WO2015071841A1 - Complexes of dabigatran and its derivatives, process for the preparation thereof and pharmaceutical compositions containing them - Google Patents

Complexes of dabigatran and its derivatives, process for the preparation thereof and pharmaceutical compositions containing them Download PDF

Info

Publication number
WO2015071841A1
WO2015071841A1 PCT/IB2014/065992 IB2014065992W WO2015071841A1 WO 2015071841 A1 WO2015071841 A1 WO 2015071841A1 IB 2014065992 W IB2014065992 W IB 2014065992W WO 2015071841 A1 WO2015071841 A1 WO 2015071841A1
Authority
WO
WIPO (PCT)
Prior art keywords
complex
dabigatran
sodium
diseases
acetate
Prior art date
Application number
PCT/IB2014/065992
Other languages
French (fr)
Inventor
Erzsébet Réka ANGI
Tamás SOLYMOSI
Richard Balázs Kárpáti
Zsófia FENYVESI
Zsolt ÖTVÖS
László MOLNÁR
Hristos Glavinas
Genovéva FILIPCSEI
Katalin Ferenczi
Gábor HELTOVICS
Original Assignee
Druggability Technologies Holdings Limited
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
Priority claimed from HU1300648A external-priority patent/HUP1300648A2/en
Priority claimed from HU1400084A external-priority patent/HUP1400084A2/en
Application filed by Druggability Technologies Holdings Limited filed Critical Druggability Technologies Holdings Limited
Publication of WO2015071841A1 publication Critical patent/WO2015071841A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/19Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles lyophilised, i.e. freeze-dried, solutions or dispersions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5138Organic macromolecular compounds; Dendrimers 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
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5146Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5146Organic macromolecular compounds; Dendrimers obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyamines, polyanhydrides
    • A61K9/5153Polyesters, e.g. poly(lactide-co-glycolide)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5192Processes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/06Antiarrhythmics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the invention is directed to a stable complex with increased apparent solubility and increased dissolution rate comprising as active compound Dabigatran, its esters, preferably Dabigatran etexilate, its salts, preferably Dabigatran etexilate mesylate, or derivatives thereof, which is useful in the treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis.
  • the complex of the present invention possesses increased apparent solubility in the whole physiologically relevant pH range resulting in increased absorption and higher overall bioavailability. This allows the reduction of dose which delivers significantly less anticoagulant to the GI tract.
  • the complex Dabigatran, its esters, preferably Dabigatran etexilate, its salts, preferably Dabigatran etexilate mesylate, or derivatives thereof of the present invention contains no tartaric acid or any other acid which further decreases the incidence of GI tract bleeding. This also makes the bioavailability of the compound independent of the way it is administered; the final dosage form is not sensitive to being broken, chewed, or opened.
  • the complex Dabigatran, its esters, preferably Dabigatran etexilate, its salts, preferably Dabigatran etexilate mesylate, or derivatives thereof of the present invention allows safer administration of a lower dose from the active ingredient.
  • Dabigatran was discovered from a panel of chemicals with similar structure to benzamidine- based thrombin inhibitor a-NAPAP (N-alpha-(2-naphthylsulfonylglycyl)-4- amidinophenylalanine piperidide), which had been known since the 1980s as a powerful inhibitor of various serine proteases, specifically thrombin, but also trypsin.
  • a-NAPAP N-alpha-(2-naphthylsulfonylglycyl)-4- amidinophenylalanine piperidide
  • Dabigatran etexilate mesylate is ⁇ -Alanine, N-[[2-[[[4-
  • Dabigatran etexilate mesylate is a yellow-white to yellow powder.
  • a saturated solution in pure water has a solubility of 1.8 mg/mL. It is freely soluble in methanol, slightly soluble in ethanol, and sparingly soluble in isopropanol.
  • the active substance is susceptible to hydrolysis in presence of humidity under acidic conditions. Batch analysis results confirm that alkyl methane sulfonates (genotoxic impurities) that could potentially be formed during manufacturing are not formed at detectable levels during the manufacturing of the finished product.
  • the 150 mg capsule (PRAXADA) for oral administration contains 172.95 mg Dabigatran etexilate mesylate, which is equivalent to 150 mg of Dabigatran etexilate, and the following pharmaceutically acceptable excipients: acacia, dimethicone, hypromellose, hydroxypropyl cellulose, talc, and tartaric acid.
  • the capsule shell is composed of carrageenan, FD&C Blue No. 2 (150 mg only), FD&C Yellow No.
  • the 75 mg capsule contains 86.48 mg dabigatran etexilate mesylate, equivalent to 75 mg Dabigatran etexilate, and is otherwise similar to the 150 mg capsule.
  • PRAXADA should be swallowed as a whole with water, with or without food. Patients have to be instructed not to open the capsule as this may increase the risk of bleeding.
  • Dabigatran and its acyl glucuronides are competitive, direct thrombin inhibitors. Because thrombin (serine protease) enables the conversion of fibrinogen into fibrin during the coagulation cascade, its inhibition prevents the development of a thrombus. Both free and clot-bound thrombin and thrombin-induced platelet aggregation are inhibited by the active moieties.
  • thrombin serine protease
  • Dabigatran etexilate mesylate is absorbed as the Dabigatran etexilate ester. The ester is then hydrolyzed, forming Dabigatran, the active moiety. Dabigatran is metabolized to four different acyl glucuronides and both the glucuronides and Dabigatran have similar pharmacological activity. Pharmacokinetics described here refer to the sum of Dabigatran and its glucuronides. Dabigatran displays dose-proportional pharmacokinetics in healthy subjects and patients in the range of doses from 10 to 400 mg.
  • Dabigatran etexilate is a substrate of the efflux transporter P-gp.
  • P-gp efflux transporter
  • C ma x occurs at 1 hour post-administration in the fasted state.
  • Coadministration of PRADAXA with a high-fat meal delays the time to C max by approximately 2 hours but has no effect on the bioavailability of Dabigatran; PRADAXA may be administered with or without food.
  • Dabigatran is approximately 35% bound to human plasma proteins.
  • the red blood cell to plasma partitioning of Dabigatran measured as total radioactivity is less than 0.3.
  • the volume of distribution of Dabigatran is 50 to 70 L.
  • Dabigatran pharmacokinetics are dose proportional after single doses of 10 to 400 mg. Given twice daily, Dabigatran's accumulation factor is approximately two.
  • Dabigatran is eliminated primarily in the urine. Renal clearance of Dabigatran is 80% of total clearance after intravenous administration. After oral administration of radiolabeled Dabigatran, 7% of radioactivity is recovered in urine and 86% in feces. The half-life of Dabigatran in healthy subjects is 12 to 17 hours.
  • Dabigatran etexilate After oral administration, Dabigatran etexilate is converted to Dabigatran.
  • the cleavage of the Dabigatran etexilate by esterase-catalyzed hydrolysis to the active principal Dabigatran is the predominant metabolic reaction.
  • Dabigatran is not a substrate, inhibitor, or inducer of CYP450 enzymes.
  • Dabigatran is subject to conjugation forming pharmacologically active acyl glucuronides.
  • Four positional isomers, 1-0, 2-0, 3-0, and 4-O-acylglucuronide exist, and each accounts for less than 10% of total Dabigatran in plasma.
  • the European Medicines Agency has recommended updating the product information for the blood thinner PRAXADA, to give clearer guidance to doctors and patients on how to reduce and manage the risk of bleeding associated with the anticoagulant medicine. Bleeding is a well-known complication of all anticoagulant medicines and PRAXADA has therefore been kept under close review by the Agency's Committee for Medicinal Products for Human Use (CHMP) since its initial authorization (25 May 2012, EMA/337406/2012).
  • CHMP Agency's Committee for Medicinal Products for Human Use
  • PRAXADA provides an important health benefit when used as directed. Healthcare professionals who prescribe PRAXADA should carefully follow the dosing recommendations in the drug label, especially for patients with renal impairment (when kidneys don't function normally) to reduce the risk of bleeding. Patients with atrial fibrillation should not stop taking PRAXADA without first talking to their healthcare professional. Stopping use of anticoagulant medications such as PRAXADA can increase the risk of stroke. Strokes can lead to permanent disability and death (FDA Drug Safety Communication of 12/19/2012).
  • the solubility of the Dabigatran ester is pH sensitive. Below pH 4.2 its solubility is actually appreciable. Above this pH it decreases rapidly reaching practically insoluble levels in the 6-7 pH range.
  • Dabigatran etexilate is available in capsule form (PRADAXA). It is formulated together with tartaric acid to reduce the variability of Dabigatran etexilate absorption, which is dependent on an acid environment. A Dabigatran etexilate coating is applied onto a tartaric acid core to form tiny pellets ( ⁇ lmm diameter). A single capsule contains hundreds of these pellets, the exact number depending on the dose strength of the capsule. In this way, Dabigatran etexilate absorption is independent of gastrointestinal tract acidity and is not materially affected by co- administration of a proton pump inhibitor. The oral bioavailability of Dabigatran etexilate increases by 75% when the pellets are taken without the capsule shell compared to the intact capsule formulation. PRADAXA capsules should therefore not be broken, chewed, or opened before administration.
  • the low systemic oral bioavailability is attributed to the low solubility of the compound at physiologically relevant pH.
  • the above formulation strategy ensures the absorption of the compound by creating a low pH milieu in the upper intestine (the first part of the duodenum) which dissolves the compound. At the lower parts of the GI track where pH is raised to the 6- 7 range the compound precipitates. This creates a short window (around 0.5 h) for the compound to get absorbed.
  • the absolute bioavailability of Dabigatran following oral administration of Dabigatran etexilate is approximately 3 to 7% even when using the above detailed formulation strategy.
  • WO2002/000196 WO2001/085345; US 8,074,906; US 7,441,717; US 6,921,458; US 6,641,063; US 5,389,382; WO2000/040220; WO2010/146406; WO2010/146408;
  • a stable complex comprising as active compound chosen from Dabigatran, its salts or derivatives thereof; and at least one complexation agent chosen from polyvinylcaprolactam-polyvinyl acetate-polyethylene-glycol graft copolymers and poloxamers; polyvinylpyrrolidone; polyoxyglicerides; copolymers of vinylpyrrolidone and vinyl-acetate; and poly(maleic acid-co-methyl-vinyl-ether); said complex characterized in that it possesses at least one of the following properties: having a particle size in the range between 50 nm and 600 nm, preferably 50 nm and 300 nm;
  • PAMPA permeability in the physiological pH range has a PAMPA permeability of at least 0.3* 10 "6 cm/s when dispersed in FaSSIF or FeSSIF biorelevant media;
  • ATR infrared
  • the invention is a complex formula having increased and pH independent permeability in the physiological relevant pH range and enhanced biological performance including higher C ma x and AUC, faster onset of action and decreased fed/fasted effect.
  • Dabigatran is generally used for Dabigatran, its esters, such as Dabigatran etexilate or its salts such as Dabigatran etexilate mesylate, Dabigatran etexilate methanesulfonate, Dabigatran etexilate bismesylate or derivatives of Dabigatran.
  • said complexation agent is chosen from polyvinylcaprolactam-polyvinyl acetate-polyethylene-glycol graft copolymers and poloxamers; polyvinylpyrrolidone; polyoxyglicerides; copolymers of vinylpyrrolidone and vinyl-acetate; and poly(maleic acid- co-methyl -vinyl -ether) .
  • said complex further comprises at least one pharmaceutically acceptable excipient selected from the group of sodium-acetate, sodium-lauryl-sulfate, dioctyl sodium sulfosuccinate, cetylpyridinium chloride and sodium saccharine.
  • said complex has a controlled particle size in the range between 50 nm and 600 nm. In an embodiment, said particle size is between 50 nm and 300 nm.
  • said complex further comprises one or more additional active agents.
  • said additional active agent is chosen from agents useful for the prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis.
  • said complex is orally available.
  • said complex possesses at least two of the properties described in a) - f).
  • said complex possesses at least three of the properties described in a) - f).
  • said complex has an increased dissolution rate.
  • a stable complex comprising an active compound selected from the group of Dabigatran, its salt, or derivatives thereof; at least one complexation agent chosen from polyvinylcaprolactam-polyvinyl acetate-polyethylene-glycol graft copolymers and poloxamers; polyvinylpyrrolidone; polyoxyglicerides; copolymers of vinylpyrrolidone and vinyl-acetate; and poly(maleic acid-co-methyl-vinyl-ether); and at least one pharmaceutically acceptable excipient chosen from sodium-acetate, sodium-lauryl-sulfate, dioctyl sodium sulfosuccinate, cetylpyridinium chloride and sodium saccharine; wherein said complex obtained via a mixing process.
  • said complex is obtained via a continuous flow mixing process.
  • a complex comprises a complexation agent and a pharmaceutically acceptable excipient in a total amount ranging from about 1.0 weight% to about 95.0 weight % based on the total weight of the complex.
  • said complexation agent and pharmaceutically acceptable excipient comprise 50 weight% to about 95 weight% of the total weight of the complex.
