WO2022079591A1 - Pharmaceutical composition of lasmiditan and process of preparation thereof - Google Patents

Pharmaceutical composition of lasmiditan and process of preparation thereof Download PDF

Info

Publication number
WO2022079591A1
WO2022079591A1 PCT/IB2021/059337 IB2021059337W WO2022079591A1 WO 2022079591 A1 WO2022079591 A1 WO 2022079591A1 IB 2021059337 W IB2021059337 W IB 2021059337W WO 2022079591 A1 WO2022079591 A1 WO 2022079591A1
Authority
WO
WIPO (PCT)
Prior art keywords
pharmaceutical
pharmaceutical composition
lasmiditan
hemisuccinate
subject matter
Prior art date
Application number
PCT/IB2021/059337
Other languages
French (fr)
Inventor
Sagar SHETH
Mehul UMARAVKAR
Viral Vaidya
Original Assignee
Alembic Pharmaceuticals 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
Application filed by Alembic Pharmaceuticals Limited filed Critical Alembic Pharmaceuticals Limited
Publication of WO2022079591A1 publication Critical patent/WO2022079591A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • 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
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • 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
    • A61K9/2833Organic macromolecular compounds
    • A61K9/286Polysaccharides, e.g. gums; Cyclodextrin
    • A61K9/2866Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose

Definitions

  • the present subject matter relates to a solid oral pharmaceutical composition
  • a solid oral pharmaceutical composition comprising lasmiditan or a pharmaceutically acceptable salt thereof and at least one pharmaceutical acceptable excipient, wherein the solid oral pharmaceutical composition is prepared by a dry manufacturing process.
  • Lasmiditan (COL-144, LY573144) is a serotonin (5-HT) IF receptor agonist. It was developed by CoLucid (now part of Eli Lilly) for the treatment of migraine. Lasmiditan is available as hemisuccinate salt and is also known by its chemical name: 2,4,6- trifluoro- N- [6-(l- methylpiperidine -4- carbonyl) pyridine-2-yl] benzamide hemisuccinate salt and is reported to have the following chemical structure:
  • Lasmiditan is approved in the USA as 50 mg and 100 mg film coated tablets under the brand name Reyvow®. It is administered orally, as needed, with or without food for the acute treatment of migraine with or without aura in adults.
  • Lasmiditan hemisuccinate is a white, crystalline powder that is sparingly soluble in water, slightly soluble in ethanol, and soluble in methanol. A 1 mg/mL aqueous solution of lasmiditan hemisuccinate is estimated to have a pH of 6.8 at ambient conditions.
  • United States Patent No. US8748459 discloses lasmiditan and its hemisuccinate salt. It further discloses method of treatment of migraine in a mammal comprising administering to a mammal effective amount of lasmiditan or salt thereof.
  • US20100256187 discloses oral delivery compositions comprising 50 to 200mg of lasmiditan or salt therefore for the treatment of migraine.
  • US’ 187 publication also discloses lasmiditan compositions in the form of dry powder inhaler, tablet, capsule, suspension, intravenous formulation, sublingual or buccal tablet.
  • the sublingual and buccal tablets are also disclosed with HC1 and succinate salts of lasmiditan. It further discloses specific tablets containing certain excipients and also discloses wet granulation as manufacturing process for one such tablet.
  • United States Patent No. US8697876 discloses solid crystalline Form A, Form B and Form C of lasmiditan hemisuccinate.
  • US2020087279 discloses new pseudo-polymorphic crystalline Form D, Form E and Form F of lasmiditan hemisuccinate. It also discloses that starting with either Form A or the amorphous form of lasmiditan hemisuccinate, addition of water alone or water/solvent mixtures results in the conversion from Form A or the amorphous form to one of the pseudo-polymorphic Forms D, E, or F.
  • Form A is the dominant form in the absence of water (up to 10% by volume of water; a water activity of approximately 0.6) or at low water activities, e.g. in pure ethanol, at any temperature. At higher water activities, temperature and water content are determinative.
  • the present subject matter relates to a solid oral pharmaceutical composition comprising lasmiditan or its pharmaceutically acceptable salt there of as an active ingredient and at least one excipients, wherein the solid oral pharmaceutical composition is prepared by dry manufacturing process.
