are listed in the following Table.
  • Scheduled Plasma conc. of TMC125 (ng/ml) time Mean ⁇ SD Day 1 0 h Not quantifiable 1 h 35.3 ⁇ 33.1 2 h 638 ⁇ 451 3 h 1220 ⁇ 750 4 h 136 ⁇ 681 6 h 1020 ⁇ 395 8 h 680 ⁇ 285 10 h 476 ⁇ 182 12 h 392 ⁇ 166 Day 3 0 h 1230 ⁇ 341 Day 5 0 h 1630 ⁇ 513 Day 7 0 h 1830 ⁇ 769 Day 9 0 h 1830 ⁇ 720 Day 11 0 h 1940 ⁇ 758 Day 13 0 h 1870 ⁇ 740 Day 14 0 h 1660 ⁇ 578

Abstract

Solid dispersions and dosage forms comprising the hydrobromide salt of a diarylpyrimidine derivative, useful as an anti-HIV agent.

Description

  • This invention concerns the hydrobromide salt of a diarylpyrimidine derivative that is useful as an anti-HIV agent. It further concerns solid dispersions as well as pharmaceutical dosage forms comprising this salt.
  • The human immunodeficiency virus (HIV) is generally recognized as the agent causing the acquired immunodeficiency syndrome (AIDS), of which two distinct types have been identified, i.e. HIV-1 and HIV-2. Hereinafter, the term HIV is used to generically denote both these types. The spread of HIV has caused and continues to cause serious health problems throughout the world. Consequently, the search for effective pharmaceutical agents to treat HIV infection is of vital importance. AIDS patients are currently treated with a variety of agents such as HIV reverse transcriptase inhibitors (RTIs), HIV protease inhibitors (PIs) and HIV entry inhibitors. One class of anti-HIV pharmaceutical agents comprises the non-nucleoside reverse transcriptase inhibitors (NNRTIs).
  • WO 00/27825 discloses a series of pyrimidine derivatives that function as NNRTIs. A particularly successful type of NNRTI disclosed in WO 00/27825 is 4-[[4-amino-5-bromo-6-(4-cyano-2,6-dimethylphenyloxy)-2-pyrimidinyl]amino]benzonitrile, also known as etravirine or TMC125, and which is represented by the formula (A):
  • Figure US20100041687A1-20100218-C00001
  • The compound of formula (A) has high intrinsic activity not only against wild-type HIV but also against HIV strains harboring resistance-inducing mutations. Consequently, the compound of formula (A) is very useful in the treatment of individuals infected by HIV. Yet its proper functioning is impeded by its very poor solubility in water and concomitant low bioavailability.
  • One consequence of a pharmaceutical agent having low bioavailability is that the amount (or dose) administered to a patient needs to be higher than it would be if the bioavailability were higher. An increased dose however not only results in an increased risk of side effects and intolerance to the pharmaceutical agent but also leads to an increase in the size and/or the number of dosage forms required.
  • The number or volume of dosage forms that needs to be administered is commonly referred to as the “pill burden”. A high pill burden is undesirable for many reasons, such as the patient having to spend more time taking each dose and the patient having to store and/or transport a large number or volume of pills. A high pill burden also increases the risk that patients will not take their entire dose, thereby failing to comply with the prescribed dosage regimen. As well as reducing the effectiveness of the treatment, this may also lead to the disease-causing organism or virus becoming resistant to the pharmaceutical agent.
  • The problems associated with a high pill burden are multiplied where a patient must take a combination of a number of different types of pharmaceutical agents. One example of such a group of patients is that being treated for HIV. Anti-HIV treatment generally involves administration of a combination of a number of different pharmaceutical agents. In such cases, therefore, a high pill burden is a particular disadvantage. Having a high pill burden also means that patients infected with HIV may be less able to discreetly take the required doses of pharmaceutical agent(s). This may be a particular problem for those who do not wish their condition to become widely known.
  • It is known that the degree of crystallinity of a poorly water-soluble pharmaceutical agent is inversely proportional to its bioavailability. As such, the more crystalline the pharmaceutical agent is, the lower its bioavailability. Increased bioavailability may be achieved by providing the pharmaceutical agent in an amorphous form. One way of achieving this is by incorporating the active ingredient into a solid dispersion wherein the active ingredient is dispersed in amorphous form throughout a polymeric matrix, which typically is a water-soluble pharmaceutically acceptable polymeric material. Solid dispersions can be obtained in several ways, e.g. by dissolving the active ingredient and the matrix polymer, optionally in the presence of other ingredients, and allowing the solvent to evaporate. Melt extrusion offers another method to obtain solid dispersions. The spray drying technique offers still another way of obtaining solid dispersions of active ingredients. In this technique a solution of the active ingredient and a polymer, optionally together with other ingredients, is sprayed thereby allowing the solvent to evaporate thus obtaining finely dispersed particles.
  • As mentioned above, the compound of formula (A) suffers from poor bioavailability thereby impeding its proper functioning. Therefore there is need to improve the bioavailability of this active ingredient so that its beneficial properties can have full play. A higher bioavailability moreover helps to reduce the size and number of dosage forms to be administered and this in turn contributes to an even further reduced pill burden. It now has been found that the hydrobromic acid-addition salt form of the compound of formula (A) has improved bioavailability and is particularly attractive when used in the form of solid dispersions, in particular in the form of solid dispersions prepared by the spray-drying technique.
  • Thus in one aspect of this invention, there is provided a solid dispersion of a compound of formula (I)
  • Figure US20100041687A1-20100218-C00002
  • in a water-soluble polymer. In one embodiment, the said solid dispersion is a spray-dried solid dispersion.
  • In another aspect there is provided the use of a compound of formula (I) for preparing a solid dispersion of the compound of formula (I) in a water-soluble polymer. In one embodiment said use is for preparing a solid dispersion of said compound of formula (I) by spray-drying.