  • a process for the preparation of the complex comprising the steps of mixing a solution of Dabigatran, its salt, or derivatives thereof; and at least one complexation agent chosen from polyvinylcaprolactam-polyvinyl acetate-polyethylene-glycol graft copolymers and poloxamers; polyvinylpyrrolidone; polyoxyglicerides; copolymers of vinylpyrrolidone and vinyl-acetate; and poly(maleic acid-co-methyl-vinyl-ether) in a pharmaceutically acceptable solvent with an aqueous solution containing and at least one pharmaceutically acceptable excipient chosen from sodium-acetate, sodium-lauryl-sulfate, dioctyl sodium sulfosuccinate, cetylpyridinium chloride and sodium saccharine.
  • said process is performed in a continuous flow instrument.
  • said continuous flow instrument is a microfluidic flow instrument.
  • said pharmaceutically acceptable solvent is chosen from methanol, ethanol, i-propanol, n-propanol, acetone, acetonitrile, dimethyl-sulfoxyde, or tetrahydrofuran, or combinations thereof.
  • said pharmaceutically acceptable solvent and said aqueous solvent are miscible with each other.
  • said aqueous solvent comprises 0.1 to 99.9% weight of the final solution.
  • said aqueous solvent comprises 50 to 90% weight of the final solution.
  • said aqueous solvent comprises 50 to 80% weight of the final solution.
  • said aqueous solvent comprises 50 to 70% weight of the final solution. In an embodiment, said aqueous solvent comprises 50 to 60% weight of the final solution.
  • said aqueous solvent comprises 50 % weight of the final solution.
  • a pharmaceutical composition comprising the complex together with pharmaceutically acceptable carrier.
  • said composition is suitable for oral, pulmonary, rectal, colonic, parenteral, intracisternal, intravaginal, intraperitoneal, ocular, otic, local, buccal, nasal, or topical administration. In an embodiment, said composition is suitable for oral administration.
  • said complex is for use in the manufacture of a medicament for the treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis.
  • diseases in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis.
  • said complex is used for the treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis.
  • diseases in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis.
  • a method of treatment and/or prophylaxis of diseases comprises administration of a therapeutically effective amount of a complex or a pharmaceutical composition as described herein.
  • a method for reducing the therapeutically effective dosage of Dabigatran compared to orally available formulations comprises oral administration of a pharmaceutical composition as described herein.
  • a stable complex wherein said complex has a controlled particle size in the range between 50 nm and 600 nm; and wherein said complex is not obtained via a milling process or by high pressure homogenization process, encapsulation process and solid dispersion process, but it is obtained by a mixing process, preferable continuous flow mixing process.
  • said particle size is between 50 nm and 300 nm.
  • said complex shows reduced fed/fasted effect based on in vivo studies.
  • said complex shows significantly improved exposure, earlier t max , higher Cmax which will allow the oral administration and reduction of the dose.
  • said complex has a faster onset of action compared to the marketed formulations. In an embodiment, said complex is instantaneously redispersable in physiological relevant media.
  • said complex is stable in solid form and in colloid solution and/or dispersion. In an embodiment, said complex exhibits pH independent PAMPA permeability in the physiological pH range.
  • said complex has a PAMPA permeability of at least 0.3* 10 "6 cm/s when dispersed in FaSSIF or FeSSIF biorelevant media
  • said complex is characterized by infrared (ATR) spectrum having charateristic absorption peaks at 1730 cm “1 , 1608 cm “1 , 1468 cm “1 , 1437 cm “1 , 1384 cm “1 , 1328 cm “1 , 1252 cm “1 , 1193 cm “1 , 1144 cm “1 , 1118 cm “1 , 1098 cm “1 , 1052 cm “1 , 993 cm “1 , 948 cm “1 , 888 cm “1 , 837 cm “1 , 811 768 cm “1 , 754 cm “1 , 704 cm “1 , 680 cm “1 , 632 cm “1 , 604 cm “1 , 567 cm “1 , 545 cm “1 and 524 cm “1 , preferable at 1730 cm “1 , 1608 cm “1 , 1437 cm “1 , 1328 cm “1 ,! 118 cm “1 , 1098 cm “1 , and 754
  • the complexation agents and pharmaceutically acceptable excipients of the Dabigatran complex formulae of the invention are selected from the group of pharmaceutically acceptable nonionic, anionic, cationic, ionic polymers, surfactants and other types of excipients.
  • the complexation agents themselves or together with the pharmaceutically accepted excipients have the function to form a complex structure with an active pharmaceutical ingredient through non-covalent secondary interactions.
  • the secondary interactions can form through electrostatic interactions such as ionic interactions, H-bonding, dipole-dipole interactions, dipole-induced dipole interactions, London dispersion forces, ⁇ - ⁇ interactions, and hydrophobic interactions.
  • compositions may additionally include one or more pharmaceutically acceptable excipients, auxiliary materials, carriers, active agents or combinations thereof.
  • active agents may include agents useful for the treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis.
  • Another aspect of the invention is the complex formulae of the Dabigatran with complexation agents and pharmaceutically acceptable excipients in which the complexation agents and pharmaceutically acceptable excipients preferably are associated or interacted with the Dabigatran especially as the results of the mixing process, preferably continuous flow mixing process.
  • the structure of the complex Dabigatran formula is different from the core-shell type milled particle, precipitated encapsulated particles, micelles and solid dispersions.
  • the pharmaceutical composition of the invention can be formulated: (a) for administration selected from the group consisting of oral, pulmonary, rectal, colonic, parenteral, intracisternal, intravaginal, intraperitoneal, ocular, otic, local, buccal, nasal, and topical administration; (b) into a dosage form selected from the group consisting of liquid dispersions, gels, aerosols, ointments, creams, lyophilized formulations, tablets, capsules; (c) into a dosage form selected from the group consisting of controlled release formulations, fast melt formulations, delayed release formulations, extended release formulations, pulsatile release formulations, and mixed immediate release and controlled release formulations; or (d) any combination of (a), (b), and (c).
  • compositions can be formulated by adding different types of excipients for oral administration in solid, liquid, local (powders, ointments or drops), or topical administration, and the like.
  • compositions can be formulated by adding different types of pharmaceutically acceptable excipients for oral administration in solid, liquid, local (powders, ointments or drops), or topical administration, and the like.
  • a preferred dosage form of the invention is a solid dosage form, although any pharmaceutically acceptable dosage form can be utilized.
  • Solid dosage forms for oral administration include, but are not limited to, capsules, tablets, pills, powders, and granules.
  • the active agent is admixed with at least one of the following excipients: (a) one or more inert excipients (or carriers), such as sodium citrate or dicalcium phosphate; (b) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, microcrystalline cellulose and silicic acid; (c) binders, such as cellulose derivatives, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (d) humectants, such as glycerol; (e) disintegrating agents, such as crospovidon, sodium starch glycolate, effervescent compositions, croscarmellose sodium,, calcium carbonate, potato or tap
  • composition can also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • compositions of the invention include, but are not limited to (1) physical and chemical stability, (2) instantaneous redispersibility, (3) stability in colloid solution or dispersion in the therapeutic time window, (4) increased solubility compared to the conventional Dabigatran formulation at physiologically relevant pH, (5) increased and pH independent permeability in the physiological relevant pH range, (6) possibility of dose reduction, (7) reduced GI tract bleeding adverse effect due to decreased anticoagulant dose and no strong acid necessary to be added to the formula and (8) good processability.
  • One of the preferred characteristics of the complex Dabigatran formulae of the present invention is their increased apparent solubility and pH independent increased permeability.
  • the permeability of the complex Dabigatran formulae is at least 0.574* 10 " 6 cm/s, 0.376* 10 "6 cm/s and 0.421 * 10 "6 cm/s in the 5-8.5 pH range, respectively.
  • the invention relates to a method for the treatment of a subject in need for the treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesisby administering to the subject an effective amount of the complex or the pharmaceutical composition according to the invention.
  • diseases in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia
  • oncological diseases of any pathogenesis by administering to the subject an effective amount of the complex or the pharmaceutical composition according to the invention.
  • Another preferred characteristic of the complex Dabigatran formulae of the present invention relates to the enhanced pharmacokinetic performance of the complex Dabigatran formulae.
  • the complex Dabigatran is available for absorption in the whole GI tract extending the window for absorption from 0.5 hour to 4-5 hours and allows safer administration of a lower dose from the active ingredient.
  • Figure 1 shows complexation agent and pharmaceutically acceptable excipient screening for formula selection
  • Figure 2. shows particle size of the redispersed solid complex of the selected formula Figure 3. shows SEM photos of complex Dabigatran (A) and placebo sample (B)
  • Figure 4 shows infrared (ATR) spectra of Crystalline Dabigatran etexilate (A), amorphous Dabigatran etexilate (B), complex Dabigatran exeilate (C), placebo (D), Gelucire 44/14 (E), CPC (F) and Sacharine (G)
  • Figure 5 shows comparative PAMPA assays of complex Dabigatran formula and the marketed compound (Pradaxa)
  • a colloid solution of Dabigatran complex formula with the optimal ratio of the pharmaceutically acceptable excipients of the present invention was prepared by continuous flow mixing in a flow instrument.
  • 200 mg Dabigatran etexilate and 600 mg polyoxygliceride (Gelucire 44/14) dissolved in 100 mL tetrahydrofuran was used.
  • the prepared solution was passed into the instrument with 2 mL/min flow rate.
  • aqueous solvent containing 50 mg cetylpyridinium chloride and 250 mg sodium saccharine in 500 mL water was passed into the instrument with 8 mL/min flow rate, where Dabigatran formed complex Dabigatran composition.
  • the colloid solution of the complex Dabigatran is continuously produced at atmospheric pressure.
  • the produced colloid solution was frozen on dry-ice and then it was lyophilized using a freeze drier equipped with -110°C ice condenser, with a vacuum pump. Particle size of complex Dabigatran was measured in reconstituted colloid solution right after the lyophilisation. It was found to be 156 nm as shown in Figure 2.
  • Dabigatran etexilate Continuous flow mixing of Dabigatran etexilate in the presence of selected complexation agents and pharmaceutically accepted excipients, such as polyoxygliceride (Gelucire 44/14), cetylpyridinium chloride and sodium saccharine resulted in a stable complex of Dabigatran as shown in Figure 4.
  • complexation agents such as polyoxygliceride (Gelucire 44/14), cetylpyridinium chloride and sodium saccharine
  • the complex or the pharmaceutical composition according to the invention characterized by at least one of the following characteristic infrared (ATR ) peak at 1730 cm “1 , 1608 cm “1 , 1468 cm “1 , 1437 cm “1 , 1384 cm “1 , 1328 cm “1 , 1252 cm “1 , 1193 cm “1 , 1144 cm “1 , 1118 cm “1 , 1098 cm “ 1052 cm “1 , 993 cm “1 , 948 cm “1 , 888 cm “1 , 837 cm “1 , 811 768 cm “1 , 754 cm “1 , 704 cm “1 , 680 cm “1 , 632 cm “1 , 604 cm “1 , 567 cm _1 ,_545 cm “1 and 524 cm “1 , preferable at 1730 cm “1 , 1608 cm “ cm “1 , 1328 cm “1 , 1118 cm “1 , 1098 cm “1 , and 754 cm
  • PAMPA permeability measurements were performed as described by M. Kansi et al. (Journal of medicinal chemistry, 41, (1998) pp 1007) with modifications based on S. Bendels et al (Pharmaceutical research, 23 (2006) pp 2525).
  • Sample containing the reference compound was a suspension of crystals visible by the naked eye, while samples of the novel complex were opalescent colloid solutions.
  • Permeability was measured in a 96-well plate assay across an artificial membrane composed of dodecane with 20% soy lecithin supported by a PVDF membrane (Millipore, USA).
  • the receiver compartment was phosphate buffered saline (pH 7.0) supplemented with 1% sodium dodecyl sulfate.
  • the assay was performed at room temperature; incubation time was 1-24 hours.
  • the concentration in the receiver compartment was determined by UV-VIS spectrophotometry (Thermo Scientific Genesys S10).
  • PAMPA permeability of complex Dabigatran formula of Example 2 was 0.574* 10 "6 cm/s, 0.376* 10 "6 cm/s and 0.421 * 10 "6 cm/s when dispersed in a buffer pH 5.0, 6.5 or 8.5, respectively, while permeability of the marketed compound (Pradaxa) was 0.094* 10 "6 cm/s, 0.060* 10 "6 cm/s and 0.020* 10 "6 cm/s when dispersed in the same buffers as shown in Figure 5.
  • the test item was administered to the rats orally by gavage.
  • the granules of the reference item were administered through a plastic gavage into the stomach of the rats.
  • the granules were pushed out with the help of a mandrin then water was administered in a same amount as the test item administration amount.
  • Blood was collected in the following time points using sodium heparin as anticoagulant: 15m, 30m, 45m, lh, 1.5h, 2h, 4h, 6h, and 8h.
  • the plasma was separated and stored in an ultra- freezer (below -70°C) until analysis.
  • Dabigatran plasma kinetics showed high inter-individual variability after dosing the reference item.
  • the shape of the individual curves and the t max values (0.25h - 4h) showed very high variability and the C max values varied in a range (80.6 ⁇ 37.0).