  • the present subject matter also relates to a non-aqueous wet granulation process.
  • the present subject matter relates to a solid oral pharmaceutical composition
  • a solid oral pharmaceutical composition comprising lasmiditan or its pharmaceutically acceptable salt thereof and at least one pharmaceutically acceptable excipient, wherein the solid oral pharmaceutical composition is prepared by a dry manufacturing process.
  • the present subject matter relates to solid oral pharmaceutical composition
  • solid oral pharmaceutical composition comprising lasmiditan hemisuccinate as an active ingredient and at least one excipient, wherein the solid oral pharmaceutical composition is prepared by a dry manufacturing process.
  • the dry manufacturing process according to the subject matter refers to processes such as direct compression process, dry granulation process, roller compaction etc.
  • Another objective of the present subject matter is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising,
  • Another objective of the present subject matter is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising,
  • Another objective of the present subject matter is to provide pharmaceutical composition comprising,
  • Another objective of the present subject matter is to provide a pharmaceutical composition comprising,
  • Another objective of the present subject matter is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising,
  • Another objective of the present subject matter is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising,
  • Another objective of the present subject matter is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising,
  • Another objective of the present subject matter is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising,
  • the present subject matter relates to a solid oral pharmaceutical composition
  • a solid oral pharmaceutical composition comprising lasmiditan or a pharmaceutically acceptable salt thereof as an active ingredient, at least one pharmaceutically acceptable excipient, where in the solid oral pharmaceutical composition is prepared by a dry manufacturing process.
  • the dry manufacturing process according to the subject matter refers to a process generally known in the art.
  • Non-limiting examples of such processes are direct compression process, dry granulation process, roller compaction etc.
  • the non-aqueous wet granulation process according to the subject matter refers to a process generally known in the art such as rapid mixer granulation, fluidized bed granulation, spray granulation etc.
  • solvent used in non-aqueous wet granulation process is selected from isopropyl alcohol, ethanol, methanol, acetone, dichloromethane, acetonitrile or mixture thereof.
  • composition may comprise lasmiditan in the ‘free base form’ or as a pharmaceutically acceptable salt, or as any mixture thereof.
  • lasmiditan present in pharmaceutical composition is in the form of hemisuccinate salt form.
  • lasmiditan hemisuccinate is in crystalline form.
  • lasmiditan hemisuccinate crystalline form is polymorphic Form A (wherein polymorphic Form A of the lasmiditan hemisuccinate may be defined in any of the ways described in United States Patent No. US8697876).
  • crystalline lasmiditan hemisuccinate salt exhibits an x-ray diffraction pattern using Cu-Ka radiation includes peaks at about 15.3, 16.4, 19.3, 22.1, 23.6 and 25.9 degrees 29.
  • composition comprising,
  • composition comprising,
  • Suitable diluents can be selected from the group of, but not limited to microcrystalline cellulose (MCC), pregelatinized starch, sugar and sugar alcohol such as mannitol, lactose, fructose, isomalt, lactitol, maltitol, sorbitol, erythritol, maltose, polydextrose, sucrose, trehalose, xylitol, calcium carbonate, dicalcium phosphate, calcium sulfate, cellulose acetate, ethylcellulose, inulin, magnesium carbonate, magnesium oxide, maltodextrin, sodium bicarbonate, sodium carbonate and sodium chloride.
  • MCC microcrystalline cellulose
  • sugar alcohol such as mannitol, lactose, fructose, isomalt, lactitol, maltitol, sorbitol, erythritol, maltose, polydextrose, sucrose, trehalose,
  • the diluent is microcrystalline cellulose.
  • binder is intended to be interpreted in the context of pharmaceutical formulation science.
  • Suitable binder according to the present subject matter can be selected from the group of, but not limited to povidone, pregelatinized starch, cellulose, methyl cellulose, ethyl cellulose, cellulose derivatives (e.g., hydroxypropyl methyl cellulose (HPMC), hydroxypropyl cellulose (HPC)), and polyethylene glycol.
  • the binder is povidone.
  • disintegrant and “disintegrants” are intended to be interpreted in the context of pharmaceutical formulation science.