  • In another aspect there is provided a method of preparing a solid dispersion of the compound of formula (I) in a solid polymeric matrix, said method comprising dispersing the compound of formula (I) into a solid polymeric matrix. In one embodiment said method is by spray-drying.
  • In a further aspect there is provided a pharmaceutical formulation comprising a solid dispersion of a compound of formula (I) in a solid polymeric matrix, and a carrier. In one embodiment, said formulation comprises a spray-dried solid dispersion of a compound of formula (I).
  • The compound of formula (I), i.e. 4-[[4-amino-5-bromo-6-(4-cyano-2,6-dimethyl-phenyloxy)-2-pyrimidinyl]amino]benzonitrile hydrobromide, which is the hydrobromide salt of the compound of formula (A), is deemed novel. Therefore, in a further aspect, the present invention concerns said compound of formula (I). In one embodiment, the compound of formula (I) is in amorphous form. In a further aspect of the invention there are provided pharmaceutical formulations comprising the compound of formula (I) and a carrier. In these formulations, the compound of formula (I) is present in an effective amount, i.e. an amount that is effective to inhibit HIV.
  • Contrary to a number of other salts of compound (A) that are unstable, the compound of formula (I) is a stable salt. For example the maleate and methane sulfonate salts of compound (A) have been found to be unstable, because of their partial or complete decomposition to compound (A) and the free acid.
  • The compound of formula (A) may be prepared according to the method described in WO 00/27825. The compound of formula (I) may be prepared by treating the compound of formula (A) with hydrobromic acid.
  • The compound of formula (I) is useful in the treatment of individuals infected by HIV. It can be used to inhibit HIV and additionally to treat conditions associated with HIV infection. The latter include AIDS as well as secondary conditions associated with HIV infection such as thrombocytopaenia, Kaposi's sarcoma, and infection of the central nervous system characterized by progressive demyelination, resulting in dementia and symptoms such as progressive dysarthria, ataxia, and disorientation. Other conditions associated with HIV infection comprise peripheral neuropathy, progressive generalized lymphadenopathy (PGL), and AIDS-related complex (ARC).
  • It has been found that the compound of formula (I) has increased bioavailability and that dosage forms comprising the compound of formula (I) advantageously have a lower pill burden.
  • The compound of formula (I) is preferably highly amorphous, that is, it has a low level of crystallinity. By “amorphous” is meant that the active agent is in a non-crystalline state. This again advantageously increases the bioavailability of the compound of formula (I) thereby decreasing the amount of compound that needs to be administered to a patient resulting in a reduced pill burden.
  • Preferably, the degree of crystallinity of the pharmaceutical agent in the solid pharmaceutical composition, as characterized by X-ray powder diffraction (XRPD), is about 20% or less, about 15% or less, about 10% or less, about 9% or less, about 8% or less, about 7% or less, about 6% or less, about 5% or less, about 4% or less, about 3% or less, about 2% or less, about 1% or less, about 0.5% or less, or about 0.1% or less.
  • The compound of formula (I) may be formulated into a dosage form for administration to a patient. Typical dosage forms include dosage forms for administration orally, rectally, vaginally, percutaneously or by parenteral injection. Suitable dosage forms for oral administration include tablets, capsules, powders, pills, suspensions, and solutions. Preferred dosage forms for oral administration are tablets and capsules. Suitable dosage forms for rectal or vaginal administration include gels, suppositories, or pastes.
  • The compound of formula (I) may be combined with any suitable pharmaceutically acceptable excipient appropriate for the particular dosage form being prepared. For example, for dosage forms for oral administration, suitable excipients include water, glycols, oils, alcohols, starches, sugars, kaolin, diluents, lubricants, binders, and disintegrating agents. Where the dosage form is a solution, polyethylene glycol is preferably used as the vehicle wherein the compound of formula (I) is dissolved. More preferably the polyethylene glycol is PEG 400.
  • The compound of formula (I) is particularly useful to formulate into a solid dispersion comprising the compound of formula (I), a pharmaceutically acceptable water-soluble polymer, and optionally one or more excipients. Thus formulated, the bioavailability of the compound of formula (I) is improved. Different types of solid dispersions exist. In a first type of solid dispersion the pharmaceutical agent is molecularly dispersed, substantially homogeneously, throughout the polymer. This is generally described as a “solid solution”. A second type of solid dispersion is where there are islands of crystalline or semi-crystalline pharmaceutical agent dispersed throughout the polymer. A third type of solid dispersion is where there are islands or clusters of pharmaceutical agent in amorphous form dispersed throughout the polymer.
  • Spray-drying offers an attractive technique for preparing solid dispersions because it is a continuous process that can be used at large scale. In the spray-drying process, the amount of the compound of formula (I) in the solution introduced to the spray-drying chamber is typically about 10% to about 60%, about 15% to about 50%, about 20% to about 40%, or about 25% to about 35% by weight compared to the total weight of the compound of formula (I), water-soluble polymer and optional excipients. The amount of compound dissolved is determined by its solubility in the solvent(s) chosen.
