NZ623423B2 - Solid dosage forms of (s)-ethyl 2-amino-3-(4-(2-amino-6-((r)-1-(4-chloro-2-(3-methyl-1h-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoate - Google Patents

Solid dosage forms of (s)-ethyl 2-amino-3-(4-(2-amino-6-((r)-1-(4-chloro-2-(3-methyl-1h-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoate Download PDF

Info

Publication number
NZ623423B2
NZ623423B2 NZ623423A NZ62342312A NZ623423B2 NZ 623423 B2 NZ623423 B2 NZ 623423B2 NZ 623423 A NZ623423 A NZ 623423A NZ 62342312 A NZ62342312 A NZ 62342312A NZ 623423 B2 NZ623423 B2 NZ 623423B2
Authority
NZ
New Zealand
Prior art keywords
tablet
phenyl
amino
trifluoroethoxy
chloro
Prior art date
Application number
NZ623423A
Other versions
NZ623423A (en
Inventor
Jinling Chen
Matthew S Deaver
Richard J Holl
Kalyan Nuguru
Original Assignee
Tersera Therapeutics Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Tersera Therapeutics Llc filed Critical Tersera Therapeutics Llc
Priority to NZ717137A priority Critical patent/NZ717137B2/en
Priority claimed from PCT/US2012/060338 external-priority patent/WO2013059146A1/en
Publication of NZ623423A publication Critical patent/NZ623423A/en
Publication of NZ623423B2 publication Critical patent/NZ623423B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1617Organic compounds, e.g. phospholipids, fats
    • A61K9/1623Sugars or sugar alcohols, e.g. lactose; Derivatives thereof; Homeopathic globules
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1652Polysaccharides, e.g. alginate, cellulose derivatives; Cyclodextrin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • A61K9/2077Tablets comprising drug-containing microparticles in a substantial amount of supporting matrix; Multiparticulate tablets
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2095Tabletting processes; Dosage units made by direct compression of powders or specially processed granules, by eliminating solvents, by melt-extrusion, by injection molding, by 3D printing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2893Tablet coating processes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Abstract

Provided are solid stable pharmaceutical dosage forms comprising (S)-ethyl 2-amino-3-(4-(2-amino-6-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoate (telotristat). In an embodiment the dosage form is a tablet comprising (S)-ethyl 2-amino-3-(4-(2-amino-6-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoate, lactose, hydroxy propyl cellulose, croscarmellose sodium, magnesium stearate, and silicon dioxide; wherein the tablet forms less than 1.0 percent (S)-2-amino-3-(4-(2-amino-6-((R)-1-(4- chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoic acid when stored at 40°C and 75% relative humidity for six months; and wherein the tablet has a disintegration time of less than 5.5 minutes in water. -(2-amino-6-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoate, lactose, hydroxy propyl cellulose, croscarmellose sodium, magnesium stearate, and silicon dioxide; wherein the tablet forms less than 1.0 percent (S)-2-amino-3-(4-(2-amino-6-((R)-1-(4- chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoic acid when stored at 40°C and 75% relative humidity for six months; and wherein the tablet has a disintegration time of less than 5.5 minutes in water.