  • Suitable disintegrants according to the present subject matter can be selected from but not limited to croscarmellose sodium (CCS), sodium starch glycolate (SSG), pregelatinized starch, alginic acid, sodium alginate, carboxymethylcellulose calcium, chitosan, crospovidone, glycine, guar gum, hydroxypropyl cellulose, low-substituted hydroxypropyl cellulose, magnesium aluminum silicate, methylcellulose, povidone, sodium alginate, sodium carboxymethylcellulose and starch.
  • CCS croscarmellose sodium
  • SSG sodium starch glycolate
  • pregelatinized starch alginic acid, sodium alginate, carboxymethylcellulose calcium, chitosan, crospovidone, glycine, guar gum, hydroxypropyl cellulose, low-substituted hydroxypropyl cellulose, magnesium aluminum si
  • one or more disintegrants are selected from the group of croscarmellose sodium, pregelatinized starch or a mixture thereof.
  • Suitable pharmaceutical surfactant can be selected from but not limited to sodium lauryl sulfate (SLS), poloxamers, polysorbate, sodium dodecyl benzene sulfonate, alkali metal or ammonium salts of lauroyl sarcosinate, myristoyl sarcosinate, palmitoyl sarcosinate, stearoyl sarcosinate and oleoyl sarcosinate, polyoxyethylene sorbitan monostearate, isostearate and laurate, sodium lauryl sulfoacetate, N-lauroyl sarcosine, the sodium, potassium, and ethanolamine salts of N-lauroyl, N-myristoyl or N-palmitoyl sarcosine, polyethylene oxide condensates of alkyl phenol
  • the surfactant is sodium lauryl sulphate.
  • lubricant and “lubricants” are intended to be interpreted in the context of pharmaceutical formulation science.
  • Suitable pharmaceutical lubricant according to the present subject matter can be selected from but not limited to magnesium, aluminium, zinc or calcium stearate, glyceryl behenate, glyceryl dibehenate, glyceryl monostearate, glyceryl palmitostearate, a mixture of behenate esters of glycerine (e.g.
  • glyceryl dibehenate tribehenin and glyceryl behenate
  • magnesium stearate myristic acid, palmitic acid, stearic acid, talc, tribehenin, sodium stearyl fumarate (SSF) and sucrose stearate.
  • SSF sodium stearyl fumarate
  • the one or more pharmaceutical lubricants comprise magnesium stearate, stearic acid or a mixture thereof.
  • the lubricant is magnesium stearate.
  • Glidants are frequently used in tablet formulations to improve flow.
  • glidant and “glidants” are intended to be interpreted in the context of pharmaceutical formulation science.
  • Suitable glidants according to the present subject matter can be selected from but not limited to magnesium oxide, colloidal silicon dioxide, pyrogenic silica and hydrated sodium silicoaluminate.
  • glidant is colloidal silicon dioxide.
  • a pharmaceutical tablet comprising the pharmaceutical composition as defined herein.
  • a pharmaceutical tablet comprising a pharmaceutical composition in the form of tablet core, wherein the tablet core is coated with a layer of coating.
  • the coating is a film coating.
  • the film coating may be applied using conventional methods.
  • a coating can be used to provide protection against, for example, moisture ingress or degradation by light, to colour the formulation or to modify or control the release of lasmiditan hemisuccinate from the formulation.
  • the pharmaceutical composition is a pharmaceutical tablet composition (for oral administration).
  • the pharmaceutical composition is a pharmaceutical tablet composition suitable for oral administration to a human.
  • the pharmaceutical composition is a pharmaceutical tablet composition suitable for oral administration for the treatment of migraine.
  • the pharmaceutical tablet comprises 100 mg lasmiditan free base, equivalent to 115.65 mg of lasmiditan hemisuccinate respectively.
  • the pharmaceutical tablet comprises 50 mg lasmiditan free base, equivalent to 57.824 mg of lasmiditan hemisuccinate respectively.
  • a pharmaceutical composition as defined herein, for the manufacture of a medicament for the treatment of migraine.
  • a pharmaceutical composition for use as a medicament.
  • a pharmaceutical composition is a pharmaceutical tablet.
  • a method of acute treatment of migraine with or without aura in adults in need thereof comprises the oral administration of the pharmaceutical tablet(s), as needed, to the patient.
  • lasmiditan hemisuccinate tablet is prepared by dry manufacturing process or a non-aqueous wet manufacturing process.