  • Polymers suitable for use in the solid dispersions of the compound of formula (I) comprise any that are pharmaceutically acceptable, water-soluble and substantially unreactive towards the pharmaceutical agent. Suitable polymers include cellulosic polymers, such as methyl cellulose, ethyl cellulose, hydroxymethyl cellulose, hydroxy-ethyl cellulose, hydroxypropyl cellulose, hydroxybutyl cellulose, hydroxyethylmethyl cellulose, hydroxypropylmethyl cellulose (HPMC), e.g. HPMC 2910, carboxymethyl cellulose, hydroxypropylmethyl cellulose phthalate (HPMCP), e.g. HP 50, hydroxy-propylmethyl cellulose acetate succinate (HPMCAS), cellulose acetate trimellitate (CAT), hydroxypropylcellulose acetate phthalate (HPCAP), hydroxypropylmethyl cellulose acetate phthalate (HPMCAP), methyl cellulose acetate phthalate (MCAP), and mixtures thereof such as a mixture of hydroxypropyl cellulose and ethyl cellulose. Suitable polymers also include polyvinyl pyrrolidone, copolyvidone, which is polyvinyl pyrrolidone copolymerized with vinyl acetate, and aminoalkyl methacrylate copolymers, such as Eudragit E® 100 (Röhm GmbH, Germany). Preferably, the polymer is hydroxypropylmethyl cellulose (HPMC), polyvinyl pyrrolidone or copolyvidone. A particularly preferred hydroxypropylmethyl cellulose is HPMC 2910 5 mPa·s. Particularly preferred polyvinyl pyrrolidones are PVP K12, PVP K30, or PVP K90 and a particularly preferred copolyvidone is PVP-co-VA64.
  • The amount of water-soluble polymer in the spray dried product may be in the range from about 30% to about 75%, in particular about 40% to about 75%, or about 50% to about 75% or about 60% to about 70%, by weight relative to the total weight of the spray dried product comprising compound (I), i.e. TMC125.HBr, water-soluble polymer and optional excipients. The amount of water-soluble polymer in the feed mixture can be calculated based on these percentages and on the amount of solvent used.
  • The weight:weight ratio of water-soluble polymer to the compound of formula (I) may be in the range from about 10:1 to about 1:10, in particular from about 10:1 to about 1:1, more in particular from about 5:1 to about 1:1, or from about 4:1 to about 1:1; or from about 3:1 to about 1:1, e.g. a ratio of about 3:1, of about 3.5:1, or of about 3.6:1. The ratio of water-soluble polymer to pharmaceutical agent is believed to affect the degree of crystallinity of the pharmaceutical agent in the resulting solid pharmaceutical composition. However, it is also desirable to reduce the amount of polymer in relation to the pharmaceutical agent in order to maximize the amount of pharmaceutical agent in the resulting pharmaceutical composition.
  • The solvent used in the spraying process may be any solvent that is inert with respect to the compound of formula (I) and that is able to dissolve the compound of formula (I) and the water-soluble polymer. Mixtures of solvents may be used. Suitable solvents include acetone, tetrahydrofuran (THF), dichloromethane, ethanol (anhydrous or aqueous), methanol, and combinations thereof. Where the polymer is HPMC, the solvent is preferably a mixture of dichloromethane and ethanol, more preferably a mixture of dichloromethane and ethanol, the latter in particular being anhydrous ethanol, in a 9:1 ratio by weight. Where the polymer is polyvinyl pyrrolidone or copolyvidone, the solvent is preferably acetone. The amount of solvent present in the feed mixture will be such that TMC125 and the water-soluble polymer are dissolved and that the feed mixture has sufficient low viscosity for it to be sprayed. In one embodiment the amount of solvent in the feed mixture will be at least 80%, in particular at least 90%, preferably at least 95%, the percentages expressing the weight amount of solvent to the total weight of the feed mixture.
  • Optionally, the feed mixture may contain further excipients such as to improve the properties of the feed mixture or the resulting solid pharmaceutical composition, for example to improve the handling or processing properties. The addition of excipients obviously results in these being incorporated in the solid dispersion. Regardless of whether or not excipients are added to the feed mixture, these may also be mixed with the resulting solid pharmaceutical composition during formulation into a desired dosage form.
  • One type of excipient that may be mixed into the feed mixture is microcrystalline cellulose (“MCC”). The microcrystalline cellulose (MCC) that can be used has an average particle size that is selected such that when mixed into the solution of pharmaceutical agent and water-soluble polymer, the resulting feed mixture is able to pass through the atomizing means into the spray-drying chamber without clogging or blocking the atomizer. As such, the size of the MCC is limited by the size of the atomizing means provided on the spray-drying chamber. For example, where the atomizing means is a nozzle, the size of the nozzle bore will affect the size range of the MCC that may be used. The average particle size of the MCC may be in the range of from 5 μm to 50 μm, in particular from 10 μm to 30 μm, e.g. about 20 μm.
  • Microcrystalline cellulose that can be used comprises the Avicel™ series of products available from FMC BioPolymer, in particular Avicel PH 105® (20 μm), Avicel PH 101® (50 μm), Avicel PH 301® (50 μm);
  • the microcrystalline cellulose products available from JRS Pharma, in particular Vivapur® 105 (20 μm), Vivapur® 101 (50 μm), Emcocel® SP 15 (15 μm), Emcocel® 50 M 105 (50 μm), Prosolv® SMCC 50 (50 μm);
    the microcrystalline cellulose products available from DMV, in particular Pharmacel® 105 (20 μm), Pharmacel® 101 (50 μm);
    the microcrystalline cellulose products available from Blanver, in particular Tabulose (Microcel)® 101 (50 μm), Tabulose (Microcel)® 103 (50 μm);
    the microcrystalline cellulose products available from Asahi Kasei Corporation, such as Ceolus® PH-F20JP (20 μm), Ceolus® PH-101 (50 μm), Ceolus® PH-301 (50 μm), Ceolus® KG-802 (50 μm).
  • A particularly preferred microcrystalline cellulose is Avicel PH 105® (20 μm).
  • The amount of MCC in the spray dried product may be in the range from about 5% to about 25%, in particular about 7.5% to about 20%, or about 10% to about 15% or about 10% to about 12.5%, by weight relative to the total weight of the spray dried product comprising TMC125, water-soluble polymer, MCC and optional excipients. The weight ratio of the amounts of MCC to TMC125 in the spray dried product can be calculated based on these percentages and in particular may be in the range of from about 2:1 to about 1:5, in particular from about 1:1 to 1:7, preferably about 1:2. The amount of MCC in the feed mixture can be calculated based on these percentages and on the amount of solvent used. In view of the desirability of keeping the concentration of pharmaceutical agent in the resulting solid pharmaceutical composition as high as possible, the concentration of MCC is preferably kept as low as possible.