Abstract

The present invention relates to pharmaceutically acceptable complex formulae comprising complexes of Dabigatran, its esters, preferably Dabigatran etexilate, its salts, preferably 5 Dabigatran etexilate mesylate, or derivatives thereof and complexation agents and pharmaceutically accepted excipients, process for the preparation thereof and pharmaceutical compositions containing them. The complex formulae of the present invention have improved physicochemical properties and enhanced biological performance and are useful in the treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis. More specifically, the complex of the present invention possesses increased apparent solubility in the whole physiologically relevant pH range resulting in increased absorption and higher overall bioavailability. This allows the reduction of dose which delivers significantly less anticoagulant to the GI tract. Furthermore, the complex composition of the present invention contains no tartaric acid or any other acid which further decreases the incidence of GI tract bleeding. This also makes the bioavailability of the compound independent of the way it is administered; the final dosage form is not sensitive to being broken, chewed, or opened. Overall, the complex Dabigatran composition of the present invention allows safer administration of a lower dose from the active ingredient.

Description

COMPLEXES OF DABIGATRAN AND ITS DERIVATIVES, PROCESS FOR THE PREPARATION THEREOF AND PHARMACEUTICAL COMPOSITIONS
CONTAINING THEM
FIELD OF THE INVENTION
The invention is directed to a stable complex with increased apparent solubility and increased dissolution rate comprising as active compound Dabigatran, its esters, preferably Dabigatran etexilate, its salts, preferably Dabigatran etexilate mesylate, or derivatives thereof, which is useful in the treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis. More specifically, the complex of the present invention possesses increased apparent solubility in the whole physiologically relevant pH range resulting in increased absorption and higher overall bioavailability. This allows the reduction of dose which delivers significantly less anticoagulant to the GI tract. Furthermore, the complex Dabigatran, its esters, preferably Dabigatran etexilate, its salts, preferably Dabigatran etexilate mesylate, or derivatives thereof of the present invention contains no tartaric acid or any other acid which further decreases the incidence of GI tract bleeding. This also makes the bioavailability of the compound independent of the way it is administered; the final dosage form is not sensitive to being broken, chewed, or opened. Overall, the complex Dabigatran, its esters, preferably Dabigatran etexilate, its salts, preferably Dabigatran etexilate mesylate, or derivatives thereof of the present invention allows safer administration of a lower dose from the active ingredient.
BACKGROUND OF THE INVENTION
Background Regarding Dabigatran Dabigatran was discovered from a panel of chemicals with similar structure to benzamidine- based thrombin inhibitor a-NAPAP (N-alpha-(2-naphthylsulfonylglycyl)-4- amidinophenylalanine piperidide), which had been known since the 1980s as a powerful inhibitor of various serine proteases, specifically thrombin, but also trypsin. Addition of ethyl ester and hexyloxycarbonyl carbamide hydrophobic side chains led to the orally absorbed prodrug. In order to improve the solubility of the prodrug its salts form was formulated as final drug product.
The chemical name for Dabigatran etexilate mesylate, is β-Alanine, N-[[2-[[[4-
[[[(hexyloxy)carbonyl]amino]iminomethyl] I ] ami no] methyl ] - 1 -methyl - 1 H- benzimidazol-5-yl]carbonyl]-N-2-pyridinyl-,ethyl ester, methanesulfonate. The empirical formula is C34H41N7O5 · CH4O3S and the molecular weight is 723.86 (mesylate salt), 627.75 (free base). The structural formula is:
Figure imgf000003_0001
Dabigatran etexilate mesylate is a yellow-white to yellow powder. A saturated solution in pure water has a solubility of 1.8 mg/mL. It is freely soluble in methanol, slightly soluble in ethanol, and sparingly soluble in isopropanol.
Based on these physicochemical and biopharmaceutical properties of Dabigatran etexilate mesylate and the clinical requirements for a reliable drug release a multilayer pellet was developed. Other formulation approaches were evaluated during early development, but were inferior to the selected formulation in terms of drug load, stability and in vivo performance.
The active substance is susceptible to hydrolysis in presence of humidity under acidic conditions. Batch analysis results confirm that alkyl methane sulfonates (genotoxic impurities) that could potentially be formed during manufacturing are not formed at detectable levels during the manufacturing of the finished product. The 150 mg capsule (PRAXADA) for oral administration contains 172.95 mg Dabigatran etexilate mesylate, which is equivalent to 150 mg of Dabigatran etexilate, and the following pharmaceutically acceptable excipients: acacia, dimethicone, hypromellose, hydroxypropyl cellulose, talc, and tartaric acid. The capsule shell is composed of carrageenan, FD&C Blue No. 2 (150 mg only), FD&C Yellow No. 6, hypromellose, potassium chloride, titanium dioxide, and black edible ink. The 75 mg capsule contains 86.48 mg dabigatran etexilate mesylate, equivalent to 75 mg Dabigatran etexilate, and is otherwise similar to the 150 mg capsule.
PRAXADA should be swallowed as a whole with water, with or without food. Patients have to be instructed not to open the capsule as this may increase the risk of bleeding.
Dabigatran and its acyl glucuronides are competitive, direct thrombin inhibitors. Because thrombin (serine protease) enables the conversion of fibrinogen into fibrin during the coagulation cascade, its inhibition prevents the development of a thrombus. Both free and clot-bound thrombin and thrombin-induced platelet aggregation are inhibited by the active moieties.
Dabigatran etexilate mesylate is absorbed as the Dabigatran etexilate ester. The ester is then hydrolyzed, forming Dabigatran, the active moiety. Dabigatran is metabolized to four different acyl glucuronides and both the glucuronides and Dabigatran have similar pharmacological activity. Pharmacokinetics described here refer to the sum of Dabigatran and its glucuronides. Dabigatran displays dose-proportional pharmacokinetics in healthy subjects and patients in the range of doses from 10 to 400 mg.
The absolute bioavailability of Dabigatran following oral administration of Dabigatran etexilate is approximately 3 to 7%. Dabigatran etexilate is a substrate of the efflux transporter P-gp. After oral administration of Dabigatran etexilate in healthy volunteers, Cmax occurs at 1 hour post-administration in the fasted state. Coadministration of PRADAXA with a high-fat meal delays the time to Cmax by approximately 2 hours but has no effect on the bioavailability of Dabigatran; PRADAXA may be administered with or without food.
The oral bioavailability of Dabigatran etexilate increases by 75% when the pellets are taken without the capsule shell compared to the intact capsule formulation. PRADAXA capsules should therefore not be broken, chewed, or opened before administration.
Dabigatran is approximately 35% bound to human plasma proteins. The red blood cell to plasma partitioning of Dabigatran measured as total radioactivity is less than 0.3. The volume of distribution of Dabigatran is 50 to 70 L. Dabigatran pharmacokinetics are dose proportional after single doses of 10 to 400 mg. Given twice daily, Dabigatran's accumulation factor is approximately two. Dabigatran is eliminated primarily in the urine. Renal clearance of Dabigatran is 80% of total clearance after intravenous administration. After oral administration of radiolabeled Dabigatran, 7% of radioactivity is recovered in urine and 86% in feces. The half-life of Dabigatran in healthy subjects is 12 to 17 hours.
After oral administration, Dabigatran etexilate is converted to Dabigatran. The cleavage of the Dabigatran etexilate by esterase-catalyzed hydrolysis to the active principal Dabigatran is the predominant metabolic reaction. Dabigatran is not a substrate, inhibitor, or inducer of CYP450 enzymes. Dabigatran is subject to conjugation forming pharmacologically active acyl glucuronides. Four positional isomers, 1-0, 2-0, 3-0, and 4-O-acylglucuronide exist, and each accounts for less than 10% of total Dabigatran in plasma.
The most commonly reported adverse reactions are bleedings occurring in total in approximately 16.5 % in patients with atrial fibrillation treated for the prevention of stroke and systemic embolic events. A dose response with respect to bleeding was seen for Dabigatran etexilate. Major or severe bleeding may occur and, regardless of location, may lead to disabling, life-threatening or even fatal outcomes.
The European Medicines Agency has recommended updating the product information for the blood thinner PRAXADA, to give clearer guidance to doctors and patients on how to reduce and manage the risk of bleeding associated with the anticoagulant medicine. Bleeding is a well-known complication of all anticoagulant medicines and PRAXADA has therefore been kept under close review by the Agency's Committee for Medicinal Products for Human Use (CHMP) since its initial authorization (25 May 2012, EMA/337406/2012).
FDA has not changed its recommendations regarding PRAXADA. PRAXADA provides an important health benefit when used as directed. Healthcare professionals who prescribe PRAXADA should carefully follow the dosing recommendations in the drug label, especially for patients with renal impairment (when kidneys don't function normally) to reduce the risk of bleeding. Patients with atrial fibrillation should not stop taking PRAXADA without first talking to their healthcare professional. Stopping use of anticoagulant medications such as PRAXADA can increase the risk of stroke. Strokes can lead to permanent disability and death (FDA Drug Safety Communication of 12/19/2012). The solubility of the Dabigatran ester is pH sensitive. Below pH 4.2 its solubility is actually appreciable. Above this pH it decreases rapidly reaching practically insoluble levels in the 6-7 pH range.
Dabigatran etexilate is available in capsule form (PRADAXA). It is formulated together with tartaric acid to reduce the variability of Dabigatran etexilate absorption, which is dependent on an acid environment. A Dabigatran etexilate coating is applied onto a tartaric acid core to form tiny pellets (~lmm diameter). A single capsule contains hundreds of these pellets, the exact number depending on the dose strength of the capsule. In this way, Dabigatran etexilate absorption is independent of gastrointestinal tract acidity and is not materially affected by co- administration of a proton pump inhibitor. The oral bioavailability of Dabigatran etexilate increases by 75% when the pellets are taken without the capsule shell compared to the intact capsule formulation. PRADAXA capsules should therefore not be broken, chewed, or opened before administration.
The low systemic oral bioavailability is attributed to the low solubility of the compound at physiologically relevant pH. The above formulation strategy ensures the absorption of the compound by creating a low pH milieu in the upper intestine (the first part of the duodenum) which dissolves the compound. At the lower parts of the GI track where pH is raised to the 6- 7 range the compound precipitates. This creates a short window (around 0.5 h) for the compound to get absorbed. As a result of this pH dependent solubility the absolute bioavailability of Dabigatran following oral administration of Dabigatran etexilate is approximately 3 to 7% even when using the above detailed formulation strategy. This makes it necessary to administer high doses which deliver substantial amount of tartaric acid and an anticoagulant to the GI tract. This results in GI tract bleeding as a very common adverse effect of the drug. The risk of major bleeds was similar with PRADAXA 150 mg and warfarin across major subgroups defined by baseline characteristics, with the exception of age, where there was a trend towards a higher incidence of major bleeding on PRADAXA (hazard ratio 1.2, 95% CI: 1.0 to 1.4) for patients >75 years of age. There was a higher rate of major gastrointestinal bleeds in patients receiving PRADAXA 150 mg than in patients receiving warfarin (1.6% vs. 1.1%, respectively, with a hazard ratio vs. warfarin of 1.5, 95% CI, 1.2 to 1.9), and a higher rate of any gastrointestinal bleeds (6.1% vs. 4.0%, respectively). It has been hypothesized that tartaric acid may promote GI bleeding through caustic injury, this is likely to be the case because the anti-platelet medication Aggrenox® (aspirin and dipyridamole), which is formulated with similar amounts of tartaric acid, is also associated with increased GI bleeding.
A variety of strategies have been used to attempt to overcome these issues, see for example US 5, 145,684; US 5,518,187; US 5,862,999; US 5,718,388; US 6,431,478; US 6,582,285; US
6,742,734; US2006/0165806; WO2009/075401; US 8,092,785; WO2003/002258; US
7,951,399; US 7,575, 184; US 6,814,319; WO0032313; WO2011/131943; WO2012/042224,
WO2006/060698; WO2006/096462; US2006/0159766; US2006/0188566; WO2006/110811;
WO2002/000196; WO2001/085345; US 8,074,906; US 7,441,717; US 6,921,458; US 6,641,063; US 5,389,382; WO2000/040220; WO2010/146406; WO2010/146408;
WO2010/146409; WO2011/158053; WO2010/146407; WO2011/154755; WO2011/158052;
WO2009/133418; US 5,091, 187; US 6,607,784; US 5,780,062; US 5,560,932; US 4,540,602;
US 5, 133,908; US 5,716,642; US2002/0127278; US 7,244,451; US2005/020294;
WO2009/007992; WO2007/150030; US 6,018,080; WO2001/003670; US2002/0168402; WO2006/100281; US2006/0013888; US 5, 141,961; US 5,646, 131; US 4,894,235; US
2003/0104065; US 5,646, 131; US 6,596,263; US 6,699,849; EP1348432; US 5324718;
WO2000/016744; WO2005/037250; WO2012/160559; WO2006/021160; WO2008/002121;
WO2006/112541; US 8, 158,162; WO2010/133611; US 7,550, 158; US 6,706,283; US
8,354, 122; EP1027886; US 6,677,362; US 5,340,591; US 7,994,214; US 8,337,899; US 8,343,548; US 4,996,047; US 5,980,882; EP 0911039; US 6,436,912; US 6,458,347; US
6,291,671; US 5,324,718; US 6,699,849; US 6,406,713; US 4,687,762; US 4,489,065;
WO2003/086363; US2004/0224024; US2007/0110804; US 6,063,370; EP 0987023; US
7,537,786; CN102558153; WO2013124340; EP2588090; WO2012027543; EP2631234;
WO2011110876; EP2288335; WO2012044595; WO2010020601; WO2013110567; EP2328581 and US20100322870.
DESCRIPTION OF THE INVENTION
Disclosed herein is a stable complex comprising as active compound chosen from Dabigatran, its salts or derivatives thereof; and at least one complexation agent chosen from polyvinylcaprolactam-polyvinyl acetate-polyethylene-glycol graft copolymers and poloxamers; polyvinylpyrrolidone; polyoxyglicerides; copolymers of vinylpyrrolidone and vinyl-acetate; and poly(maleic acid-co-methyl-vinyl-ether); said complex characterized in that it possesses at least one of the following properties: having a particle size in the range between 50 nm and 600 nm, preferably 50 nm and 300 nm;
is instantaneously redispersable in buffers in physiological pH range ;
is stable in solid form and in colloid solution and/or dispersion;
exhibits pH independent PAMPA permeability in the physiological pH range has a PAMPA permeability of at least 0.3* 10"6 cm/s when dispersed in FaSSIF or FeSSIF biorelevant media;
is characterized by infrared (ATR) spectrum having main/characteristic absorption peaks at 1730 cm"1, 1608 cm"1, 1468 cm"1, 1437 cm"1, 1384 cm"1, 1328 cm"1, 1252 cm"1, 1193 cm"1, 1144 cm"1, 1118 cm"1, 1098 cm"1, 1052 cm"1, 993 cm"1, 948 cm"1, 888 cm"1, 837 cm"1, 811 768 cm"1, 754 cm"1, 704 cm"1, 680 cm"1, 632 cm"1, 604 cm"1, 567 cm"1, 545 cm"1 and 524 cm"1, preferable at 1730 cm"1, 1608 cm"1, 1437 cm"1, 1328 cm"1,! 118 cm"1, 1098 cm"1, and 754 cm"1.
The invention is a complex formula having increased and pH independent permeability in the physiological relevant pH range and enhanced biological performance including higher Cmax and AUC, faster onset of action and decreased fed/fasted effect.
We have found that only selected combinations of complexation agents and pharmaceutically accepted excipients disclosed in the present invention result in a stable complex formula having improved physicochemical characteristics and enhanced biological performance.
The expression Dabigatran is generally used for Dabigatran, its esters, such as Dabigatran etexilate or its salts such as Dabigatran etexilate mesylate, Dabigatran etexilate methanesulfonate, Dabigatran etexilate bismesylate or derivatives of Dabigatran.
In an embodiment, said complexation agent is chosen from polyvinylcaprolactam-polyvinyl acetate-polyethylene-glycol graft copolymers and poloxamers; polyvinylpyrrolidone; polyoxyglicerides; copolymers of vinylpyrrolidone and vinyl-acetate; and poly(maleic acid- co-methyl -vinyl -ether) . In an embodiment, said complex further comprises at least one pharmaceutically acceptable excipient selected from the group of sodium-acetate, sodium-lauryl-sulfate, dioctyl sodium sulfosuccinate, cetylpyridinium chloride and sodium saccharine.
In an embodiment, said complex has a controlled particle size in the range between 50 nm and 600 nm. In an embodiment, said particle size is between 50 nm and 300 nm.
In an embodiment, said complex further comprises one or more additional active agents.
In an embodiment, said additional active agent is chosen from agents useful for the prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis. In an embodiment, said complex is orally available.
In an embodiment, said complex possesses at least two of the properties described in a) - f).
In an embodiment, said complex possesses at least three of the properties described in a) - f).
In an embodiment, said complex has an increased dissolution rate.
Further disclosed herein is a stable complex comprising an active compound selected from the group of Dabigatran, its salt, or derivatives thereof; at least one complexation agent chosen from polyvinylcaprolactam-polyvinyl acetate-polyethylene-glycol graft copolymers and poloxamers; polyvinylpyrrolidone; polyoxyglicerides; copolymers of vinylpyrrolidone and vinyl-acetate; and poly(maleic acid-co-methyl-vinyl-ether); and at least one pharmaceutically acceptable excipient chosen from sodium-acetate, sodium-lauryl-sulfate, dioctyl sodium sulfosuccinate, cetylpyridinium chloride and sodium saccharine; wherein said complex obtained via a mixing process.