  • Table 2 Pharmaceutical composition of Lasmiditan hemisuccinate.
  • Lasmiditan hemisuccinate, microcrystalline cellulose (Avicel PH 112), povidone, croscarmellose sodium (Ac-di-sol) and sodium lauryl sulfate were sifted and mixed properly to get uniform blend.
  • Magnesium stearate was sifted and a blend obtained in step 1 was lubricated using magnesium stearate in step 2.
  • step 3 The lubricated blend of step 2 was compressed into tablets. Core tablets were coated using Opadry dispersion with inlet temperature of 60-65°C and product temperature 38-44°C.
  • Above example 2 can be prepared using a dry manufacturing process, e.g. direct compression process, dry granulation process, roller compaction etc.
  • Dissolution tests The dissolution described herein was performed according to the general procedure of the United States Pharmacopoeia using Apparatus II (Paddle), at 75 rpm with 500 ml of 0.01N HC1 media at a temperature of 37° C. The samples were withdrawn at 5, 10, 15, 20 and 30 minutes.
  • the intragranular material was sifted and mixed properly to get uniform blend.
  • Magnesium stearate was sifted and a blend obtained in step 1 was lubricated using magnesium stearate in step 2.
  • step 3 The lubricated blend of step 2 was compressed into tablets. Core tablets were coated using Opadry dispersion with inlet temperature of 60-65°C and product temperature of 38-44°C.
  • Step 1 excipients were mixed with lasmiditan and pass through Quadro comil with 0.610mm screen.
  • step 4 Lubricated blend of step 4 was compressed into tablets. Core tablets were coated using Opadry dispersion with inlet temperature of 60-65°C and product temperature of 38-44°C.
  • Lasmiditan hemisuccinate, microcrystalline cellulose (Avicel PH 112), L- HPC LH-11 and sodium lauryl sulphate were passed through suitable sieve and mixed properly to get uniform blend.
  • step 2 The material obtained in step 1 was subjected to roller compaction and granules were prepared.
  • Step 3 material was lubricated using magnesium stearate and compressed into tablets. Core tablets were coated using Opadry dispersion with inlet temperature of 60-65°C and product temperature of 38-44°C.
  • Lasmiditan hemisuccinate, microcrystalline cellulose (Avicel PH 112), PVPK-30, croscarmellose sodium and sodium lauryl sulphate were passed through suitable sieve and mixed properly to get uniform blend.
  • step 3 The material obtained in step 3 was milled and granules were obtained.
  • Extragranular microcrystalline cellulose (Avicel PH 112) and croscarmellose sodium were sieved and mixed with step 4 granules.
  • Step 5 material was lubricated using magnesium stearate and compressed into tablets. Core tablets were coated using Opadry dispersion with inlet temperature of 60-65°C and product temperature of 38-44°C.
  • Lasmiditan hemisuccinate, sodium lauryl sulfate, microcrystalline cellulose (Avicel PH 112) and L HPC LH 11/ croscarmellose sodium were passed through suitable sieve and mixed properly to get uniform blend.
  • Step 1 material was granulated with solvent (IPA/Ethanol).
  • step 6 Lubricated blend of step 6 was compressed into tablet at target weight i.e. 230 mg.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present subject matter relates to a solid oral pharmaceutical composition comprising lasmiditan or pharmaceutically acceptable salt thereof and at least one pharmaceutically acceptable excipient, wherein the solid oral pharmaceutical composition is prepared by a dry manufacturing process.

Description

PHARMACEUTICAL COMPOSITION OF LASMIDITAN AND PROCESS
OF PREPARATION THEREOF
TECHNICAL FIELD:
The present subject matter relates to a solid oral pharmaceutical composition comprising lasmiditan or a pharmaceutically acceptable salt thereof and at least one pharmaceutical acceptable excipient, wherein the solid oral pharmaceutical composition is prepared by a dry manufacturing process.
BACKGROUND:
Lasmiditan (COL-144, LY573144) is a serotonin (5-HT) IF receptor agonist. It was developed by CoLucid (now part of Eli Lilly) for the treatment of migraine. Lasmiditan is available as hemisuccinate salt and is also known by its chemical name: 2,4,6- trifluoro- N- [6-(l- methylpiperidine -4- carbonyl) pyridine-2-yl] benzamide hemisuccinate salt and is reported to have the following chemical structure:
Figure imgf000002_0001
Lasmiditan is approved in the USA as 50 mg and 100 mg film coated tablets under the brand name Reyvow®. It is administered orally, as needed, with or without food for the acute treatment of migraine with or without aura in adults.