  • A skilled person will be aware of the types of excipients suitable for inclusion in the feed mixture and/or in the resulting pharmaceutical composition. These include surfactants, solubilizers, disintegrants, pigments, flavorings, fillers, lubricants, glidants, preservatives, thickening agents, buffering agents and pH modifiers. Typical surfactants include sodium lauryl sulphate, Cremophor RH 40, Vitamin E TPGS, and polysorbates, such as Tween 20®. Typical pH modifiers are acids, such as citric acid or succinic acid, bases, or buffers.
  • The solid pharmaceutical composition produced by the process of the invention typically comprises particles having an average effective particle size in the range of from about 10 μm to about 150 μm, or about 15 μm to about 100 μm, particularly about 20 μm to about 80 μm, or 30 μm to about 50 μm, preferably about 40 μm. As used herein, the term average effective particle size has its conventional meaning as known to the person skilled in the art and can be measured by art-known particle size measuring techniques such as, for example, sedimentation field flow fractionation, photon correlation spectroscopy, laser diffraction or disk centrifugation. The average effective particle sizes mentioned herein may be related to weight distributions of the particles. In that instance, by “an average effective particle size of about 150 μm” it is meant that at least 50% of the total weight of the particles is composed of particles having a particle size of less than the effective average of 150 μm, and the same applies to the other effective particle sizes mentioned. In a similar manner, the average effective particle sizes may be related to volume distributions of the particles but usually this will result in the same or about the same value for the average effective particle size.
  • The so-called “span” of the spray-dried particles may be lower than about 3, in particular lower than about 2.5, e.g. the span is about 2. Usually the span will not be lower than about 1. As used herein the term “span” is defined by the formula (D90−D10)/D50 wherein D90 is the particle diameter corresponding to the diameter of particles that make up 90% of the total weight of all particles of equal or smaller diameter and wherein D50 and D10 are the diameters for 50 respectively 10% of the total weight of all particles.
  • The spray-dried particles may be formulated into a pharmaceutical formulation. The latter comprises the spray-dried particles and a carrier, which may comprise one or more pharmaceutically acceptable ingredients such as, for example, a lubricant, a binder, thickener, and the like ingredients. Where the formulation is a suspension or a paste, the spray-dried particles may be added to a suitable liquid, such as water.
  • The pharmaceutical formulations in turn may be converted into an appropriate dosage form. Typical dosage forms include dosage forms for oral administration, such as tablets, capsules, suspensions and pastilles, and dosage forms for rectal or vaginal administration, such as gels, suppositories or pastes. The spray-dried particles may be subjected to further processing steps depending on the nature of the final dosage form. For example, the pharmaceutical composition may be subjected to a post-drying process, or may undergo tabletting or encapsulating processes. Depending on whether the dosage form comprising the spray-dried particles is intended for immediate release or controlled release, further processing steps may be required, such as the incorporation of a disintegrant for immediate release products or the coating of the powder with an enteric layer for controlled release products. Suitable disintegrants include microcrystalline cellulose, starch, sodium starch glycolate, crosslinked carboxy-methylcellulose sodium, and crosslinked PVP.
  • The effective daily amount of the compound of formula (I) is from about 1 mg/kg to about 20 mg/kg body weight, or from about 2 mg/kg to about 10 mg/kg body weight, or from about 5 mg/kg to about 8 mg/kg body weight, or from about 5 mg/kg to about 6 mg/kg body weight. The effective daily amounts per patient can be obtained by multiplying the before-mentioned amounts by 70 or by the actual weight of the patient. A person skilled in the art will be able to determine the exact dose and frequency of administration required, which will depend on a number of factors, such as the severity of the condition being treated, the age and weight of the individual being treated and whether that individual is taking any other medication. The effective daily amount of the compound of formula (I) may be administered as a single dose or as two, three, four or more sub-doses at appropriate intervals throughout the day. The dose or sub-doses of the compound of formula (I) may be formulated as unit dosage forms containing a specified amount of the compound, typically from about 10 to about 1000 mg, from about 50 to about 800 mg, from about 100 to about 500 mg, from about 100 mg to about 300 mg, e.g. about 200 mg or about 240 mg of the compound of formula (I).
  • As used herein the term “about” has its usual meaning as understood by the person skilled in the relevant art. The term “about” may alternatively be interpreted as meaning that the numerical value mentioned in connection with the term “about” can deviate with +/−10%, or with +/−5%, or with +/−1%. All documents cited herein are incorporated by reference in their entirety. Specific embodiments of the present invention are now described, by way of example only, and should not be construed as limiting the invention thereto.
  • EXAMPLE 1 Preparation of Salts of the Compound of Formula (A)
  • The compound of formula (A), that is, 4-[[4-amino-5-bromo-6-(4-cyano-2,6-dimethyl-phenyloxy)-2-pyrimidinyl]amino]benzonitrile, was dissolved in dichloromethane and aqueous hydrobromic acid was added. The mixture was evaporated to dry yielding the hydrobromide salt of (A), i.e. the compound of formula (I). Other addition salts of the compound of formula (A) can be obtained similarly and are shown in the following Table 1.