In an embodiment, said complex is obtained via a continuous flow mixing process.
In an embodiment, a complex comprises a complexation agent and a pharmaceutically acceptable excipient in a total amount ranging from about 1.0 weight% to about 95.0 weight % based on the total weight of the complex.
In an embodiment, said complexation agent and pharmaceutically acceptable excipient comprise 50 weight% to about 95 weight% of the total weight of the complex. Further disclosed herein is a process for the preparation of the complex, comprising the steps of mixing a solution of Dabigatran, its salt, or derivatives thereof; and at least one complexation agent chosen from polyvinylcaprolactam-polyvinyl acetate-polyethylene-glycol graft copolymers and poloxamers; polyvinylpyrrolidone; polyoxyglicerides; copolymers of vinylpyrrolidone and vinyl-acetate; and poly(maleic acid-co-methyl-vinyl-ether) in a pharmaceutically acceptable solvent with an aqueous solution containing and at least one pharmaceutically acceptable excipient chosen from sodium-acetate, sodium-lauryl-sulfate, dioctyl sodium sulfosuccinate, cetylpyridinium chloride and sodium saccharine.
In an embodiment, said process is performed in a continuous flow instrument. In an embodiment, said continuous flow instrument is a microfluidic flow instrument.
In an embodiment, said pharmaceutically acceptable solvent is chosen from methanol, ethanol, i-propanol, n-propanol, acetone, acetonitrile, dimethyl-sulfoxyde, or tetrahydrofuran, or combinations thereof.
In an embodiment, said pharmaceutically acceptable solvent and said aqueous solvent are miscible with each other.
In an embodiment, said aqueous solvent comprises 0.1 to 99.9% weight of the final solution.
In an embodiment, said aqueous solvent comprises 50 to 90% weight of the final solution.
In an embodiment, said aqueous solvent comprises 50 to 80% weight of the final solution.
In an embodiment, said aqueous solvent comprises 50 to 70% weight of the final solution. In an embodiment, said aqueous solvent comprises 50 to 60% weight of the final solution.
In an embodiment, said aqueous solvent comprises 50 % weight of the final solution.
In an embodiment, a pharmaceutical composition comprising the complex together with pharmaceutically acceptable carrier.
In an embodiment, said composition is suitable for oral, pulmonary, rectal, colonic, parenteral, intracisternal, intravaginal, intraperitoneal, ocular, otic, local, buccal, nasal, or topical administration. In an embodiment, said composition is suitable for oral administration.
In an embodiment, said complex is for use in the manufacture of a medicament for the treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis.
In an embodiment, said complex is used for the treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis.
In an embodiment, a method of treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis comprises administration of a therapeutically effective amount of a complex or a pharmaceutical composition as described herein.
In an embodiment, a method for reducing the therapeutically effective dosage of Dabigatran compared to orally available formulations comprises oral administration of a pharmaceutical composition as described herein.
Further disclosed herein is a stable complex wherein said complex has a controlled particle size in the range between 50 nm and 600 nm; and wherein said complex is not obtained via a milling process or by high pressure homogenization process, encapsulation process and solid dispersion process, but it is obtained by a mixing process, preferable continuous flow mixing process.
In an embodiment, said particle size is between 50 nm and 300 nm.
In an embodiment, said complex shows reduced fed/fasted effect based on in vivo studies.
In an embodiment, said complex shows significantly improved exposure, earlier tmax, higher Cmax which will allow the oral administration and reduction of the dose.
In an embodiment, said complex has a faster onset of action compared to the marketed formulations. In an embodiment, said complex is instantaneously redispersable in physiological relevant media.
In an embodiment, said complex is stable in solid form and in colloid solution and/or dispersion. In an embodiment, said complex exhibits pH independent PAMPA permeability in the physiological pH range.
In an embodiment, said complex has a PAMPA permeability of at least 0.3* 10"6 cm/s when dispersed in FaSSIF or FeSSIF biorelevant media
In an embodiment, said complex is characterized by infrared (ATR) spectrum having charateristic absorption peaks at 1730 cm"1, 1608 cm"1, 1468 cm"1, 1437 cm"1, 1384 cm"1, 1328 cm"1, 1252 cm"1, 1193 cm"1, 1144 cm"1, 1118 cm"1, 1098 cm"1, 1052 cm"1, 993 cm"1, 948 cm"1, 888 cm"1, 837 cm"1, 811 768 cm"1, 754 cm"1, 704 cm"1, 680 cm"1, 632 cm"1, 604 cm"1, 567 cm"1, 545 cm"1 and 524 cm"1, preferable at 1730 cm"1, 1608 cm"1, 1437 cm"1, 1328 cm"1,! 118 cm"1, 1098 cm"1, and 754 cm"1. In an embodiment, said complex has decreased incidence of GI tract bleeding adverse side effect when compared to the marketed form.
The complexation agents and pharmaceutically acceptable excipients of the Dabigatran complex formulae of the invention are selected from the group of pharmaceutically acceptable nonionic, anionic, cationic, ionic polymers, surfactants and other types of excipients. The complexation agents themselves or together with the pharmaceutically accepted excipients have the function to form a complex structure with an active pharmaceutical ingredient through non-covalent secondary interactions. The secondary interactions can form through electrostatic interactions such as ionic interactions, H-bonding, dipole-dipole interactions, dipole-induced dipole interactions, London dispersion forces, π-π interactions, and hydrophobic interactions. The complexation agents, pharmaceutically accepted excipients and active ingredients are selected from the group of complexation agents, pharmaceutically accepted excipients and active ingredients which are able to form such complex structures through non-covalent secondary interactions. In some embodiments, the compositions may additionally include one or more pharmaceutically acceptable excipients, auxiliary materials, carriers, active agents or combinations thereof. In some embodiments, active agents may include agents useful for the treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis.
Another aspect of the invention is the complex formulae of the Dabigatran with complexation agents and pharmaceutically acceptable excipients in which the complexation agents and pharmaceutically acceptable excipients preferably are associated or interacted with the Dabigatran especially as the results of the mixing process, preferably continuous flow mixing process. In some embodiment, the structure of the complex Dabigatran formula is different from the core-shell type milled particle, precipitated encapsulated particles, micelles and solid dispersions.
The pharmaceutical composition of the invention can be formulated: (a) for administration selected from the group consisting of oral, pulmonary, rectal, colonic, parenteral, intracisternal, intravaginal, intraperitoneal, ocular, otic, local, buccal, nasal, and topical administration; (b) into a dosage form selected from the group consisting of liquid dispersions, gels, aerosols, ointments, creams, lyophilized formulations, tablets, capsules; (c) into a dosage form selected from the group consisting of controlled release formulations, fast melt formulations, delayed release formulations, extended release formulations, pulsatile release formulations, and mixed immediate release and controlled release formulations; or (d) any combination of (a), (b), and (c).
The compositions can be formulated by adding different types of excipients for oral administration in solid, liquid, local (powders, ointments or drops), or topical administration, and the like.
The compositions can be formulated by adding different types of pharmaceutically acceptable excipients for oral administration in solid, liquid, local (powders, ointments or drops), or topical administration, and the like.
A preferred dosage form of the invention is a solid dosage form, although any pharmaceutically acceptable dosage form can be utilized. Solid dosage forms for oral administration include, but are not limited to, capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active agent is admixed with at least one of the following excipients: (a) one or more inert excipients (or carriers), such as sodium citrate or dicalcium phosphate; (b) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, microcrystalline cellulose and silicic acid; (c) binders, such as cellulose derivatives, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (d) humectants, such as glycerol; (e) disintegrating agents, such as crospovidon, sodium starch glycolate, effervescent compositions, croscarmellose sodium,, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates and sodium carbonate; (f) solution retarders, such as acrylates, cellulose derivatives, paraffin; (g) absorption accelerators, such as quaternary ammonium compounds; (h) wetting agents, such as polysorbates, cetyl alcohol and glycerol monostearate; (i) adsorbents, such as kaolin and bentonite; and j) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. For capsules, tablets, and pills, the dosage forms may also comprise buffering agents.
Besides such inert diluents, the composition can also include adjuvants, such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Advantages of the complex Dabigatran formulae of the invention include, but are not limited to (1) physical and chemical stability, (2) instantaneous redispersibility, (3) stability in colloid solution or dispersion in the therapeutic time window, (4) increased solubility compared to the conventional Dabigatran formulation at physiologically relevant pH, (5) increased and pH independent permeability in the physiological relevant pH range, (6) possibility of dose reduction, (7) reduced GI tract bleeding adverse effect due to decreased anticoagulant dose and no strong acid necessary to be added to the formula and (8) good processability. Beneficial features of the present invention are as follows: the good/instantaneous redispersibility of solid complex formulae of Dabigatran in water, biologically relevant media, e.g.; physiological saline solution, pH=2.5 HC1 solution, FessiF and FassiF media and gastro intestinal fluids and adequate stability in colloid solutions and/or dispersion in the therapeutic time window. One of the preferred characteristics of the complex Dabigatran formulae of the present invention is their increased apparent solubility and pH independent increased permeability. In some embodiments, the permeability of the complex Dabigatran formulae is at least 0.574* 10" 6 cm/s, 0.376* 10"6 cm/s and 0.421 * 10"6 cm/s in the 5-8.5 pH range, respectively.
Finally, the invention relates to a method for the treatment of a subject in need for the treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesisby administering to the subject an effective amount of the complex or the pharmaceutical composition according to the invention.
Another preferred characteristic of the complex Dabigatran formulae of the present invention relates to the enhanced pharmacokinetic performance of the complex Dabigatran formulae. The complex Dabigatran is available for absorption in the whole GI tract extending the window for absorption from 0.5 hour to 4-5 hours and allows safer administration of a lower dose from the active ingredient.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1. shows complexation agent and pharmaceutically acceptable excipient screening for formula selection
Figure 2. shows particle size of the redispersed solid complex of the selected formula Figure 3. shows SEM photos of complex Dabigatran (A) and placebo sample (B)
Figure 4. shows infrared (ATR) spectra of Crystalline Dabigatran etexilate (A), amorphous Dabigatran etexilate (B), complex Dabigatran exeilate (C), placebo (D), Gelucire 44/14 (E), CPC (F) and Sacharine (G)
Figure 5. shows comparative PAMPA assays of complex Dabigatran formula and the marketed compound (Pradaxa)
Figure 6. shows pharmacokinetic properties of Dabigatran following the oral administration of Pradaxa (granules from the capsule) and complex Dabigatran Etexilate (dispersed in pH 8.5 Tris buffer) in rats in the fasted state at 3 mg/kg dose (n=4) EXAMPLES
1. Selection of complexation agents and pharmaceutically accepted stabilizers for the production of complex Dabigatran formulae
Several pharmaceutically accepted complexation agents and pharmaceutically accepted excipients and their combinations were tested in order to select the formulae having instantaneous redispersibility as shown in Figure 1. One of the examples that displayed an acceptable level of redispersibility was selected for further analysis.
2. Process for producing stable colloid solution and or dispersion of solid complex Dabigatran formula
A colloid solution of Dabigatran complex formula with the optimal ratio of the pharmaceutically acceptable excipients of the present invention was prepared by continuous flow mixing in a flow instrument. As a starting solution, 200 mg Dabigatran etexilate and 600 mg polyoxygliceride (Gelucire 44/14) dissolved in 100 mL tetrahydrofuran was used. The prepared solution was passed into the instrument with 2 mL/min flow rate. Meanwhile, aqueous solvent containing 50 mg cetylpyridinium chloride and 250 mg sodium saccharine in 500 mL water was passed into the instrument with 8 mL/min flow rate, where Dabigatran formed complex Dabigatran composition. The colloid solution of the complex Dabigatran is continuously produced at atmospheric pressure. The produced colloid solution was frozen on dry-ice and then it was lyophilized using a freeze drier equipped with -110°C ice condenser, with a vacuum pump. Particle size of complex Dabigatran was measured in reconstituted colloid solution right after the lyophilisation. It was found to be 156 nm as shown in Figure 2.
3. Structural analysis
Morphology of complex Dabigatran etexilate of Example 2 was investigated using FEI Quanta 3D scanning electron microscope. The morphology of the complex of the present invention was compared to the placebo sample prepared in the lack of Dabigatran etexilate. Complex Dabigatran of the present invention consists of spherical particles. In the lack of the active compound, the pharmaceutically acceptable excipients do not form spherical particles as shown in Figure 3. Structural analysis was performed by using Bruker Vertex 70 FT-IR spectrometer with Bruker Platinum diamond ATR unit. Continuous flow mixing of Dabigatran etexilate in the presence of selected complexation agents and pharmaceutically accepted excipients, such as polyoxygliceride (Gelucire 44/14), cetylpyridinium chloride and sodium saccharine resulted in a stable complex of Dabigatran as shown in Figure 4. In a preferred embodiment the complex or the pharmaceutical composition according to the invention characterized by at least one of the following characteristic infrared (ATR ) peak at 1730 cm"1, 1608 cm"1, 1468 cm"1, 1437 cm"1, 1384 cm"1, 1328 cm"1, 1252 cm"1, 1193 cm"1, 1144 cm"1, 1118 cm"1, 1098 cm" 1052 cm"1, 993 cm"1, 948 cm"1, 888 cm"1, 837 cm"1, 811 768 cm"1, 754 cm"1, 704 cm"1, 680 cm"1, 632 cm"1, 604 cm"1, 567 cm_1,_545 cm"1 and 524 cm"1, preferable at 1730 cm"1, 1608 cm" cm"1, 1328 cm"1, 1118 cm"1, 1098 cm"1, and 754 cm"1.
4. Comparative in vitro PAMPA assays
PAMPA permeability measurements were performed as described by M. Kansi et al. (Journal of medicinal chemistry, 41, (1998) pp 1007) with modifications based on S. Bendels et al (Pharmaceutical research, 23 (2006) pp 2525). Sample containing the reference compound was a suspension of crystals visible by the naked eye, while samples of the novel complex were opalescent colloid solutions. Permeability was measured in a 96-well plate assay across an artificial membrane composed of dodecane with 20% soy lecithin supported by a PVDF membrane (Millipore, USA). The receiver compartment was phosphate buffered saline (pH 7.0) supplemented with 1% sodium dodecyl sulfate. The assay was performed at room temperature; incubation time was 1-24 hours. The concentration in the receiver compartment was determined by UV-VIS spectrophotometry (Thermo Scientific Genesys S10).
PAMPA permeability of complex Dabigatran formula of Example 2 was 0.574* 10"6 cm/s, 0.376* 10"6 cm/s and 0.421 * 10"6 cm/s when dispersed in a buffer pH 5.0, 6.5 or 8.5, respectively, while permeability of the marketed compound (Pradaxa) was 0.094* 10"6 cm/s, 0.060* 10"6 cm/s and 0.020* 10"6 cm/s when dispersed in the same buffers as shown in Figure 5.
5. Comparative in vivo study The main objective of this study was the pharmacokinetic evaluation of a Dabigatran etexilate complex formulation following a single oral administration at 3 mg/kg dose to jugular vein cannulated male Wistar rats. The mean metabolite, Dabigatran was measured in plasma using a reliable LC/MS6MS method.
The test item substance was formulated in 0.1M Tris buffer (pH=8.5). The test item was administered to the rats orally by gavage. The granules of the reference item were administered through a plastic gavage into the stomach of the rats. The granules were pushed out with the help of a mandrin then water was administered in a same amount as the test item administration amount.
Blood was collected in the following time points using sodium heparin as anticoagulant: 15m, 30m, 45m, lh, 1.5h, 2h, 4h, 6h, and 8h. The plasma was separated and stored in an ultra- freezer (below -70°C) until analysis.
Dabigatran plasma kinetics showed high inter-individual variability after dosing the reference item. After dosing of the reference item the shape of the individual curves and the tmax values (0.25h - 4h) showed very high variability and the Cmax values varied in a range (80.6 ± 37.0).
For the test item peak concentrations of Dabigatran occurred at 0.25h - 0.5h after administration in all the four rats. The average Cmax value was 210 ± 154 ng/ml. The elimination of Dabigatran from the plasma was relatively fast: the mean half-life was 1.60 ± 0.291 h. The AUCinf values were 470 ± 243 ng*h/ml. This total exposure of the complex formulation was compared to the AUCinf value obtained for a reference formulation (Pradaxa) after administration of the same oral dose of Dabigatran etexilate. The AUCinf of Pradaxa was 280 ± 93.7 ng*h/ml. The relative oral bioavailability (Frel) of the investigated nano- formulation proved to be 168% (Figure 6).