Lasmiditan hemisuccinate is a white, crystalline powder that is sparingly soluble in water, slightly soluble in ethanol, and soluble in methanol. A 1 mg/mL aqueous solution of lasmiditan hemisuccinate is estimated to have a pH of 6.8 at ambient conditions. United States Patent No. US8748459 discloses lasmiditan and its hemisuccinate salt. It further discloses method of treatment of migraine in a mammal comprising administering to a mammal effective amount of lasmiditan or salt thereof.
United States Patent Application Publication US20100256187 (US’ 187 publication) discloses oral delivery compositions comprising 50 to 200mg of lasmiditan or salt therefore for the treatment of migraine.
US’ 187 publication also discloses lasmiditan compositions in the form of dry powder inhaler, tablet, capsule, suspension, intravenous formulation, sublingual or buccal tablet. The sublingual and buccal tablets are also disclosed with HC1 and succinate salts of lasmiditan. It further discloses specific tablets containing certain excipients and also discloses wet granulation as manufacturing process for one such tablet.
United States Patent No. US8697876 discloses solid crystalline Form A, Form B and Form C of lasmiditan hemisuccinate.
United States Patent Application No. US2020087279 (US’279) discloses new pseudo-polymorphic crystalline Form D, Form E and Form F of lasmiditan hemisuccinate. It also discloses that starting with either Form A or the amorphous form of lasmiditan hemisuccinate, addition of water alone or water/solvent mixtures results in the conversion from Form A or the amorphous form to one of the pseudo-polymorphic Forms D, E, or F. Form A is the dominant form in the absence of water (up to 10% by volume of water; a water activity of approximately 0.6) or at low water activities, e.g. in pure ethanol, at any temperature. At higher water activities, temperature and water content are determinative.
Further US’279 disclose that conversion of Form A to Form D occurs as water is introduced, for example, through high humidity conditions or in solvent-water mixtures with a high water activity. Form D is the most stable form at intermediate water activities and higher temperatures. Polymorphic Form A of lasmiditan hemisuccinate is unstable in the presence of water and can convert to other pseudo-polymorphic Forms such as D, E or F. It also discloses that lasmiditan in the form of the dihydrate form D can be produced by subjecting the form A of lasmiditan to a wet granulation process using purified water as the granulating medium.
There is a need to develop a solid oral pharmaceutical composition containing lasmiditan or its pharmaceutically acceptable salt thereof in stable crystalline form, wherein the solid oral pharmaceutical composition is prepared by dry manufacturing process. Alternatively, the manufacturing process is a non-aqueous wet granulation process.
Accordingly, the present subject matter relates to a solid oral pharmaceutical composition comprising lasmiditan or its pharmaceutically acceptable salt there of as an active ingredient and at least one excipients, wherein the solid oral pharmaceutical composition is prepared by dry manufacturing process. The present subject matter also relates to a non-aqueous wet granulation process.
SUMMARY OF THE SUBJECT MATTER:
The present subject matter relates to a solid oral pharmaceutical composition comprising lasmiditan or its pharmaceutically acceptable salt thereof and at least one pharmaceutically acceptable excipient, wherein the solid oral pharmaceutical composition is prepared by a dry manufacturing process.
Further, the present subject matter relates to solid oral pharmaceutical composition comprising lasmiditan hemisuccinate as an active ingredient and at least one excipient, wherein the solid oral pharmaceutical composition is prepared by a dry manufacturing process. The dry manufacturing process according to the subject matter refers to processes such as direct compression process, dry granulation process, roller compaction etc.
Another objective of the present subject matter is to provide a pharmaceutical composition comprising,
(a) Lasmiditan or a pharmaceutically acceptable salt thereof,
(b) one or more pharmaceutical diluent,
(c) one or more pharmaceutical binder,
(d) one or more pharmaceutical disintegrant,
(e) one or more pharmaceutical surfactant,
(f) one or more pharmaceutical lubricant.