  • Acid Salt of compound of formula (I) obtained
    Hydrobromic acid Hydrobromide (i.e. compound of formula (I))
    Hydrochloric acid Hydrochloride
    Maleic acid Maleate
    Methanesulfonic acid Methanesulfonate
    Fumaric acid Fumarate
    Sulfuric acid Sulfate
  • Capsules were filled with the various salts of the compound of formula (I) in the amounts shown in the following Table 2.
  • Salt of compound (I) Amount of salt per capsule in mg
    Hydrobromide 118.6
    Hydrochloride 108.4
    Maleate 63.3
    Methane sulfonate 122.1
    Fumarate 63.3
    Sulfate 122.5
  • EXAMPLE 2 In-Vivo Study of Bioavailability in Dogs of the Compound of Formula (I) Compared with Salts of the Compound of Formula (I)
  • Four male dogs were used per tested formulation. The animals were randomized over the test groups by a computer-generated random algorithm according to body weight, with all animals within ±20% of the sex mean. A health inspection was performed prior to commencement of treatment to ensure that the animals were in a good state of health. The dogs were subjected to an acclimatization period of at least 5 days before the start of treatment under laboratory conditions. A controlled environment was maintained in the room. The animals were dosed once orally with the formulations of example 1, and blood was collected from all animals under light ether anaesthesia at regular intervals for a period of 12, 24 or 48 hours after dosing.
  • The mean pharmacokinetic parameters relating to the mean plasma concentration with time of each test formulation are shown in Table 3. The standard deviation, where calculated, is shown between brackets. Analysis of the peak plasma concentration (Cmax) and bioavailability in terms of area under the curve (AUC0-last) shows that the hydrobromide salt of compound (I), that is the compound of formula (I) has advantageously improved bioavailability in dogs compared with other salts of compound (I). tmax is the time taken to reach the peak plasma concentration. tlast is the last time at which the peak plasma concentration was determined.
  • Methane-
    HBr Maleate sulfonate Fumarate Sulfate
    Parameter salt HCl salt salt salt salt salt
    tmax (h) 4.5 (2.1) 1.5 (0.7)  2.0 (—) 1.5 (0.7)  2.0 (—) 2.0 (0)  
    tlast (h) 48 (0)  48 (0)    24 (—) 48 (0)    12 (—) 48 (0) 
    AUC0-last 5855 (1456)  1875 (158.7) 446.5 (—)  3753 (803.3) 102.7 (—)  3143 (794.3)
    (ng · h/ml)
    (without dose
    normalization)
    Cmax (ng/ml) 365.5 (212.8) 222.0 (86.27) 52.00 (—) 409.0 (57.98)  15.2 (—) 294.0 (29.70)
    (without dose
    normalization)
    AUC0-last 516.1 (83.02) 168.2 (29.43) 77.70 (—) 318.8 (35.56) 20.67 (—) 279.9 (49.45)
    (ng · h/ml)
    (dose
    normalized to 1 mg/kg)
    Cmax (ng/ml) 31.73 (16.05) 20.19 (9.513   9.048 (—) 34.87 (1.337) 3.058 (—)  26.34 (0.6133)
    (dose
    normalized to 1 mg/kg)
  • EXAMPLE 3 Spray-Dried Formulation Comprising the Compound of Formula (I)
  • Formulation G
    Compound of formula (I)  59.3 mg
    HPMC 5 m Pa · s 177.9 mg
  • To prepare formulation G, the compound of formula (I) was dissolved in a first beaker in methanol/dichloromethane 50/50 v/v % at a concentration of 6.5 g per 750 ml of methanol/dichloromethane. HPMC 2910 5 mPa·s was dissolved in methanol/dichloro-methane 50/50 v/v % in a second beaker. The concentration of HPMC 2910 5 mPa·s in the solution was 6.5 g per 100 ml methanol/dichloromethane.
  • The two solutions were subsequently mixed and then spray-dried using the following spray-drying operating conditions:
  • Inlet air temperature: 80° C.
    Outlet air temperature: 40-55° C.
    Flow rate of the solution: 12 ml/min
    Air pressure: 1 bar
  • After spray-drying, the spray dried powder was dried under reduced pressure at 40° C. for 5 days. The resulting formulation was filled into Swedish-orange capsules.
  • An in-vivo dog study of the bioavailability of capsules comprising formulation G was carried out as described in Example 2. The mean pharmacokinetic parameters are shown in Table 6. The standard deviation, where calculated, is shown between brackets.
  • Parameter Formulation G
    tmax (h) 1.0 (1.0-2.0)
    tlast (h) 48 (0)
    AUC0-last (ng · h/ml) 13752 (2378)
    (without dose normalization)
    Cmax (ng/ml) 1263 (165.6)
    (without dose normalization)
    AUC0-last (ng · h/ml) 1179 (293.7)
    (dose normalized to 1 mg/kg)
    Cmax (ng/ml) 107.0 (15.38)
    (dose normalized to 1 mg/kg)
  • EXAMPLE 5 In-Vivo Study of Bioavailability in Humans of the Compound of Formula (I) Formulation L
  • To prepare formulation L, 118.6 mg of the compound of formula (I) was dissolved in a first beaker in methanol/dichloromethane 50/50 v/v % at a concentration of 6.5 g per 750 ml of methanol/dichloromethane In a second beaker, 234.8 mg HPMC 2910 5 mPa·s was dissolved in methanol/dichloromethane 50/50 v/v %. The concentration of HPMC 2910 5 mPa·s in the solution was 6.5 g per 100 ml methanol/dichloromethane. The two solutions were subsequently mixed and then sprayed onto 121 mg of HMPC 5 mPa·s. The resulting powder was filled into Swedish-orange capsules. One capsule of formulation L contained 118.6 mg of the compound of formula (I), which is equivalent to 100 mg of compound (A).
  • A randomized human bioequivalence trial was carried out to monitor bioavailability in humans of formulation L.
  • For formulation L, nine subjects were given 800 mg of formulation L twice daily for 13 days, with a single dose on day 14. A 12 h pharmacokinetic profile was determined on day 1 and on day 14. Pre-dose concentrations were determined every other day. The results are listed in the following Table.
  • Scheduled Plasma conc. of TMC125 (ng/ml)
    time Mean ± SD
    Day 1
    0 h Not quantifiable
    1 h  35.3 ± 33.1
    2 h  638 ± 451
    3 h 1220 ± 750
    4 h  136 ± 681
    6 h 1020 ± 395
    8 h  680 ± 285
    10 h   476 ± 182
    12 h   392 ± 166
    Day 3
    0 h 1230 ± 341
    Day 5
    0 h 1630 ± 513
    Day 7
    0 h 1830 ± 769
    Day 9
    0 h 1830 ± 720
    Day 11
    0 h 1940 ± 758
    Day 13
    0 h 1870 ± 740
    Day 14
    0 h 1660 ± 578
  • For formulation L, pre-dose levels reached steady-state between 3 to 5 days. Good bioavailability was observed.