Claims

Claims
1. A stable complex comprising as active compound selected from the group of Dabigatran, its esters, preferably Dabigatran etexilate, its salts, preferably Dabigatran etexilate mesylate, or derivatives thereof; and at least one complexation agent selected from the group of a polyvinylcaprolactam-polyvinyl acetate-polyethylene-glycol graft copolymers and poloxamers; polyvinylpyrrolidone; polyoxyglicerides; copolymers of vinylpyrrolidone and vinyl-acetate; and poly(maleic acid-co-methyl-vinyl-ether); said complex characterized in that it possesses at least one of the following properties: a) having a particle size in the range between 50 nm and 600 nm, preferably 50 nm and 300 nm;
b) is instantaneously redispersable in buffers in physiological pH range ;
c) is stable in solid form and in colloid solution and/or dispersion;
d) exhibits pH independent PAMPA permeability in the physiological pH range e) has a PAMPA permeability of at least 0.3* 10"6 cm/s when dispersed in FaSSIF or FeSSIF biorelevant media;
f) is characterized by infrared (ATR) spectrum having main/characteristic absorption peaks at 1730 cm"1, 1608 cm"1, 1437 cm"1, 1328 cm"1,! 118 cm"1, 1098 cm"1, and 754 cm"1;
g) has reduced fed/fasted effect based on in vivo studies.
h) has significantly improved exposure, earlier tmax, higher Cmax which will allow the oral administration and reduction of the dose.
i) has a faster onset of action compared to the marketed formulations.
2. The complex according to Claim 1, wherein said complexation agent is selected from the group of polyvinylcaprolactam-polyvinyl acetate-polyethylene-glycol graft copolymers and poloxamers; polyvinylpyrrolidone; polyoxyglicerides; copolymers of vinylpyrrolidone and vinyl-acetate; and poly(maleic acid-co-methyl-vinyl-ether).
3. A complex according to Claims 1 or 2, wherein said complex further comprises at least one pharmaceutically accepted excipient selected from the group of sodium-lauryl-sulfate; dioctyl sodium sulfosuccinate; cetylpyridinium chloride, sodium-acetate and sodium saccharine, preferably cetylpyridinium chloride and sodium saccharine.
The complex according to any of Claims 1 to 3, wherein said complex further comprises one or more additional active agents, preferable the additional active agent is selected from the group of agents useful for the treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis.
The complex according to Claim 1, wherein said complex is orally bioavailable.
A stable complex according to any Claims 1 to 5comprising an active compound selected from the group of Dabigatran, its esters, preferably Dabigatran etexilate, its salts, preferably Dabigatran etexilate mesylate, or derivatives thereof; at least one complexation agent selected from the group of a polyvinylcaprolactam-polyvinyl acetate-polyethylene- glycol graft copolymers and poloxamers; polyvinylpyrrolidone; polyoxyglicerides; copolymers of vinylpyrrolidone and vinyl-acetate; and poly(maleic acid-co-methyl-vinyl- ether), and at least one pharmaceutically accepted excipient chosen from the group of sodium-lauryl-sulfate; dioctyl sodium sulfosuccinate; cetylpyridinium chloride, sodium- acetate and sodium saccharine; wherein said complex obtained via a mixing process, preferable continuous flow mixing process, more preferable microfluidic flow mixing process.
A complex according to Claims 1 to 6 comprising complexation agents selected from the group of polyvinylcaprolactam-polyvinyl acetate-polyethylene-glycol graft copolymers and/or poloxamers and/or polyvinylpyrrolidone and/or copolymers of vinylpyrrolidone and vinyl-acetate and/or polyoxyglicerides and pharmaceutically accepted excipients selected from the group of sodium-acetate and/or sodium-lauryl-sulfate and/or dioctyl sodium sulfosuccinate and/or cetylpyridinium chloride and/or sodium saccharine in an amount ranging from about 1.0 weight% to about 90.0 weight % based on the total weight of the complex.
A process for the preparation of the complex according to Claims 1 to 6, comprising the steps of mixing a solution of Dabigatran, its esters, preferably Dabigatran etexilate, its salts, preferably Dabigatran etexilate mesylate, or derivatives thereof, and at least one complexation agent selected from the group of polyvinylcaprolactam-polyvinyl acetate- polyethylene-glycol graft copolymers and/or poloxamers and/or polyvinylpyrrolidone and/or copolymers of vinylpyrrolidone and vinyl-acetate and/or polyoxyglicerides in a pharmaceutically acceptable solvent with an aqueous solution containing at least one pharmaceutically accepted excipient chosen from sodium-acetate, sodium-lauryl-sulfate, dioctyl sodium sulfosuccinate, cetylpyridinium chloride and sodium saccharine..
9. The process according to Claim 7, wherein said process is performed in a continuous flow instrument, preferable in microfluidic instrument.
10. The process according to Claims 7 or 8, wherein said pharmaceutically acceptable solvent is selected from the group of methanol, ethanol, isopropanol, n-propanol, acetone, acetonitrile, dimethyl-sulfoxide, tetrahydrofuran, and combinations thereof.
11. The process according to any of Claims 7 to 9, wherein the solvent and the aqueous solvent are miscible with each other and the aqueous solvent comprises 0.1 to 99.9% weight of the final solution.
12. A pharmaceutical composition comprising the complex according to any of Claims 1 to 6 together with pharmaceutically acceptable carrier.
13. A pharmaceutical composition according to Claim 11, wherein said composition is suitable for oral, pulmonary, rectal, colonic, parenteral, intracisternal, intravaginal, intraperitoneal, ocular, otic, local, buccal, nasal, or topical administration, preferable the composition is suitable for oral administration.
14. A complex according to any of Claims 1 or to 6 for use in the manufacture of a medicament for the treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis.
15. The use of the complex according to any of Claims 1 or to 6 for the treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis.
16. A method of treatment and/or prophylaxis of diseases, in particular thrombotic diseases, stroke, cardiac infarction and/or atrial fibrillation and cardiac arrhythmia, as well as oncological diseases of any pathogenesis comprising administration of a therapeutically effective amount of the complex according to any of Claims 1 to 6 or the pharmaceutical composition according to Claim 12 orl3.
17. A stable complex wherein said complex has a controlled particle size in the range between 50 nm and 600 nm, preferable the particle size is between 50 nm and 300 nm; and wherein said complex is obtained according to any of claims 7 to 11.
PCT/IB2014/065992 2013-11-12 2014-11-12 Complexes of dabigatran and its derivatives, process for the preparation thereof and pharmaceutical compositions containing them WO2015071841A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
HUP1300648 2013-11-12
HU1300648A HUP1300648A2 (en) 2013-11-12 2013-11-12 Complexes of dabigatran and its derivatives, process for the preparation thereof and pharmaceutical compositions containing them
HUP1400084 2014-02-17
HU1400084A HUP1400084A2 (en) 2014-02-17 2014-02-17 Complexes of dabigatran and its derivatives, process for the preparation thereof and pharmaceutical composition containing them

Publications (1)

Publication Number Publication Date
WO2015071841A1 true WO2015071841A1 (en) 2015-05-21

Family

ID=89991406

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2014/065992 WO2015071841A1 (en) 2013-11-12 2014-11-12 Complexes of dabigatran and its derivatives, process for the preparation thereof and pharmaceutical compositions containing them

Country Status (1)

Country Link
WO (1) WO2015071841A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105534939A (en) * 2015-12-29 2016-05-04 郑州大明药物科技有限公司 Dabigatran etexilate mesylate oral solid preparation and preparation method thereof
WO2017187336A1 (en) * 2016-04-25 2017-11-02 Druggability Technologies Ip Holdco Limited Complexes of ivacaftor and its salts and derivatives, process for the preparation thereof and pharmaceutical compositions containing them
WO2017187340A1 (en) * 2016-04-25 2017-11-02 Druggability Technologies Ip Holdco Limited Pharmaceutical combination composition comprising complex formulations of ivacaftor and lumacaftor and their salts and derivatives, process for their preparation thereof and pharmaceutical compositions containing them
WO2017187338A1 (en) * 2016-04-25 2017-11-02 Druggability Technologies Ip Holdco Limited Complexes of lumacaftor and its salts and derivatives, process for the preparation thereof and pharmaceutical compositions containing them
US10376501B2 (en) 2016-04-25 2019-08-13 Druggability Technologies Ip Holdco Limited Complexes of lumacaftor and its salts and derivatives, process for the preparation thereof and pharmaceutical compositions containing them
US10383865B2 (en) 2016-04-25 2019-08-20 Druggability Technologies Ip Holdco Limited Pharmaceutical combination composition comprising complex formulations of Ivacaftor and Lumacaftor and their salts and derivatives, process for their preparation thereof and pharmaceutical compositions containing them
CN111163807A (en) * 2017-09-19 2020-05-15 耶路撒冷希伯来大学伊森姆研究发展有限公司 Lipophilic peptide prodrugs
CN111902161A (en) * 2018-04-04 2020-11-06 上海汉都医药科技有限公司 Pharmaceutical composition containing dabigatran etexilate and preparation method thereof
CN113893356A (en) * 2020-11-27 2022-01-07 上海博志研新药物技术有限公司 Dabigatran etexilate mesylate inclusion compound, preparation method and application
US11965041B2 (en) 2017-09-19 2024-04-23 Technische Universitaet Muenchen N-methylated cyclic peptides and their prodrugs