Description

WO 59146 SOLID DOSAGE FORMS OF (S)—ETHYL 2—AMINO—3—(4—(2—AMINO—6—((R)—1—(4— CHLORO—2—(3—M ETHYL—1H—PYRAZOL—1—YL)PHENYL)—2,2,2— OROETHOXY)PYRIM|DIN—4—YL)PHENYL)PROPANOATE This ation claims priority to US. provisional patent application no. 61/547,894, filed October 17, 2011, the entirety of which is orated herein by reference. 1. FIELD OF THE INVENTION This invention relates to solid pharmaceutical dosage forms of (S)—ethyl 2-amino—3—(4— (2-amino-6—((R)—1—(4-chloro(3—methyl-1H-pyrazolyl)phenyl)—2,2,2-trifluoroethoxy)pyrimidin- 4-yl)phenyl)propanoate (telotristat). 2. OUND OF THE INVENTION The compound (S)-ethyl 2-amino(4-(2-amino((R)-1—(4-chloro(3-methyl-1H- pyrazolyl)phenyl)—2,2,2-trifluoroethoxy)pyrimidinyl)phenyl)propanoate (telotristat) is an inhibitor of tryptophan hydroxylase, the enzyme responsible for the rate-limiting step in biosynthesis of 5-hydroxytryptamine onin). See, e.g., US. patent no. 7,709,493. The compound is believed to be useful in the treatment of es and disorders associated with abnormal levels of serotonin, such as diarrhea-predominant ble bowel syndrome and carcinoid syndrome. Unfortunately, istat’s physicochemical properties make its incorporation into a commercially viable dosage form difficult.
Telotristat hydrolyzes when contacted with water. Dosage forms comprising it must, 2O therefore, limit this degradation as much as possible, and must be made using methods that limit the compounds exposure to re. The poor flowability of telotristat’s crystalline hippurate salt (telotristat etiprate) further complicates the manufacture of dosage forms comprising it. Further adding to the problem is the desire to provide single unit dosage forms that contain at least 100 mg of the compound, and that rapidly release it upon oral stration.
In view of these factors, a need exists for solid dosage forms of telotristat that can be stored at typical temperatures and humidity levels for a commercially viable period of time, and for methods of their manufacture. Preferred dosage forms should be capable of rapidly ring the compound upon oral administration. A particular need exists for a rapid release tablet formulation of telotristat with good chemical stability, satisfactory oral bioavailability, good processability, and high drug loading. 3. SUMMARY OF THE INVENTION This invention is directed to solid dosage forms of telotristat. Particular dosage forms are tablets made with the hippurate salt of telotristat (telotristat etiprate).
One embodiment of the ion encompasses a tablet le for administration to a patient comprising at least 100, 200, or 300 mg of an active pharmaceutical ient (API), which tablet has a disintegration time of less than 10, 5.0, 2.3, 2.0, or 1.8 s in water, wherein the API is telotristat or a pharmaceutically acceptable salt thereof.
Another ment encompasses a tablet suitable for administration to a patient comprising at least 100, 200, or 300 mg of an API based on free base, which tablet comprises acoating and has a disintegration time of less than 5.5, 4.5, or 4.0 minutes in water, n the API is telotristat or a pharmaceutically acceptable salt thereof.
Another embodiment encompasses a tablet having a core consisting essentially of istat hippurate, lactose, hyrdroxy propyl cellulose, croscarmellose sodium, magnesium te, and silicon dioxide.
Another embodiment encompasses a tablet comprising telotristat or a ceutically acceptable salt thereof, which forms less than 1.0, 0.8 or 0.5 percent (S)-2— amino-3—(4—(2—amino-6—((R)-1—(4-chloro(3—methyl-1H-pyrazol-l—yl)phenyl)—2,2,2— trifluoroethoxy)pyrimidinyl)phenyl)propanoic acid when stored at about 40°C and about 75% relative humidity for six months.
Another embodiment encompasses a tablet comprising telotristat or a ceutically acceptable salt thereof, which forms less than 0.5 or 0.4 percent (S)—2— amino-3—(4—(2—amino-6—((R)-1—(4-chloro(3—methyl-1H-pyrazol-l—yl)phenyl)—2,2,2— 2O trifluoroethoxy)pyrimidinyl)phenyl)propanoic acid when stored at about 40°C and about 75% relative humidity for three months.
Another embodiment encompasses a granule comprising telotristat etiprate, lactose, hydroxyl propyl cellulose, croscarmellose sodium, magnesium stearate, and silicon dioxide.
Another embodiment encompasses a method of making a tablet, which comprises: combining granules comprising intragranular ients with at least one extragranular ingredient, and compressing the granules to provide a tablet; wherein the intragranular ingredients comprise telotristat or a pharmaceutically acceptable salt f, magnesium stearate, and e; and at least one extragranular ingredient is lactose. 4. BRIEF DESCRIPTION OF THE FIGURES 3O Certain aspects of the invention can be understood with reference to the appended figures.
Figure 1 shows an X—ray powder diffraction (XRPD) pattern of a crystalline form of istat. The diffractogram was obtained using a Rigaku MiniFlex diffractometer (copper Kor radiation).
Figure 2 provides an XRPD pattern of a crystalline form of telotristat etiprate. The diffractogram was obtained using a Bruker D8 Advance r Kor radiation).
Figure 3 shows the effects of temperature, humidity and time on the formation of the hydrolysis product (S)—2-amino—3-(4-(-2amino--((R)(-4c-h-loro2--(-3methyl-1H--p-yrazol1-- yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidinyl)phenyl)propanoic acid in ent dosage forms of telotristat.
. DETAILED PTION OF THE INVENTION This invention is directed to solid pharmaceutical dosage forms in which an active pharmaceutical ingredient (API) is (S)—ethyl 2-amino(4-(2-amino---6((R)(c-hloro2--(3- methyl-1H-pyrazolyl)phenyl)-2,2,2-trifluoroethoxy)pyrimidinyl)phenyl)propanoate (telotristat): / /N MONE or a pharmaceutically able salt thereof. The compound, its salts and crystalline forms can be obtained by methods known in the art. See, e.g., U.S. patent no. 7,709,493.