Claims (10)

1. A solid dispersion comprising a compound of formula (I):
Figure US20100041687A1-20100218-C00003
in a water-soluble polymer.
2. The solid dispersion of claim 1, wherein the water-soluble polymer is selected from hydroxypropylmethyl cellulose, polyvinyl pyrrolidone, copolyvidone, and vitamin E TGPS.
3. The solid dispersion of claim 4, wherein the polymer is hydroxypropylmethyl cellulose 2910 5 cps
4. The solid dispersion of claim 3, wherein the ratio of the polymer to pharmaceutical agent is in the range from 9:1 to 1:1.
5. The solid dispersion of claim 6, wherein the ratio of polymer to pharmaceutical agent is in the range from 3:1 to 1:1.
6. A dosage form comprising the compound of claim 1 or claim 2 or the solid dispersion of claim 3.
7. The dosage form of claim 9, wherein the dosage form is selected from a tablet or a capsule.
8. A compound of formula (I):
Figure US20100041687A1-20100218-C00004
9. The compound of claim 1 having a degree of crystallinity of about 10% or less, or of about 5% or less, or of about 1% or less.
10. A pharmaceutical composition comprising a compound of formula (I) as defined in claim 8 and a carrier.
US12/517,665 2006-12-06 2007-12-06 Hydrobromide salt of an anti-hiv compound Abandoned US20100041687A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP06125547 2006-12-06
EP06125547.7 2006-12-06
PCT/EP2007/063386 WO2008068299A2 (en) 2006-12-06 2007-12-06 Hydrobromide salt of an anti-hiv compound