Another objective of the present subject matter is to provide a pharmaceutical composition comprising,
(a) Lasmiditan hemisuccinate,
(b) one or more pharmaceutical diluent,
(c) one or more pharmaceutical binder,
(d) one or more pharmaceutical disintegrant,
(e) one or more pharmaceutical surfactant,
(f) one or more pharmaceutical lubricant.
Another objective of the present subject matter is to provide pharmaceutical composition comprising,
(a) Lasmiditan hemisuccinate Form A,
(b) one or more pharmaceutical diluent,
(c) one or more pharmaceutical binder,
(d) one or more pharmaceutical disintegrant,
(e) one or more pharmaceutical surfactant,
(f) one or more pharmaceutical lubricant. Another objective of the present subject matter is to provide a pharmaceutical composition comprising,
(a) Lasmiditan hemisuccinate Form A,
(b) one or more pharmaceutical diluent,
(c) one or more pharmaceutical disintegrant,
(d) one or more pharmaceutical surfactant,
(e) one or more pharmaceutical lubricant.
Another objective of the present subject matter is to provide a pharmaceutical composition comprising,
(a) Lasmiditan hemisuccinate Form A,
(b) one or more pharmaceutical diluent,
(c) one or more pharmaceutical disintegrant,
(d) one or more pharmaceutical surfactant,
(e) one or more pharmaceutical lubricant,
(f) one or more pharmaceutical glidant.
Another objective of the present subject matter is to provide a pharmaceutical composition comprising,
(a) Lasmiditan hemisuccinate Form A,
(b) one or more pharmaceutical diluent,
(c) one or more pharmaceutical disintegrant,
(d) one or more pharmaceutical surfactant,
(e) one or more pharmaceutical lubricant. wherein a pharmaceutical composition is prepared by dry manufacturing process.
Another objective of the present subject matter is to provide a pharmaceutical composition comprising,
(a) Lasmiditan hemisuccinate Form A,
(b) one or more pharmaceutical diluent, (c) one or more pharmaceutical binder,
(d) one or more pharmaceutical disintegrant,
(e) one or more pharmaceutical surfactant,
(f) one or more pharmaceutical lubricant wherein a pharmaceutical composition is prepared by dry manufacturing process.
Another objective of the present subject matter is to provide a pharmaceutical composition comprising,
(a) Lasmiditan hemisuccinate Form A,
(b) one or more pharmaceutical diluent,
(c) optionally one or more pharmaceutical binder,
(d) one or more pharmaceutical disintegrant,
(e) one or more pharmaceutical surfactant,
(f) one or more pharmaceutical lubricant wherein a pharmaceutical composition is prepared by a non-aqueous wet granulation process.
DETAILED DESCRIPTION OF THE SUBJECT MATTER:
Accordingly, the present subject matter relates to a solid oral pharmaceutical composition comprising lasmiditan or a pharmaceutically acceptable salt thereof as an active ingredient, at least one pharmaceutically acceptable excipient, where in the solid oral pharmaceutical composition is prepared by a dry manufacturing process.
The dry manufacturing process according to the subject matter refers to a process generally known in the art. Non-limiting examples of such processes are direct compression process, dry granulation process, roller compaction etc. The non-aqueous wet granulation process according to the subject matter refers to a process generally known in the art such as rapid mixer granulation, fluidized bed granulation, spray granulation etc.
In an embodiment, solvent used in non-aqueous wet granulation process is selected from isopropyl alcohol, ethanol, methanol, acetone, dichloromethane, acetonitrile or mixture thereof.
In an embodiment, the processes described on this subject matter does not involve substantial use of water.
For the purpose of the present subject matter, pharmaceutical composition may comprise lasmiditan in the ‘free base form’ or as a pharmaceutically acceptable salt, or as any mixture thereof.
In one embodiment, lasmiditan present in pharmaceutical composition is in the form of hemisuccinate salt form.
In one embodiment, lasmiditan hemisuccinate is in crystalline form.
In one embodiment, lasmiditan hemisuccinate crystalline form is polymorphic Form A (wherein polymorphic Form A of the lasmiditan hemisuccinate may be defined in any of the ways described in United States Patent No. US8697876).