Citations (106)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4489065A (en) 1981-07-02 1984-12-18 Valcor Scientific Ltd. Chondroitin drug Complexes
US4540602A (en) 1979-04-13 1985-09-10 Freund Industry Company, Limited Process for the preparation of activated pharmaceutical compositions
US4687762A (en) 1984-03-31 1987-08-18 Green Cross Corporation Water soluble drug complex and method for production of same
WO1989008127A1 (en) * 1988-02-29 1989-09-08 Gaf Chemicals Corporation Solubilization of complexes of water-insoluble organic compounds by aqueous solutions of polyvinylpyrrolidone
US4894235A (en) 1984-10-23 1990-01-16 Dr. Rentschler, Arzneimmittel Gmbh & Co. Nifedipine-containing form of administration and method for its production
US4996047A (en) 1988-11-02 1991-02-26 Richardson-Vicks, Inc. Sustained release drug-resin complexes
US5091187A (en) 1990-04-26 1992-02-25 Haynes Duncan H Phospholipid-coated microcrystals: injectable formulations of water-insoluble drugs
US5133908A (en) 1986-12-31 1992-07-28 Centre National De La Recherche Scientifique (Cnrs) Process for the preparation of dispersible colloidal systems of a substance in the form of nanoparticles
US5141961A (en) 1991-06-27 1992-08-25 Richrdson-Vicks Inc. Process for solubilizing difficulty soluble pharmaceutical actives
US5145684A (en) 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
US5324718A (en) 1992-07-14 1994-06-28 Thorsteinn Loftsson Cyclodextrin/drug complexation
US5340591A (en) 1992-01-24 1994-08-23 Fujisawa Pharmaceutical Co., Ltd. Method of producing a solid dispersion of the sparingly water-soluble drug, nilvadipine
US5389382A (en) 1986-12-19 1995-02-14 Sandoz Ltd. Hydrosols of pharmacologically active agents and their pharmaceutical compositions comprising them
US5518187A (en) 1992-11-25 1996-05-21 Nano Systems L.L.C. Method of grinding pharmaceutical substances
US5560932A (en) 1995-01-10 1996-10-01 Nano Systems L.L.C. Microprecipitation of nanoparticulate pharmaceutical agents
US5646131A (en) 1994-02-22 1997-07-08 The Arab Company For Drug Industries And Medical Applicances (Acdima) Method for solubilizing drugs using cyclodextrins and carboxylic acids
US5716642A (en) 1995-01-10 1998-02-10 Nano Systems L.L.C. Microprecipitation of nanoparticulate pharmaceutical agents using surface active material derived from similar pharmaceutical agents
US5718388A (en) 1994-05-25 1998-02-17 Eastman Kodak Continuous method of grinding pharmaceutical substances
US5780062A (en) 1994-11-09 1998-07-14 The Ohio State University Research Foundation Small particle formation
US5862999A (en) 1994-05-25 1999-01-26 Nano Systems L.L.C. Method of grinding pharmaceutical substances
EP0911039A2 (en) 1997-04-16 1999-04-28 Medeva Pharmaceutical Manufacturing, Inc. Drug-resin complexes stabilized by chelating agents
US6018080A (en) 1992-12-24 2000-01-25 Glaxo Wellcome Inc. Atovaquone pharmaceutical compositions
EP0987023A1 (en) 1997-04-17 2000-03-22 Panacea Biotec Limited Transition metal complexes of non steroidal anti-inflammatory drugs
WO2000016744A1 (en) 1998-09-23 2000-03-30 Guju Pharm. Co., Ltd. A pharmaceutical solution for the treatment of erectile dysfunction, prepared by sedds formulation
US6063370A (en) 1996-04-05 2000-05-16 The Board Of Trustees Of The University Of Illinois Macromolecular complexes for drug delivery
WO2000032313A1 (en) 1998-12-01 2000-06-08 Aventis Pharma Limited Milling process for the production of finely milled medicinal substances
WO2000040220A1 (en) 1999-01-06 2000-07-13 Korea Research Institute Of Chemical Technology Method of preparing pharmaceutical active ingredient comprising water-insoluble drug and pharmaceutical composition for oral administration comprising the same
EP1027886A2 (en) 1999-02-10 2000-08-16 Pfizer Products Inc. Pharmaceutical solid dispersions
WO2001003670A1 (en) 1999-07-13 2001-01-18 Pharmasol Gmbh Method for controlled production of ultrafine microparticles and nanoparticles
US6291671B1 (en) 1996-06-06 2001-09-18 Daiichi Pharmaceutical Co., Ltd. Process for producing drug complexes
WO2001085345A1 (en) 2000-05-10 2001-11-15 Rtp Pharma Inc. Media milling
WO2002000196A2 (en) 2000-06-28 2002-01-03 Smithkline Beecham P.L.C. Wet milling process
US6406713B1 (en) 1987-03-05 2002-06-18 The Liposome Company, Inc. Methods of preparing low-toxicity drug-lipid complexes
US6431478B1 (en) 1999-06-01 2002-08-13 Elan Pharma International Limited Small-scale mill and method thereof
US6436912B1 (en) 1996-06-06 2002-08-20 Daiichi Pharmaceutical Co., Ltd. Drug complexes
US20020127278A1 (en) 2000-12-22 2002-09-12 Kipp James E. Microprecipitation method for preparing submicron suspensions
US6458347B1 (en) 1996-04-15 2002-10-01 Asahi Kasei Kabushiki Kaisha Drug complex
WO2003002258A1 (en) 2001-06-29 2003-01-09 Eurand Pharmaceuticals Ltd Drug activation process and vibrational mill therefor
US20030104065A1 (en) 2000-03-09 2003-06-05 Arne Brodin Method of preparing solid dispersions
US6582285B2 (en) 2000-04-26 2003-06-24 Elan Pharmainternational Ltd Apparatus for sanitary wet milling
US6596263B1 (en) 1995-07-14 2003-07-22 Stc English Ideas, Inc. Cyclodextrin compositions
EP1348432A1 (en) 2001-01-04 2003-10-01 Daiichi Pharmaceutical Co., Ltd. Cyclodextrin-containing pharmaceutical preparation
WO2003086363A1 (en) 2002-04-08 2003-10-23 Lavipharm Laboratories Inc. Drug-complex microparticles and methods of making/using same
US6641063B2 (en) 1998-12-01 2003-11-04 Aventis Pharma Limited Milling process for the production of finely milled medicinal substances
US6677362B1 (en) 1991-12-18 2004-01-13 Warner-Lambert Company Solid pharmaceutical dispersions
US6699849B1 (en) 1998-02-23 2004-03-02 Cyclops, Ehf. Cyclodextrin complexes of benzodiazepines
US6706283B1 (en) 1999-02-10 2004-03-16 Pfizer Inc Controlled release by extrusion of solid amorphous dispersions of drugs
US6742734B2 (en) 2001-06-05 2004-06-01 Elan Pharma International Limited System and method for milling materials
US6814319B2 (en) 2000-12-06 2004-11-09 Pharmacia & Upjohn Company Laboratory scale milling process
US20040224024A1 (en) 2003-04-23 2004-11-11 Massachusetts Institute Of Technology Controlled drug release formulations containing polyion complexes
US20050020294A1 (en) 2001-12-07 2005-01-27 Marko Kantola System for transmitting information in a digital radio network system
WO2005037250A1 (en) 2003-10-17 2005-04-28 Amylin Pharmaceuticals, Inc. Self emulsifying drug delivery systems for hydrophobic therapeutic compounds
US6921458B2 (en) 2002-12-19 2005-07-26 Acusphere, Inc. Methods and apparatus for making particles using spray dryer and in-line jet mill
US20060013888A1 (en) 2003-09-29 2006-01-19 Ronald G. Udell Solubilized CoQ-10
WO2006021160A1 (en) 2004-08-27 2006-03-02 Shijiazhuang Pharma. Group Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., Ltd. Butylbenzene phthalein self-emulsifying drug delivery system, its preparation method and application
WO2006060698A1 (en) 2004-12-03 2006-06-08 Elan Pharma International Ltd. Nanoparticulate benzothiophene formulations
US20060159766A1 (en) 2004-12-15 2006-07-20 Elan Pharma International Limited Nanoparticulate tacrolimus formulations
US20060165806A1 (en) 2005-01-06 2006-07-27 Elan Pharma International Limited Nanoparticulate candesartan formulations
US20060188566A1 (en) 2005-02-24 2006-08-24 Elan Pharma International Limited Nanoparticulate formulations of docetaxel and analogues thereof
WO2006096462A1 (en) 2005-03-03 2006-09-14 Elan Pharma International Limited Nanoparticulate compositions of heterocyclic amide derivatives
WO2006100281A2 (en) 2005-03-22 2006-09-28 Losan Pharma Gmbh Solubilized ibuprofen
WO2006110811A1 (en) 2005-04-12 2006-10-19 Elan Pharma International Limited Nanoparticulate quinazoline derivative formulations
WO2006112541A1 (en) 2005-04-20 2006-10-26 Chugai Seiyaku Kabushiki Kaisya Method for the design of self-emulsifying drug formulations
US20070110804A1 (en) 2003-06-20 2007-05-17 Royer Biomedical, Inc. Drug polymer complexes
US7244451B2 (en) 2000-09-21 2007-07-17 Elan Pharma International Ltd. Methods of making nanoparticulate drug compositions comprising copolymers of vinyl pyrrolidone and vinyl acetate as surface stabilizers
WO2007150030A2 (en) 2006-06-23 2007-12-27 Massachusetts Institute Of Technology Microfluidic synthesis of organic nanoparticles
WO2008002121A2 (en) 2006-06-28 2008-01-03 Hovid, Berhad A self-emulsifying composition for poorly bioavailable drugs
US7441717B1 (en) 2007-10-31 2008-10-28 Eastman Kodak Company Micromedia milling process
WO2009007992A2 (en) 2007-04-20 2009-01-15 Sun Pharmaceutical Industries Limited Pharmaceutical composition produced by microprecipitation
US7537786B2 (en) 2005-02-25 2009-05-26 Nec Corporation Complex of drug-carbon nanohorn and a process for producing the complex
WO2009075401A1 (en) 2007-12-12 2009-06-18 Chung-Ang University Industry-Academy Cooperation Foundation Manufacturing method of functional drug nanoparticles using milling and functional drug nanoparticle formulation manufactured thereby
WO2009118322A1 (en) * 2008-03-28 2009-10-01 Boehringer Ingelheim International Gmbh Process for preparing orally administered dabigatran formulations
WO2009133418A1 (en) 2008-04-28 2009-11-05 Nangenex Nanotechnology Incorporated Instrument and process for nanoparticles production in continuous flow mode
WO2010020601A1 (en) 2008-08-19 2010-02-25 Boehringer Ingelheim International Gmbh Dabigatran in tumour therapy
WO2010133611A1 (en) 2009-05-18 2010-11-25 Royal College Of Surgeons In Ireland Solid drug dispersions
WO2010146406A1 (en) 2009-06-19 2010-12-23 Nanoform Hungary Ltd. Nanoparticulate telmisartan compositions and process for the preparation thereof
US20100322870A1 (en) 2008-11-11 2010-12-23 Boehringer Ingelheim International Gmbh Method for treating or preventing thrombosis using dabigatran etexilate or a salt thereof with improved efficacy over conventional warfarin therapy
WO2010146408A2 (en) 2009-06-19 2010-12-23 Nangenex, Inc. Nanoparticulate olmesartan medoxomil compositions, process for the preparation thereof and pharmaceutical compositions containing them
WO2010146407A1 (en) 2009-06-19 2010-12-23 Nanoform Hungary Ltd. Nanostructured sildenafil base, its pharmaceutically acceptable salts and co-crystals, compositions of them, process for the preparation thereof and pharmaceutical compositions containing them
WO2010146409A2 (en) 2009-06-19 2010-12-23 Nangenex, Inc. Nanoparticulate candesartan cilexetil compositions, process for the preparation thereof and pharmaceutical compositions containing them
US7951399B2 (en) 2001-06-29 2011-05-31 Eurand Pharmaceuticals Ltd. Process for activation of drugs in a vibrational mill
EP2328581A1 (en) 2008-08-19 2011-06-08 Boehringer Ingelheim International GmbH Use of dabigatranetexilate for treating patients with pulmonary hypertension
US7994214B2 (en) 2003-08-29 2011-08-09 Lifecycle Pharma A/S Solid dispersions comprising tacrolimus
WO2011110876A1 (en) 2010-02-02 2011-09-15 Egis Gyógyszergyár Nyilvánosan Működő Részvénytársaság Novel salts for the manufacture of pharmaceutical compositions
WO2011110478A1 (en) * 2010-03-08 2011-09-15 Ratiopharm Gmbh Dabigatran etexilate-containing pharmaceutical composition
WO2011131943A2 (en) 2010-04-20 2011-10-27 Cipla Limited Pharmaceutical compositions
US8074906B2 (en) 2005-07-07 2011-12-13 Nanotherapeutics, Inc. Process for milling and preparing powders and compositions produced thereby
WO2011154755A1 (en) 2010-06-08 2011-12-15 Nanoform Cardiovascular Therapeutics Ltd. Nanostructured atorvastatin, its pharmaceutically acceptable salts and compositions of them, process for the preparation thereof and pharmaceutical compositions containing them
WO2011158053A1 (en) 2010-06-18 2011-12-22 Nanoform Cardiovascular Therapeutics Ltd. Nanostructured aprepitant compositions, process for the preparation thereof and pharmaceutical compositions containing them
WO2011158052A1 (en) 2010-06-18 2011-12-22 Nanoform Cardiovascular Therapeutics Ltd. Nanostructured ezetimibe compositions, process for the preparation thereof and pharmaceutical compositions containing them
US8092785B2 (en) 2004-08-23 2012-01-10 Glaxo Group Limited Process for milling poorly soluble drugs in presence of liquid propellants
WO2012027543A1 (en) 2010-08-25 2012-03-01 Teva Pharmaceuticals Usa, Inc. Solid state forms of dabigatran etexilate, dabigatran etexilate mesylate and processes for preparation thereof
WO2012042224A2 (en) 2010-10-01 2012-04-05 Cipla Limited Pharmaceutical composition
WO2012044595A1 (en) 2010-09-27 2012-04-05 Ratiopharm Gmbh Dabigatran etexilate bismesylate salt, solid state forms and process for preparation thereof
US8158162B2 (en) 2001-11-14 2012-04-17 Jarrow Formulas, Inc. Eutectic-based self-nanoemulsified drug delivery system
CN102558153A (en) 2012-02-08 2012-07-11 北京阜康仁生物制药科技有限公司 Novel pharmaceutical salt of dabigatran etexilate and preparation method thereof
WO2012160559A1 (en) 2011-05-22 2012-11-29 Rappaport Family Institute For Research In The Medical Sciences Pharmaceutical compositions of d-alpha-tocopheryl acetate
US8337899B2 (en) 1997-08-11 2012-12-25 Bend Research, Inc. Solid pharmaceutical dispersions with enhanced bioavailability
US8343548B2 (en) 2006-08-08 2013-01-01 Shin-Etsu Chemical Co., Ltd. Solid dosage form comprising solid dispersion
US8354122B2 (en) 2006-04-20 2013-01-15 Shin-Etsu Chemical Co., Ltd. Solid dispersion preparation
EP2588090A2 (en) 2010-07-01 2013-05-08 Krka Tovarna Zdravil, D.D., Novo Mesto Pharmaceutical oral dosage forms comprising dabigatran etexilate and its pharmaceutically acceptable salts
WO2013110567A1 (en) 2012-01-24 2013-08-01 Boehringer Ingelheim International Gmbh Novel orally administered dabigatran formulation
EP2631234A1 (en) 2012-02-23 2013-08-28 Esteve Química, S.A. Solid forms of dabigatran etexilate mesylate and processes for their preparation
WO2013124340A1 (en) 2012-02-21 2013-08-29 Laboratorios Del Dr. Esteve, S.A. Oral pharmaceutical compositions of dabigatran etexilate
WO2013175494A2 (en) * 2012-04-10 2013-11-28 Rubicon Research Private Limited Controlled release pharmaceutical formulations of direct thrombin inhibitors
WO2014131376A1 (en) * 2013-02-26 2014-09-04 Elmarco S.R.O. Electrospun nanofibers comprising pharmaceutically active agents