Particular dosage forms se crystalline telotristat freebase. One form of this nd has a g point of about 104°C as determined by differential scanning calorimetry (DSC) (onset temperature). As used in connection with DSC temperatures, the term “about” means 13°C. This form provides an X-ray powder diffraction (XRPD) pattern that contains peaks at about 10.7, 12.2, 12.8, 17.7 and/or 22.0 degrees 29. As used in connection with XPRD peaks, the term “about” means 10.3 degrees 29. As those skilled in the art are well aware, the relative intensities of peaks in an XRPD pattern of a crystalline 2O material can vary depending on how the sample is prepared and how the data is collected.
With this in mind, an example of an XRPD pattern of this crystalline form is provided in Figure Particular dosage forms comprise the hippurate salt of telotristat (telotristat hippurate; telotristat etiprate). A particular crystalline form of this salt has a melting point of about 142°C (DSC onset temperature, with a peak at about . A particular crystalline form provides an XRPD pattern that contains peaks at about 8.2, 9.5, 12.6, 16.9, 21.8, 22.0, 22.7, 24.3 and/or 29.1 degrees 29. An example of an XRPD n of this form is provided in Figure 2.
When contacted with water, telotristat can hydrolyze to form (S)—2-amino(4-(2- amino--((R)(4--c-hloro2--(3-methyl-1H--pyrazol-y-l)phenyl)2, 2, 2--trifluoroethoxy)pyrimidin- yl)phenyl)propanoic acid. Preferred dosage forms of this ion minimize this degradation.
Figure 3 shows the difference between two tablets of the invention—formulations 6 and 8, described in the examples below—and a capsule dosage form that was used in human Phase 1 and 2 clinical . The capsules contained a mixture of 250 mg telotristat and 2% magnesium stearate. Both tablet formulations are clearly more stable than the e formulation.
The bioavailability of an API can greatly depend on the ation in which it is red to a patient. Here, tablets that y disintegrate when stered to a patient are desired. Particular non-coated tablets of this invention have a disintegration time of less than 2.3, 2.0, or 1.8 minutes in water, or less than 4.0, 3.0, or 2.7 minutes in 0.1 N HCI.
Particular -film coated tablets of the invention have a disintegration time of less than 5.5, 4.5, or 4.0 s in water, or less than 5.4, 5.0, or 4.8 minutes in 0.1 N HCI. As used herein, the term “disintegration time" refers to disintegration time in 100 mL of purified water or 0.1 N HCI as measured according to test USP <701>. The disintegration of a tablet can be affected by the disintegrants it contains. Examples of disintegrants include alginates, oses, croscarmellose , crospovidone, and sodium starch glycolate. A preferred disintegrant is croscarmellose sodium.
The ability of a tablet to rapidly disintegrate or dissolve must be balanced, however, with the necessity that the tablet not fall apart in its packaging. Thus, particular s of the invention have a hardness greater than 8, 9, or 10 kP, and a friability of less than 0.4, 0.3, or 2O 0.25 (percent loss).
The hardness and stability of a tablet are affected by the excipients it contains. The excipients can also affect the ease with which a tablet is made (e.g., by affecting how well the ingredients from which it is made flow and ss). Particular tablets of the invention se telotristat te, cellulose, lactose, rmellose sodium, magnesium stearate, and silicon dioxide This invention encompasses methods of making solid dosage forms of telotristat and salts thereof that limit the compounds exposure to water and address the poor flow properties exhibited by many of its forms. In a particular embodiment, roller compaction is used to prepare a granular material (“granulate”) made of up granules comprising the 3O compound, which is then combined with additional excipients and compressed to provide a tablet core. The core is then optionally coated to increase the stability of the resulting tablet.
Particulate granules comprise telotristat etiprate, ypropyl cellulose, lactose, rmellose sodium, ium stearate, and silicon dioxide. Preferred granulates flow and compress well, allowing the ready manufacture of tablets possessing the desired hardness, stability, and disintegration properties described herein.
The solid dosage forms (e.g., tablets) of the invention can be packaged by methods and using containers known in the art. The packaging material may form a plurality of divided containers such as a divided bottle or a divided foil packet. The container can be in any conventional shape or form as known in the art which is made of a pharmaceutically acceptable al, for example a paper or cardboard box, a glass or plastic bottle orjar with or without desiccant, a re-sealable bag (for example, to hold a “refill” of tablets for placement into a different container), or a blister pack (e.g., Aclar bilsters or oil blisters) with individual doses for pressing out of the pack according to a therapeutic schedule. In a preferred ment, tablets are stored in an induction-sealed HDPE bottle with a desiccant pack. 6. EXAMPLES 6.1. Tablet and Ingredient Characterization egration g was performed as per USP <701> using the test for uncoated tables and plain coated tablets. The disintegration was performed in 1000 mL purified water or 0.1 N HCI. Disintegration endpoint was determined visually.