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2007/063386 A-371-Of-International WO2008068299A2 (en) 2006-12-06 2007-12-06 Hydrobromide salt of an anti-hiv compound

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/795,873 Continuation US11253479B2 (en) 2006-12-06 2017-10-27 Hydrobromide salt of an anti-HIV compound

Publications (1)

Publication Number Publication Date
US20100041687A1 true US20100041687A1 (en) 2010-02-18

Family

ID=37962213

Family Applications (2)

Application Number Title Priority Date Filing Date
US12/517,665 Abandoned US20100041687A1 (en) 2006-12-06 2007-12-06 Hydrobromide salt of an anti-hiv compound
US15/795,873 Active 2029-08-15 US11253479B2 (en) 2006-12-06 2017-10-27 Hydrobromide salt of an anti-HIV compound

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/795,873 Active 2029-08-15 US11253479B2 (en) 2006-12-06 2017-10-27 Hydrobromide salt of an anti-HIV compound

Country Status (5)

Country Link
US (2) US20100041687A1 (en)
EP (1) EP2104491B1 (en)
JP (1) JP5464584B2 (en)
ES (1) ES2624593T3 (en)
WO (1) WO2008068299A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110294802A1 (en) * 2007-12-17 2011-12-01 Mcinally Thomas Pharmaceutically acceptable salts of methyl (3-{ [[3-(6- amino- 2-butoxy-8-oxo-7, 8-dihydro-9h-purin-9-yl) propyl] (3- morpholin-4-ylpropyl) amino] methyl }phenyl) acetate and their use in therapy

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010006697A2 (en) * 2008-06-30 2010-01-21 Tibotec Pharmaceuticals Powders for reconstitution
EP2413936B1 (en) * 2009-03-30 2016-11-02 Janssen Sciences Ireland UC Co-crystal of etravirine and nicotinamide
WO2010131118A2 (en) * 2009-05-12 2010-11-18 Pliva Hrvatska D.O.O. Polymorphs of etravirine and processes for preparation thereof
US8153790B2 (en) 2009-07-27 2012-04-10 Krizmanic Irena Process for the preparation and purification of etravirine and intermediates thereof
GR1007010B (en) 2009-10-08 2010-10-07 Χημικα Και Βιοφαρμακευτικα Εργαστηρια Πατρων Αε (Cbl-Patras), INSULINOID Peptides
US20130123498A1 (en) 2011-10-19 2013-05-16 Maja Sepelj MAJER Process for the preparation of etravirine and intermediates in the synthesis thereof
RU2728555C1 (en) * 2019-12-24 2020-07-30 Общество с ограниченной ответственностью "Балтфарма" Method for producing etravirine

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050142188A1 (en) * 1998-07-17 2005-06-30 Janssen Pharmaceutica, N.V. Pellets having a core coated with an antifungal and a polymer

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1214013C (en) * 1998-11-10 2005-08-10 詹森药业有限公司 HIV replication inhibiting pyrimidines
ES2258018T3 (en) * 1999-09-24 2006-08-16 Janssen Pharmaceutica N.V. SOLID ANTIVIRAL DISPERSIONS.
EP1648467B1 (en) * 2003-07-17 2018-03-21 Janssen Sciences Ireland UC Process for preparing particles containing an antiviral

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050142188A1 (en) * 1998-07-17 2005-06-30 Janssen Pharmaceutica, N.V. Pellets having a core coated with an antifungal and a polymer