Patent Citations (112)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4540602A (en) 1979-04-13 1985-09-10 Freund Industry Company, Limited Process for the preparation of activated pharmaceutical compositions
US4489065A (en) 1981-07-02 1984-12-18 Valcor Scientific Ltd. Chondroitin drug Complexes
US4687762A (en) 1984-03-31 1987-08-18 Green Cross Corporation Water soluble drug complex and method for production of same
US4894235A (en) 1984-10-23 1990-01-16 Dr. Rentschler, Arzneimmittel Gmbh & Co. Nifedipine-containing form of administration and method for its production
US5389382A (en) 1986-12-19 1995-02-14 Sandoz Ltd. Hydrosols of pharmacologically active agents and their pharmaceutical compositions comprising them
US5133908A (en) 1986-12-31 1992-07-28 Centre National De La Recherche Scientifique (Cnrs) Process for the preparation of dispersible colloidal systems of a substance in the form of nanoparticles
US6406713B1 (en) 1987-03-05 2002-06-18 The Liposome Company, Inc. Methods of preparing low-toxicity drug-lipid complexes
WO1989008127A1 (en) * 1988-02-29 1989-09-08 Gaf Chemicals Corporation Solubilization of complexes of water-insoluble organic compounds by aqueous solutions of polyvinylpyrrolidone
US4996047A (en) 1988-11-02 1991-02-26 Richardson-Vicks, Inc. Sustained release drug-resin complexes
US5091187A (en) 1990-04-26 1992-02-25 Haynes Duncan H Phospholipid-coated microcrystals: injectable formulations of water-insoluble drugs
US5145684A (en) 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
US5141961A (en) 1991-06-27 1992-08-25 Richrdson-Vicks Inc. Process for solubilizing difficulty soluble pharmaceutical actives
US6677362B1 (en) 1991-12-18 2004-01-13 Warner-Lambert Company Solid pharmaceutical dispersions
US5340591A (en) 1992-01-24 1994-08-23 Fujisawa Pharmaceutical Co., Ltd. Method of producing a solid dispersion of the sparingly water-soluble drug, nilvadipine
US5324718A (en) 1992-07-14 1994-06-28 Thorsteinn Loftsson Cyclodextrin/drug complexation
US5518187A (en) 1992-11-25 1996-05-21 Nano Systems L.L.C. Method of grinding pharmaceutical substances
US6018080A (en) 1992-12-24 2000-01-25 Glaxo Wellcome Inc. Atovaquone pharmaceutical compositions
US5646131A (en) 1994-02-22 1997-07-08 The Arab Company For Drug Industries And Medical Applicances (Acdima) Method for solubilizing drugs using cyclodextrins and carboxylic acids
US5718388A (en) 1994-05-25 1998-02-17 Eastman Kodak Continuous method of grinding pharmaceutical substances
US5862999A (en) 1994-05-25 1999-01-26 Nano Systems L.L.C. Method of grinding pharmaceutical substances
US5780062A (en) 1994-11-09 1998-07-14 The Ohio State University Research Foundation Small particle formation
US5716642A (en) 1995-01-10 1998-02-10 Nano Systems L.L.C. Microprecipitation of nanoparticulate pharmaceutical agents using surface active material derived from similar pharmaceutical agents
US5560932A (en) 1995-01-10 1996-10-01 Nano Systems L.L.C. Microprecipitation of nanoparticulate pharmaceutical agents
US6596263B1 (en) 1995-07-14 2003-07-22 Stc English Ideas, Inc. Cyclodextrin compositions
US6063370A (en) 1996-04-05 2000-05-16 The Board Of Trustees Of The University Of Illinois Macromolecular complexes for drug delivery
US6458347B1 (en) 1996-04-15 2002-10-01 Asahi Kasei Kabushiki Kaisha Drug complex
US6291671B1 (en) 1996-06-06 2001-09-18 Daiichi Pharmaceutical Co., Ltd. Process for producing drug complexes
US6436912B1 (en) 1996-06-06 2002-08-20 Daiichi Pharmaceutical Co., Ltd. Drug complexes
EP0911039A2 (en) 1997-04-16 1999-04-28 Medeva Pharmaceutical Manufacturing, Inc. Drug-resin complexes stabilized by chelating agents
US5980882A (en) 1997-04-16 1999-11-09 Medeva Pharmaceuticals Manufacturing Drug-resin complexes stabilized by chelating agents
EP0987023A1 (en) 1997-04-17 2000-03-22 Panacea Biotec Limited Transition metal complexes of non steroidal anti-inflammatory drugs
US8337899B2 (en) 1997-08-11 2012-12-25 Bend Research, Inc. Solid pharmaceutical dispersions with enhanced bioavailability
US6699849B1 (en) 1998-02-23 2004-03-02 Cyclops, Ehf. Cyclodextrin complexes of benzodiazepines
WO2000016744A1 (en) 1998-09-23 2000-03-30 Guju Pharm. Co., Ltd. A pharmaceutical solution for the treatment of erectile dysfunction, prepared by sedds formulation
WO2000032313A1 (en) 1998-12-01 2000-06-08 Aventis Pharma Limited Milling process for the production of finely milled medicinal substances
US6641063B2 (en) 1998-12-01 2003-11-04 Aventis Pharma Limited Milling process for the production of finely milled medicinal substances
WO2000040220A1 (en) 1999-01-06 2000-07-13 Korea Research Institute Of Chemical Technology Method of preparing pharmaceutical active ingredient comprising water-insoluble drug and pharmaceutical composition for oral administration comprising the same
US6706283B1 (en) 1999-02-10 2004-03-16 Pfizer Inc Controlled release by extrusion of solid amorphous dispersions of drugs
US7550158B2 (en) 1999-02-10 2009-06-23 Bend Research, Inc. Controlled release by extrusion of solid amorphous dispersions of drugs
EP1027886A2 (en) 1999-02-10 2000-08-16 Pfizer Products Inc. Pharmaceutical solid dispersions
US6431478B1 (en) 1999-06-01 2002-08-13 Elan Pharma International Limited Small-scale mill and method thereof
WO2001003670A1 (en) 1999-07-13 2001-01-18 Pharmasol Gmbh Method for controlled production of ultrafine microparticles and nanoparticles
US20030104065A1 (en) 2000-03-09 2003-06-05 Arne Brodin Method of preparing solid dispersions
US6582285B2 (en) 2000-04-26 2003-06-24 Elan Pharmainternational Ltd Apparatus for sanitary wet milling
WO2001085345A1 (en) 2000-05-10 2001-11-15 Rtp Pharma Inc. Media milling
WO2002000196A2 (en) 2000-06-28 2002-01-03 Smithkline Beecham P.L.C. Wet milling process
US7244451B2 (en) 2000-09-21 2007-07-17 Elan Pharma International Ltd. Methods of making nanoparticulate drug compositions comprising copolymers of vinyl pyrrolidone and vinyl acetate as surface stabilizers
US6814319B2 (en) 2000-12-06 2004-11-09 Pharmacia & Upjohn Company Laboratory scale milling process
US20020168402A1 (en) 2000-12-22 2002-11-14 Kipp James E. Microprecipitation method for preparing submicron suspensions
US20020127278A1 (en) 2000-12-22 2002-09-12 Kipp James E. Microprecipitation method for preparing submicron suspensions
US6607784B2 (en) 2000-12-22 2003-08-19 Baxter International Inc. Microprecipitation method for preparing submicron suspensions
EP1348432A1 (en) 2001-01-04 2003-10-01 Daiichi Pharmaceutical Co., Ltd. Cyclodextrin-containing pharmaceutical preparation
US6742734B2 (en) 2001-06-05 2004-06-01 Elan Pharma International Limited System and method for milling materials
US7575184B2 (en) 2001-06-05 2009-08-18 Elan Pharma International Ltd. System and method for milling materials
WO2003002258A1 (en) 2001-06-29 2003-01-09 Eurand Pharmaceuticals Ltd Drug activation process and vibrational mill therefor
US7951399B2 (en) 2001-06-29 2011-05-31 Eurand Pharmaceuticals Ltd. Process for activation of drugs in a vibrational mill
US8158162B2 (en) 2001-11-14 2012-04-17 Jarrow Formulas, Inc. Eutectic-based self-nanoemulsified drug delivery system
US20050020294A1 (en) 2001-12-07 2005-01-27 Marko Kantola System for transmitting information in a digital radio network system
WO2003086363A1 (en) 2002-04-08 2003-10-23 Lavipharm Laboratories Inc. Drug-complex microparticles and methods of making/using same
US6921458B2 (en) 2002-12-19 2005-07-26 Acusphere, Inc. Methods and apparatus for making particles using spray dryer and in-line jet mill
US20040224024A1 (en) 2003-04-23 2004-11-11 Massachusetts Institute Of Technology Controlled drug release formulations containing polyion complexes
US20070110804A1 (en) 2003-06-20 2007-05-17 Royer Biomedical, Inc. Drug polymer complexes
US7994214B2 (en) 2003-08-29 2011-08-09 Lifecycle Pharma A/S Solid dispersions comprising tacrolimus
US20060013888A1 (en) 2003-09-29 2006-01-19 Ronald G. Udell Solubilized CoQ-10
WO2005037250A1 (en) 2003-10-17 2005-04-28 Amylin Pharmaceuticals, Inc. Self emulsifying drug delivery systems for hydrophobic therapeutic compounds
US8092785B2 (en) 2004-08-23 2012-01-10 Glaxo Group Limited Process for milling poorly soluble drugs in presence of liquid propellants
WO2006021160A1 (en) 2004-08-27 2006-03-02 Shijiazhuang Pharma. Group Zhongqi Pharmaceutical Technology (Shijiazhuang) Co., Ltd. Butylbenzene phthalein self-emulsifying drug delivery system, its preparation method and application
WO2006060698A1 (en) 2004-12-03 2006-06-08 Elan Pharma International Ltd. Nanoparticulate benzothiophene formulations
US20060159766A1 (en) 2004-12-15 2006-07-20 Elan Pharma International Limited Nanoparticulate tacrolimus formulations
US20060165806A1 (en) 2005-01-06 2006-07-27 Elan Pharma International Limited Nanoparticulate candesartan formulations
US20060188566A1 (en) 2005-02-24 2006-08-24 Elan Pharma International Limited Nanoparticulate formulations of docetaxel and analogues thereof
US7537786B2 (en) 2005-02-25 2009-05-26 Nec Corporation Complex of drug-carbon nanohorn and a process for producing the complex
WO2006096462A1 (en) 2005-03-03 2006-09-14 Elan Pharma International Limited Nanoparticulate compositions of heterocyclic amide derivatives
WO2006100281A2 (en) 2005-03-22 2006-09-28 Losan Pharma Gmbh Solubilized ibuprofen
WO2006110811A1 (en) 2005-04-12 2006-10-19 Elan Pharma International Limited Nanoparticulate quinazoline derivative formulations
WO2006112541A1 (en) 2005-04-20 2006-10-26 Chugai Seiyaku Kabushiki Kaisya Method for the design of self-emulsifying drug formulations
US8074906B2 (en) 2005-07-07 2011-12-13 Nanotherapeutics, Inc. Process for milling and preparing powders and compositions produced thereby
US8354122B2 (en) 2006-04-20 2013-01-15 Shin-Etsu Chemical Co., Ltd. Solid dispersion preparation
WO2007150030A2 (en) 2006-06-23 2007-12-27 Massachusetts Institute Of Technology Microfluidic synthesis of organic nanoparticles
WO2008002121A2 (en) 2006-06-28 2008-01-03 Hovid, Berhad A self-emulsifying composition for poorly bioavailable drugs
US8343548B2 (en) 2006-08-08 2013-01-01 Shin-Etsu Chemical Co., Ltd. Solid dosage form comprising solid dispersion
WO2009007992A2 (en) 2007-04-20 2009-01-15 Sun Pharmaceutical Industries Limited Pharmaceutical composition produced by microprecipitation
US7441717B1 (en) 2007-10-31 2008-10-28 Eastman Kodak Company Micromedia milling process
WO2009075401A1 (en) 2007-12-12 2009-06-18 Chung-Ang University Industry-Academy Cooperation Foundation Manufacturing method of functional drug nanoparticles using milling and functional drug nanoparticle formulation manufactured thereby
WO2009118322A1 (en) * 2008-03-28 2009-10-01 Boehringer Ingelheim International Gmbh Process for preparing orally administered dabigatran formulations
EP2288335A1 (en) 2008-03-28 2011-03-02 Boehringer Ingelheim International GmbH Process for preparing orally administered dabigatran formulations
WO2009133418A1 (en) 2008-04-28 2009-11-05 Nangenex Nanotechnology Incorporated Instrument and process for nanoparticles production in continuous flow mode
WO2010020601A1 (en) 2008-08-19 2010-02-25 Boehringer Ingelheim International Gmbh Dabigatran in tumour therapy
EP2328581A1 (en) 2008-08-19 2011-06-08 Boehringer Ingelheim International GmbH Use of dabigatranetexilate for treating patients with pulmonary hypertension
US20100322870A1 (en) 2008-11-11 2010-12-23 Boehringer Ingelheim International Gmbh Method for treating or preventing thrombosis using dabigatran etexilate or a salt thereof with improved efficacy over conventional warfarin therapy
WO2010133611A1 (en) 2009-05-18 2010-11-25 Royal College Of Surgeons In Ireland Solid drug dispersions
WO2010146407A1 (en) 2009-06-19 2010-12-23 Nanoform Hungary Ltd. Nanostructured sildenafil base, its pharmaceutically acceptable salts and co-crystals, compositions of them, process for the preparation thereof and pharmaceutical compositions containing them
WO2010146409A2 (en) 2009-06-19 2010-12-23 Nangenex, Inc. Nanoparticulate candesartan cilexetil compositions, process for the preparation thereof and pharmaceutical compositions containing them
WO2010146408A2 (en) 2009-06-19 2010-12-23 Nangenex, Inc. Nanoparticulate olmesartan medoxomil compositions, process for the preparation thereof and pharmaceutical compositions containing them
WO2010146406A1 (en) 2009-06-19 2010-12-23 Nanoform Hungary Ltd. Nanoparticulate telmisartan compositions and process for the preparation thereof
WO2011110876A1 (en) 2010-02-02 2011-09-15 Egis Gyógyszergyár Nyilvánosan Működő Részvénytársaság Novel salts for the manufacture of pharmaceutical compositions
WO2011110478A1 (en) * 2010-03-08 2011-09-15 Ratiopharm Gmbh Dabigatran etexilate-containing pharmaceutical composition
WO2011131943A2 (en) 2010-04-20 2011-10-27 Cipla Limited Pharmaceutical compositions
WO2011154755A1 (en) 2010-06-08 2011-12-15 Nanoform Cardiovascular Therapeutics Ltd. Nanostructured atorvastatin, its pharmaceutically acceptable salts and compositions of them, process for the preparation thereof and pharmaceutical compositions containing them
WO2011158052A1 (en) 2010-06-18 2011-12-22 Nanoform Cardiovascular Therapeutics Ltd. Nanostructured ezetimibe compositions, process for the preparation thereof and pharmaceutical compositions containing them
WO2011158053A1 (en) 2010-06-18 2011-12-22 Nanoform Cardiovascular Therapeutics Ltd. Nanostructured aprepitant compositions, process for the preparation thereof and pharmaceutical compositions containing them
EP2588090A2 (en) 2010-07-01 2013-05-08 Krka Tovarna Zdravil, D.D., Novo Mesto Pharmaceutical oral dosage forms comprising dabigatran etexilate and its pharmaceutically acceptable salts
WO2012027543A1 (en) 2010-08-25 2012-03-01 Teva Pharmaceuticals Usa, Inc. Solid state forms of dabigatran etexilate, dabigatran etexilate mesylate and processes for preparation thereof
WO2012044595A1 (en) 2010-09-27 2012-04-05 Ratiopharm Gmbh Dabigatran etexilate bismesylate salt, solid state forms and process for preparation thereof
WO2012042224A2 (en) 2010-10-01 2012-04-05 Cipla Limited Pharmaceutical composition
WO2012160559A1 (en) 2011-05-22 2012-11-29 Rappaport Family Institute For Research In The Medical Sciences Pharmaceutical compositions of d-alpha-tocopheryl acetate
WO2013110567A1 (en) 2012-01-24 2013-08-01 Boehringer Ingelheim International Gmbh Novel orally administered dabigatran formulation
CN102558153A (en) 2012-02-08 2012-07-11 北京阜康仁生物制药科技有限公司 Novel pharmaceutical salt of dabigatran etexilate and preparation method thereof
WO2013124340A1 (en) 2012-02-21 2013-08-29 Laboratorios Del Dr. Esteve, S.A. Oral pharmaceutical compositions of dabigatran etexilate
EP2631234A1 (en) 2012-02-23 2013-08-28 Esteve Química, S.A. Solid forms of dabigatran etexilate mesylate and processes for their preparation
WO2013175494A2 (en) * 2012-04-10 2013-11-28 Rubicon Research Private Limited Controlled release pharmaceutical formulations of direct thrombin inhibitors
WO2014131376A1 (en) * 2013-02-26 2014-09-04 Elmarco S.R.O. Electrospun nanofibers comprising pharmaceutically active agents