Dissolution was determined in 900 mL of 0.1 N HCI at 37°C using USP Apparatus 2 (paddles) set at 50 rpm. Filtrates of the dissolution test solution were collected at specific time als. The samples were analyzed by high performance liquid chromatography (HPLC) usinga PhenomenexSynergi 4p Max-RP column and a mobile phase of 70/30/02 (v/v/v) methanol/water/phosphoric acid at a flow rate of 1.0 mL/min. The HPLC system utilized ultraviolet (UV) detection at a wavelength of 237 nm. 2O Granulation particle size was determined using a sieve method, wherein the tare weight of each of several sieves (mesh 25, 40, 60, 100, 140, 230, and Fines) was recorded, the sieves were stacked in order of the coarsest sieve on top and the finest on , and imately 5 grams of the granulate material was transferred to the top sieve. The assembly was secured and placed in an ATM Sonic sifter, the pulse amplitude and sift amplitudes both set to 5. After 5 minutes, the assembly was d and the individual sieves d. Flow properties were determined using a J.R. Johanson Flow lndicizer. 6.2. General Tablet Preparation Tablets comprising 300 mg (measured as free base) of the API telotristat in the hippurate salt form were made in two general steps. First, granules comprising crystalline telotristat te and selected excipients (intragranular ents) were prepared. The material was compressed using a roller compactor and milled. The intragranular material was then combined with additional excipients (“extragranular components"), and the resulting mixture was ssed to provide the tablets. In some cases, the s were coated.
WO 59146 Batches were ed by screening all intragranular materials except ium stearate through a 20-mesh screen. Components were blended in an appropriately sized V- blender for 10 minutes. lntragranular magnesium stearate was combined with a portion of the blend and co-screened through a 20-mesh screen. The screened magnesium stearate blend was then charged into the V-blender and blended for an additional three s. The blend was then roller compacted using a Vector TF-Mini roller compactor with a target ribbon thickness of 1.5 mm. The ribbons were milled by sequentially oscillating them through a 14- mesh and 20-mesh screen. All extragranular components except magnesium stearate were combined and screened through a 20-mesh screen. Approximately half of the ation was charged into the V-blender ed by the screened extragranular components. The remaining half of the granulation was charged into the V-blender and blended for five minutes. A small portion of the blend was removed and combined with the magnesium stearate and passed through a 20-mesh screen. The ium stearate blend was charged into the V-blender and blended for an additional three minutes. The final blend was compressed into LX—1606 300-mg tablets. Some s were film coated in a Strea 1 Fluid Bed Coater with Opadry 2 Clear to a 4% weight gain. 6.3. Formulation 1 In this example, tablets were made from the ingredients listed below in Table 1: Table 1 lntragranular Components (mg/tablet) API 402.12* Citric Acid, Anhydrous 83.79 Lactose, Anhydrous 90.77 Hydroxy Propyl ose 34.91 Croscarmellose Sodium 20.95 Magnesium Stea rate 3.49 Extragranular Components (mg/tablet) Lactose, Anhydrous 34.28 Croscarmellose Sodium 20.95 Colloidal Silicon Dioxide 3.49 Core Tablet Total 700.0 *Equivalent to 300 mg telotristat free base First, the intragranular components were mixed and roller compacted with a roller pressure of 70 kg/cm2. The ribbons were 0.99 — 1.42 mm in ess. A bench-top ribbon disintegration test was med by g a one inch section of ribbon in a beaker containing approximately 500 mL of DI water and allowed to disintegrate. The ribbon disintegrated in 12.5 minutes. tion of the roller compactor rollers indicated that some sticking had occurred. Ribbons were milled by oscillating sequentially through a 14-mesh and 20-mesh screen. The granulation was blended with ranular components and physical tests were med. The granules flowed poorly, and the initial tablets exhibited weight ions and low average tablet weight. Striations and chipping were also observed on the first tablets produced. lnitial tablets also failed a friability test loss limit of S 0.8%, yet ng prevented the compression forces from being increased to improve the friability.
These problems were addressed by increasing the extragranular magnesium te by 0.25%, and blended with the remaining blend (the amount of magnesium stearate shown in Table 1 reflects this additional amount). The resulting final blend was compressed into tablets (0.300" x 0.680" capsule shaped tooling). No further sticking was observed.
Characteristics of the granulation and tablets are shown below in Table 2: Table 2 Approximate Ribbon Disintegration Time (min) 12.5 Bulk Density (g/mL) 0.6644 Tapped Density (g/mL) 0.886 Average Flow Rate Index (kg/sec) 0.511 Core Hardness Range (kP) 8.1 — 12.0 Average Core Weight (g) 0.679 Average Tablet Thickness (mm) 5.65 Tablet lity (% loss) 0.3 The tablets’ dissolution properties are shown below in Table 3: Table 3 6.4. ation 2 In this example, tablets were made from the ingredients listed below in Table 4: Table 4 lntragranular Components (mg/tablet) API 403.13 Citric Acid, Anhydrous 84.00 Microcrystalline Cellulose 89.25 Hydroxy Propyl Cellulose 35.00 Croscarmellose Sodium 28.00 Magnesium te 5.25 Extragranular Components (mg/tablet) Microcrystalline Cellulose 18.62 Croscarmellose Sodium 28.00 Colloidal n Dioxide 3.50 Core Tablet Total 700.0 First, the intragranular components were mixed and roller compacted with a roller pressure of 45 kg/cm2. The ribbon thicknesses ranged from 1.16 — 1.46 mm. Bench-top ribbon disintegration test resulted in a disintegration time of 3 minutes. Some sticking was noted during the roller compaction of the blend. The ribbons were milled by oscillating tially h a 14-mesh and 20—mesh screen. The ribbons were hard and more difficult to mill. Approximately 0.75% of the batch did not pass through the oscillator. The granulation was blended with extragranular components and physical tests were performed.