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Reier et al. ("Microcrystalline cellulose in tableting." Journal of Pharmaceutical Sciences 55.5 (1966): 510-514) *
Scholler et al. (Substantial improvement of oral bioavailability of TMC125 using new tablet formulations in healthy volunteers.. Poster Exhibition: The 3rd IAS Conference on HIV Pathogenesis and Treatment: Abstract no. TuPe3.1B11 July 24 - 27, 2005; Rio de Janeiro, Brazil). *
Siepmann et al. ("Modeling of drug release from delivery systems based on hydroxypropyl methylcellulose (HPMC)." Advanced drug delivery reviews 48.2 (2001): 139-157) *
Winston et al. ("The clinical pharmacology of antiretrovirals in development." (2006): 447-458) *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110294802A1 (en) * 2007-12-17 2011-12-01 Mcinally Thomas Pharmaceutically acceptable salts of methyl (3-{ [[3-(6- amino- 2-butoxy-8-oxo-7, 8-dihydro-9h-purin-9-yl) propyl] (3- morpholin-4-ylpropyl) amino] methyl }phenyl) acetate and their use in therapy
US8673907B2 (en) * 2007-12-17 2014-03-18 Astrazeneca Ab Pharmaceutically acceptable salts of methyl (3-{ [[3-(6-amino- 2-butoxy-8-oxo-7,8-dihydro-9H-purin-9-yl) propyl] (3-morpholin-4-ylpropyl) amino] methyl }phenyl) acetate and their use in therapy

Also Published As

Publication number Publication date
ES2624593T3 (en) 2017-07-17
EP2104491A2 (en) 2009-09-30
WO2008068299A3 (en) 2009-04-23
JP5464584B2 (en) 2014-04-09
US11253479B2 (en) 2022-02-22
WO2008068299A2 (en) 2008-06-12
US20180049986A1 (en) 2018-02-22
EP2104491B1 (en) 2017-02-22
JP2010511674A (en) 2010-04-15

Similar Documents

Publication Publication Date Title
US11253479B2 (en) Hydrobromide salt of an anti-HIV compound
US6517871B1 (en) Bioenhanced formulations comprising eprosartan in oral solid dosage form
US20230381194A1 (en) Suspension for oral administration comprising amorphous tolvaptan
JP2011516613A (en) An oral pharmaceutical composition in a solid dispersion, preferably comprising posaconazole and HPMCAS
US20100190809A1 (en) Combination formulations
EP2749270B1 (en) Racecadotril and pharmaceutical compositions thereof
US20150104511A1 (en) Pharmaceutical Antiretroviral Combinations Comprising Lamivudine, Festinavir and Nevirapine
JP5554571B2 (en) Powder for restoration
US9603803B2 (en) Process for preparing spray dried formulations of 4-[[6-amino-5-bromo-2-[(4-cyanophenyl)amino]-4-pyrimidinyl]oxy]-3,5-dimethylbenzonitrile
US20090088424A1 (en) Methods and compositions for controlling the bioavailability of poorly soluble drugs
US11576917B2 (en) Pharmaceutical compositions comprising Ibrutinib
EP1033974B1 (en) High drug load immediate and modified release oral dosage formulations and processes for their manufacture
US20160213569A1 (en) Pharmaceutical compositions of ranolazine and dronedarone
US20080182908A1 (en) Pharmaceutical compositions comprising memantine
WO2023220112A1 (en) Glp1 tablet compositions

Legal Events

Date Code Title Description
AS Assignment

Owner name: JANSSEN PHARMACEUTICA NV,BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:THONE, DANIEL JOSEPH CHRISTIAAN;REEL/FRAME:024378/0004

Effective date: 20080121

AS Assignment

Owner name: TIBOTEC PHARMACEUTICALS LTD., IRELAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:JANSSEN PHARMACEUTICA NV;REEL/FRAME:035455/0442

Effective date: 20080125

Owner name: JANSSEN R&D IRELAND, IRELAND

Free format text: CHANGE OF NAME;ASSIGNOR:TIBOTEC PHARMACEUTICALS;REEL/FRAME:035455/0639

Effective date: 20120106

Owner name: TIBOTEC PHARMACEUTICALS, IRELAND

Free format text: CHANGE OF NAME;ASSIGNOR:TIBOTEC PHARMACEUTICALS LTD.;REEL/FRAME:035466/0249

Effective date: 20080729

AS Assignment

Owner name: JANSSEN SCIENCES IRELAND UC, IRELAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:JANSSEN R & D IRELAND;REEL/FRAME:035496/0382

Effective date: 20141229

STCB Information on status: application discontinuation

Free format text: ABANDONED -- AFTER EXAMINER'S ANSWER OR BOARD OF APPEALS DECISION

AS Assignment

Owner name: JANSSEN SCIENCES IRELAND UNLIMITED COMPANY, IRELAND

Free format text: CHANGE OF NAME;ASSIGNOR:JANSSEN SCIENCES IRELAND UC;REEL/FRAME:058295/0511

Effective date: 20181008