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
GAREKANI H A ET AL: "Increasing the aqueous solubility of acetaminophen in the presence of polyvinylpyrrolidone and investigation of the mechanisms involved", DRUG DEV IND PHARM, vol. 29, no. 2, February 2003 (2003-02-01), pages 173 - 179, XP002735410 *
M. KANSI ET AL., JOURNAL OF MEDICINAL CHEMISTRY, vol. 41, 1998, pages 1007
S. BENDELS ET AL., PHARMACEUTICAL RESEARCH, vol. 23, 2006, pages 2525
SARAVANA KUMAR K ET AL: "Dissolution Enhancement of Poorly Soluble Drugs by Using Complexation Technique - A Review", J. PHARM. SCI. & RES., vol. 5, no. 5, May 2013 (2013-05-01), pages 120 - 124, XP002735411 *
T. REINTJES: "Solubility Enhancement with BASF Pharma Polymers: Solubilizer Compendium", October 2011 (2011-10-01), XP002735409, Retrieved from the Internet <URL:http://www.pharma-ingredients.basf.com/Documents/ENP/Brochure/EN/b_03_110921e_Solubility_Enhance_Compendium.pdf> [retrieved on 20150203] *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105534939A (en) * 2015-12-29 2016-05-04 郑州大明药物科技有限公司 Dabigatran etexilate mesylate oral solid preparation and preparation method thereof
US10383865B2 (en) 2016-04-25 2019-08-20 Druggability Technologies Ip Holdco Limited Pharmaceutical combination composition comprising complex formulations of Ivacaftor and Lumacaftor and their salts and derivatives, process for their preparation thereof and pharmaceutical compositions containing them
US10376501B2 (en) 2016-04-25 2019-08-13 Druggability Technologies Ip Holdco Limited Complexes of lumacaftor and its salts and derivatives, process for the preparation thereof and pharmaceutical compositions containing them
WO2017187338A1 (en) * 2016-04-25 2017-11-02 Druggability Technologies Ip Holdco Limited Complexes of lumacaftor and its salts and derivatives, process for the preparation thereof and pharmaceutical compositions containing them
CN109475548A (en) * 2016-04-25 2019-03-15 成药技术Ip控股有限公司 According to cutting down the compound of Kato and its salt and derivative, preparation method and containing their pharmaceutical composition
CN109475547A (en) * 2016-04-25 2019-03-15 成药技术Ip控股有限公司 The compound of Lu Makatuo and its salt and derivative, preparation method and the pharmaceutical composition containing them
CN109475546A (en) * 2016-04-25 2019-03-15 成药技术Ip控股有限公司 Including according to the pharmaceutical combination composition for the compound formulation for cutting down Kato and Lu Makatuo and its salt and derivative, preparation method and containing their pharmaceutical composition
WO2017187340A1 (en) * 2016-04-25 2017-11-02 Druggability Technologies Ip Holdco Limited Pharmaceutical combination composition comprising complex formulations of ivacaftor and lumacaftor and their salts and derivatives, process for their preparation thereof and pharmaceutical compositions containing them
WO2017187336A1 (en) * 2016-04-25 2017-11-02 Druggability Technologies Ip Holdco Limited Complexes of ivacaftor and its salts and derivatives, process for the preparation thereof and pharmaceutical compositions containing them
CN111163807A (en) * 2017-09-19 2020-05-15 耶路撒冷希伯来大学伊森姆研究发展有限公司 Lipophilic peptide prodrugs
US11965041B2 (en) 2017-09-19 2024-04-23 Technische Universitaet Muenchen N-methylated cyclic peptides and their prodrugs
CN111902161A (en) * 2018-04-04 2020-11-06 上海汉都医药科技有限公司 Pharmaceutical composition containing dabigatran etexilate and preparation method thereof
EP3777892A4 (en) * 2018-04-04 2021-06-02 Shanghai WD Pharmaceutical Co., Ltd Pharmaceutical composition containing dabigatran etexilate and preparation method thereof
US11707455B2 (en) 2018-04-04 2023-07-25 Shanghai Wd Pharmaceutical Co., Ltd Pharmaceutical composition containing dabigatran etexilate and preparation method thereof
CN111902161B (en) * 2018-04-04 2023-12-15 上海汉都医药科技有限公司 Pharmaceutical composition containing dabigatran etexilate and preparation method thereof
CN113893356A (en) * 2020-11-27 2022-01-07 上海博志研新药物技术有限公司 Dabigatran etexilate mesylate inclusion compound, preparation method and application

Similar Documents

Publication Publication Date Title
WO2015071841A1 (en) Complexes of dabigatran and its derivatives, process for the preparation thereof and pharmaceutical compositions containing them
ES2627820T3 (en) Histone deacetylase inhibitor formulations in combination with bendamustine and uses thereof
CN103282036B (en) For treating the composition and method of myelofibrosis
WO2008072535A1 (en) Pharmaceutical composition containing low-substituted hydroxypropylcellulose
TW201731509A (en) Dosing regimens for 2-hydroxy-6-((2-(1-isopropyl-1H-pyrazol-5-yl)pyridin-3-yl)methoxy)benzaldehyde
EA015102B1 (en) Nanoparticulate imatinib mesylate formulations
EP3522872B1 (en) Amlodipine formulations
WO2004054574A1 (en) Solid drug for oral use
US20230338287A1 (en) Amlodipine formulations
JP2012530127A (en) Nanoparticulate candesartan cilexetil composition, process for its preparation and pharmaceutical composition containing them
JP2012530126A (en) Nanoparticulate olmesartan medoxomil composition, method for its preparation, and pharmaceutical composition containing them
ES2706067T3 (en) A pharmaceutical composition containing candesartan cilexetil and amlodipine
KR20160128449A (en) Pharmaceutical preparation containing calcium antagonist/angiotensin ii receptor antagonist
CN101653440B (en) Treatment composition containing amlodipine series salt and pril medicament
AU2006257428B2 (en) Oral solid pharmaceutical formulation of the tubulin inhibitor indibulin
CN101468002A (en) Therapeutic composition containing amlodipine salt and losartan medicine
AU2017382160A1 (en) Pharmaceutical formulations of suvorexant
WO2002067904A1 (en) Pharmaceutical composition which comprise a solid dispersion of a hydroxypropylmethylcellulose phthalate polymer
CN103006648A (en) Maleic acid levorotation amlodipine drug active pharmaceutical composition and preparation method thereof
EP2839835B1 (en) Encapsulated formulation
CN101433536A (en) Therapeutic compositions containing amlodipine niacin and losartan medicament
NO317771B1 (en) Eprosartanarginyl charge neutralization complex and a process for its preparation and use thereof
US10376501B2 (en) Complexes of lumacaftor and its salts and derivatives, process for the preparation thereof and pharmaceutical compositions containing them
CN102327220B (en) Solid loratadine lipidosome preparation
WO2007091253A2 (en) Pharmaceutical compositions comprising clopidogrel and vitamins which reduce homocysteine levels

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14815023

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14815023

Country of ref document: EP

Kind code of ref document: A1