Granulation exhibited poor flow characteristics, although the compression was manageable.
Some sticking to tablet punches was observed initially during compression, which subsided after the punches were cleaned. The tablets exhibited a dull appearance, which did not improve when the compression force was increased. 6.5. Formulation 3 In this e, tablets were prepared using the ingredients listed below in Table 5: Table 5 lntragranular Components (mg/tablet) Citric Acid, Anhydrous rystalline Cellulose Hydroxy Propyl Cellulose Crospovidone 28.00 Magnesium Stearate 5.25 Extragranular Components (mg/tablet) Microcrystalline Cellulose 18.62 Crospovidone 28.00 Colloidal Silicon Dioxide 3.50 Core Tablet Total 700.0 The mixture of intragranular components was roller ted with a roller pressure of 50 kg/cm2. The ribbon thicknesses ranged from 1.40 — 1.90 mm. Bench-top ribbon egration test resulted in an undesirable disintegration time of 11 minutes. Some sticking was observed during the roller compaction process. The ribbons were similar to ation 2 and were difficult to mill. Granulation was blended with ranular components and physical tests were performed. The granulation exhibited poorflow, and some rat-holing was observed in the hopper during compression, which was me by agitating the hopper. Tablet compression was completed with no observable problems.
However, tablet disintegration testing in water and 0.1N HCI resulted in disintegration times icantly longer than those observed the other formulations, suggesting that in these formulations, crospovidone is a less effective egrant than croscarmellose . 6.6. ation 4 In this example, granules were prepared using the ingredients listed below in Table 6: Table 6 lntragranular Components (mg/tablet) Citric Acid, Anhydrous Mannitol Microcrystalline Cellulose Hydroxy Propyl Cellulose Crospovidone Magnesium Stea rate Because the disintegration tests run on the ribbons made from this mixture showed a disintegration time of 11 minutes, further work on this formulation was not done. 6.7. Formulation 5 In this example, tablets were prepared using the ingredients listed below in Table 7: Table 7 lntragranular Components (mg/tablet) API 403.13 Citric Acid, Anhydrous 84.00 ol 44.45 Hydroxy Propyl Cellulose 35.00 Croscarmellose Sodium 28.00 Magnesium Stearate 5.25 Extragranular Components (mg/tablet) Microcrystalline Cellulose 18.62 Croscarmellose Sodium 28.00 Colloidal n Dioxide 3.50 Core Tablet Total 700.0 The e of intragranular ents was roller compacted with a roller pressure of 50 kg/cm2. The ribbon thickness ranged from 1.37 — 1.83 mm. Bench-top ribbon disintegration time was 1 minute. Minor sticking was observed throughout the roller compaction process. The granulation was blended with the extragranular ents and physical tests were performed. The granulation exhibited poor flow, but tablet compression was completed with no observable problems. The formulation was capable of achieving hardnesses exceeding 18 kP. Tablet disintegration testing in water and 0.1N HCI resulted in able disintegration times for an immediate release : 2.0 s in water, 4.0 — .25 minutes in 0.1N HCI. However, assay and related substance testingindicated that an unacceptable amount of what is believed to be a hydrolysis product of the API increased significantly at the one-month time point when stored at 5% RH without desiccant.
An additional batch of Formulation 5 was manufactured, and in this case, the resulting tablets were coated with Opadry Clear. The granulation lot was roller ted with a roller pressure of 50 kg/cm2, affording a ribbon thickness ranging from 1.24 — 1.57 mm. Bench-top ribbon disintegration time was 3.25 minutes. Minor sticking to the rollers was observed throughout the roller compaction process. Blend was also observed to be sticking to the walls of the hopper and exhibited poor flow. The granulation was blended with the extragranular components and physical tests were performed. ng was observed after 5 minutes of tablet compression. The punches were cleaned and compression was restarted, but tablet ng resumed immediately, suggesting that the granulation may require additional lubrication or increased lubrication time to overcome sticking issues. The resulting tablets were coated to a 4% weight gain. The dissolution profile of these tablets was acceptable, although the disintegration times in water and 0.1 N HCI were significantly longer than the uncoated tablets. Assay and related substance gindicated that coating the tablet to a theoretical weight gain of 4% ses the level of degradation, a level which is further decreased with the use of desiccant. 6.8. Formulation 6 In this example, tablets were prepared using the ingredients listed below in Table 8: Table 8 lntragranular Components (mg/tablet) API 403.13 Mannitol 86.45 Microcrystalline Cellulose 86.45 Hydroxy Propyl Cellulose 35.00 rmellose Sodium 28.00 Magnesium Stearate 5.25 Extragranular Components (mg/tablet) Microcrystalline ose 18.97 Croscarmellose Sodium 28.00 Colloidal Silicon e 3.50 Core Tablet Total 700.0 The mixture of intragranular ents was roller compacted with a roller pressure of 50 kg/cm2. The ribbon thickness ranged from 1.11 — 1.52 mm. Bench-top ribbon disintegration time was 1 . Very little sticking was observed throughout the roller compaction process. Although the granulation exhibited poor flow, it was blended and compressed into tablets, during which some sticking was observed. Tablets exhibited some chipping during friability testing. Dissolution and egration times were: 1.3 — 1.5 minutes in water; 1.5 — 2.8 minutes in 0.1 N HCI. These tablets particularly stable (0.23 area percent after 1 month at 40°C/75 % relative humidity), and more so when stored with desiccant (0.16 area percent after 1 month at 40°C/75 % ve humidity). 6.9. Formulation 7 In this example, tablets were prepared using the ingredients listed below in Table 9: Table 9 lntragranular Components (mg/tablet) API 403.13 Citric Acid, Anhydrous 84.00 Lactose, ous 80.50 Hydroxy Propyl Cellulose 35.00 Croscarmellose Sodium 28.00 Magnesium Stearate 5.25 Extragranular Components (mg/tablet) Lactose, Anhydrous 27.37 Croscarmellose Sodium 28.00 Colloidal Silicon Dioxide 3.50 Core Tablet Total 700.0 The mixture of intragranular components was roller compacted with a roller pressure of 50 . The ribbon thickness ranged from 1.45 — 1.63 mm. Bench-top ribbon disintegration time was 3 minutes. Very little sticking was observed during roller compaction.
Granulation, which exhibited poorflow, was d and compressed into tablets. Sticking was observed on the punch faces and die walls during tablet compression. Chipping was also noted during friability testing. Dissolution and disintegration times were acceptable, although assay and related substance testing indicated a significant se in apparent API hydrolysis product when stored for one month under rated conditions without desiccant (1.01 area percent after 1 month at 5 % relative humidity). Desiccant decreased the observed level of hydrolysis product to 0.16. 6.10. ation 8 In this example, tablets were prepared using the ingredients listed below in Table 10: Table 10 lntragranular Components (mg/tablet) Lactose, Anhydrous Hydroxy Propyl Cellulose Croscarmellose Sodium Magnesium Stea rate Extragranular Components (mg/tablet) Lactose, ous 27.37 Croscarmellose Sodium 21.00 Colloidal Silicon Dioxide 3.50 Core Tablet Total 700.0 The mixture of intragranular components was roller compacted with a roller re of 55 kg/cm2. The ribbon thickness ranged from 1.07 — 1.52 mm. The material processed very well, yielding long ribbons. Bench-top ribbon egration time was 2.5 minutes.
Approximately 2% of the ribbons did not pass through the 20-mesh oscillating screen.
Granulation was blended and ssed into tablets. The blend compressed well and no sticking was observed. Some minor picking was observed.
Physical characteristics of the granulation and tablets are shown below in Table 11: Table 11 Approximate Ribbon Disintegration Time (min) Core Hardness Range (kP) 8.5 — 11.9 Average Core Weight (g) 0.711 Average Tablet Thickness (mm) Tablet Friability (% loss) The tablets’ disintegration profile was acceptable: uncoated tablets disintegrated in 1.8 — 2.3 minutes in water and 2.7 — 4.0 minutes in 0.1N HCI; coated tablets disintegrated in 3.1 — 5.5 minutes in water and 4.4 — 5.4 minutes in 0.1N HCI. The dissolution e of the tablets is shown below in Table 12: Table 12 Mean % Label Claim Time (min) ed Tablets Coated Tablets 93.6 84.6 98.3 94.7 99.5 96.0 45 99.8 96.0 This formulation performed well during the stability study, with little of the hydrolysis product observed in the ed s without desiccant (0.39 area percent after 1 month 2012/060338 at 40°C/75 % relative humidity), in uncoated s with desiccant (0.32 area t after 1 month at 40°C/75 % relative humidity), in coated tablets with desiccant (0.31 area percent after 1 month at 40°C/75 % relative humidity), in Aclar blisters (0.42 area percent after 1 month at 40°C/75 % relative humidity), and in foil/foil blisters(0.39 area percent after 1 month at 40°C/75 % relative ty). 6.11. Stability Determination The stability of tablets was determined by a reverse-phase HPLC-based method ing the ing conditions: Column: Waters XTerra MS C18 (4.6 X 150 mm, 3.5 pm Particle Size) Autosampler 5°C Temperature: Mobile Phase A: 0.05% TFA in Water Mobile Phase B: 0.05% TFA in ACN Flow Rate: 1.0 mL/minute Detection 254 nm Wavelength: Data Acquisition 41 minutes Time: Data Output: Ensure Peak is on Scale The pump program used was: Time (min) % Mobile Phase A % Mobile Phase B A standard solution was prepared by dissolving telotristat etiprate in THF with a concentration of approximately 0.25 ug/mL. s were prepared from 300 mg tablets as follows: 1) at least 4 tablets were weighed; 2) then crushed using a mortar and pestle; 3) an amount of equivalent to about 50 mg drug substance (i.e., about 117 mg) was weighed and transferred to a 100-mL volumetric flask; 4) then diluted to about 1/2 to 2/3 volume with diluent (THF); 5) the flask was then placed on a shaker for at least 20 minutes at low speed; 6) the volume was then further diluted with diluent and mixed well; 7) an aliquot was centrifuged for about 5 s at approximately 3000 RPM; 8) an aliquot of the supernate was then withdrawn for injection; 9) steps 3 through 8 were repeated for a total of two replicates for injection; 10) the average retention time of the API peak was then determined for the first six injections of the rd solution; and 2) the ratio of the retention time of any peaks in the sample preparation to the average retention time of the API peak in the first six injections of the standard was then calculated.
Potency was determined using the following equation: API (mg) per tablet = (Asample 7" Wtota|)/ (RFstd 7" e 7" Ntotal) *DFsample where: Asample = API sample peak area; Wtotal = total weight of the tablets (mg); DFsample = sample dilution volume in mL (100 mL for the 300-mg s); RFstd = rd average response factor (1st 6 ions); Wsample = Individual sample weight (mg); and Ntotal = Number of tablets used (at least 4). Individual impurities were determined as a percent of the total integrated peak area.
All references cited herein (e.g., patents and patent applications) are incorporated herein in their entireties.

Claims (17)

CLAIMS What is claimed is:
1. A tablet sing (S)-ethyl o(4-(2-amino((R)(4-chloro(3-methyl- 1H-pyrazolyl)phenyl)-2,2,2-trifluoroethoxy)pyrimidinyl)phenyl)propanoate or a pharmaceutically acceptable salt thereof, e, hydroxy propyl cellulose, croscarmellose sodium, magnesium stearate, and silicon dioxide; wherein the tablet forms less than 1.0 percent (S)amino(4-(2-amino((R)(4- chloro(3-methyl-1H-pyrazolyl)phenyl)-2,2,2-trifluoroethoxy)pyrimidinyl)phenyl)propanoic acidwhen stored at 40°C and 75% relative humidity for six months; and wherein the tablet has a disintegration time of less than 5.5 minutes in water.
2. The tablet of claim 1, which forms less than 0.5 or 0.4 percent (S)amino(4- no((R)(4-chloro(3-methyl-1H-pyrazolyl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin yl)phenyl)propanoic acid when stored at about 40°C and about 75% relative humidity for three months.
3. The tablet of claim 1, wherein the (S)-ethyl 2-amino(4-(2-amino((R)(4- chloro(3-methyl-1H-pyrazolyl)phenyl)-2,2,2-trifluoroethoxy)pyrimidinyl)phenyl)propanoate pharmaceutically acceptable salt thereof is a hippurate salt of hyl 2-amino(4-(2-amino ((R)(4-chloro(3-methyl-1H-pyrazolyl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin yl)phenyl)propanoate.
4. The tablet of claim 3, wherein the hippurate salt of(S)-ethyl o(4-(2- amino((R)(4-chloro(3-methyl-1H-pyrazolyl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin yl)phenyl)propanoate is crystalline.
5. The tablet of claim 1, wherein the tablet is enterically coated.
6. The tablet of claim 1, wherein the tablet comprises at least 100 mg (S)-ethyl 2- amino(4-(2-amino((R)(4-chloro(3-methyl-1H-pyrazolyl)phenyl)-2,2,2- trifluoroethoxy)pyrimidinyl)phenyl)propanoate or a ceutically acceptable salt thereof.
7. The tablet of claim 6, wherein the tablet comprises at least 200 mg (S)-ethyl 2- amino(4-(2-amino((R)(4-chloro(3-methyl-1H-pyrazolyl)phenyl)-2,2,2- trifluoroethoxy)pyrimidinyl)phenyl)propanoate or a pharmaceutically acceptable salt thereof.
8. The tablet of claim 1, which has a disintegration time of less than 4.5 minutes in water.
9. The tablet of claim 1, which has a disintegration time of less than 4.0 minutes in water.
10. The tablet of claim 1, which has a disintegration time of less than 5.4 minutes in 0.1 N HCl.
11. The tablet of claim 10, which has a disintegration time of less than 5.0 minutes in 0.1 N HCl.
12. The tablet of claim 1, which has a hardness greater than 8 kP.
13. The tablet of claim 12, which has a hardness greater than 9 kP.
14. The tablet of claim 13, which has a hardness greater than 10 kP.
15. The tablet of claim 1, which has a lity of less than 0.4.
16. The tablet of claim 15, which has a friability of less than 0.3.
17. The tablet of claim 16, which has a friability of less than 0.25. WO 59146 SE . cg RD E \ O Lk 3000 2500 2000 1500 1000 500 AJISNHlNI WO 59146 V N ,<_‘: % O N 7 K5 0 \ '— Lk 60 O O 0 0 LO <' M N 10 AlISNEIlNI 20:53sz 20:53sz Iol I-I Amazes Imxmtoé h moéem 6E (z) d SISA'IOEICIAH
NZ623423A 2011-10-17 2012-10-16 Solid dosage forms of (s)-ethyl 2-amino-3-(4-(2-amino-6-((r)-1-(4-chloro-2-(3-methyl-1h-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoate NZ623423B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
NZ717137A NZ717137B2 (en) 2011-10-17 2012-10-16 Solid dosage forms of (s)-ethyl 2-amino-3-(4-(2-amino-6-((r)-1-(4-chloro-2-(3-methyl-1h-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoate

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161547894P 2011-10-17 2011-10-17
US61/547,894 2011-10-17
PCT/US2012/060338 WO2013059146A1 (en) 2011-10-17 2012-10-16 Solid dosage forms of (s)-ethyl 2-amino-3-(4-(2-amino-6-((r)-1-(4-chloro-2-(3-methyl-1h-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoate

Publications (2)

Publication Number Publication Date
NZ623423A NZ623423A (en) 2016-04-29
NZ623423B2 true NZ623423B2 (en) 2016-08-02

Family

ID=

Similar Documents

Publication Publication Date Title
US11406635B2 (en) Solid dosage forms of (S)-ethyl 2-amino-3-(4-(2-amino-6-((R)-1-(4-chloro-2-(3-methyl-1H-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoate
AU2009254574B2 (en) Solid pharmaceutical formulations comprising BIBW 2992
EP3881833B1 (en) Immediate-release tablets containing a drug and processes for forming the tablets
KR101663838B1 (en) Wet granulation using a water sequestering agent
JP2019142927A (en) Pharmaceutical dosage forms
JP6737060B2 (en) Method for producing pharmaceutical composition containing irbesartan
EP3996688B1 (en) Pharmaceutical preparation
NZ623423B2 (en) Solid dosage forms of (s)-ethyl 2-amino-3-(4-(2-amino-6-((r)-1-(4-chloro-2-(3-methyl-1h-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoate
NZ717137B2 (en) Solid dosage forms of (s)-ethyl 2-amino-3-(4-(2-amino-6-((r)-1-(4-chloro-2-(3-methyl-1h-pyrazol-1-yl)phenyl)-2,2,2-trifluoroethoxy)pyrimidin-4-yl)phenyl)propanoate
JP7101464B2 (en) A method for improving the quality of azilsartan or a salt thereof and amlodipine or a salt-containing tablet thereof, and azilsartan or a salt thereof and amlodipine or a salt-containing tablet thereof and a method for producing the same.
US10420728B2 (en) Tablet and method of preparing the same
JP2022102543A (en) Tablet containing oseltamivir and method for producing the same
CN115023221A (en) Stable immediate release tablet and capsule formulations of 1- ((2S,5R) -5- ((7H-pyrrolo [2,3-D ] pyrimidin-4-yl) amino) -2-methylpiperidin-1-yl) prop-2-en-1-one
WO2017073738A1 (en) Tablet having fexofenadine as effective component thereof
CN116782888A (en) Pharmaceutical composition
WO2014191443A1 (en) Method for producing dosage form comprising odanacatib