In one embodiment, crystalline lasmiditan hemisuccinate salt exhibits an x-ray diffraction pattern using Cu-Ka radiation includes peaks at about 15.3, 16.4, 19.3, 22.1, 23.6 and 25.9 degrees 29.
Further, the present subject matter provides a pharmaceutical composition comprising,
(a) Lasmiditan hemisuccinate, (b) one or more pharmaceutical diluent,
(c) one or more pharmaceutical binder,
(d) one or more pharmaceutical disintegrant,
(e) one or more pharmaceutical surfactant,
(f) one or more pharmaceutical lubricant.
Further, the present subject matter provides a pharmaceutical composition comprising,
(a) Lasmiditan hemisuccinate,
(b) one or more pharmaceutical diluent,
(c) one or more pharmaceutical disintegrant,
(d) one or more pharmaceutical surfactant,
(e) one or more pharmaceutical lubricant.
The term “diluent” is intended to be interpreted in the context of pharmaceutical formulation science. Suitable diluents according to the present subject matter can be selected from the group of, but not limited to microcrystalline cellulose (MCC), pregelatinized starch, sugar and sugar alcohol such as mannitol, lactose, fructose, isomalt, lactitol, maltitol, sorbitol, erythritol, maltose, polydextrose, sucrose, trehalose, xylitol, calcium carbonate, dicalcium phosphate, calcium sulfate, cellulose acetate, ethylcellulose, inulin, magnesium carbonate, magnesium oxide, maltodextrin, sodium bicarbonate, sodium carbonate and sodium chloride.
In one of the aspect, the diluent is microcrystalline cellulose.
The term “binder” is intended to be interpreted in the context of pharmaceutical formulation science. Suitable binder according to the present subject matter can be selected from the group of, but not limited to povidone, pregelatinized starch, cellulose, methyl cellulose, ethyl cellulose, cellulose derivatives (e.g., hydroxypropyl methyl cellulose (HPMC), hydroxypropyl cellulose (HPC)), and polyethylene glycol. In one of the aspect, the binder is povidone.
The terms “disintegrant” and “disintegrants” are intended to be interpreted in the context of pharmaceutical formulation science. Suitable disintegrants according to the present subject matter can be selected from but not limited to croscarmellose sodium (CCS), sodium starch glycolate (SSG), pregelatinized starch, alginic acid, sodium alginate, carboxymethylcellulose calcium, chitosan, crospovidone, glycine, guar gum, hydroxypropyl cellulose, low-substituted hydroxypropyl cellulose, magnesium aluminum silicate, methylcellulose, povidone, sodium alginate, sodium carboxymethylcellulose and starch.
In one aspect, one or more disintegrants are selected from the group of croscarmellose sodium, pregelatinized starch or a mixture thereof.
The terms “surfactant” and “surfactants” are intended to be interpreted in the context of pharmaceutical formulation science. Suitable pharmaceutical surfactant according to the present subject matter can be selected from but not limited to sodium lauryl sulfate (SLS), poloxamers, polysorbate, sodium dodecyl benzene sulfonate, alkali metal or ammonium salts of lauroyl sarcosinate, myristoyl sarcosinate, palmitoyl sarcosinate, stearoyl sarcosinate and oleoyl sarcosinate, polyoxyethylene sorbitan monostearate, isostearate and laurate, sodium lauryl sulfoacetate, N-lauroyl sarcosine, the sodium, potassium, and ethanolamine salts of N-lauroyl, N-myristoyl or N-palmitoyl sarcosine, polyethylene oxide condensates of alkyl phenols, cocoamidopropyl betaine, lauramidopropyl betaine, palmityl betaine and the like.
In one aspect, the surfactant is sodium lauryl sulphate.
The terms “lubricant” and “lubricants” are intended to be interpreted in the context of pharmaceutical formulation science. Suitable pharmaceutical lubricant according to the present subject matter can be selected from but not limited to magnesium, aluminium, zinc or calcium stearate, glyceryl behenate, glyceryl dibehenate, glyceryl monostearate, glyceryl palmitostearate, a mixture of behenate esters of glycerine (e.g. a mixture of glyceryl dibehenate, tribehenin and glyceryl behenate), magnesium stearate, myristic acid, palmitic acid, stearic acid, talc, tribehenin, sodium stearyl fumarate (SSF) and sucrose stearate.
In one aspect, the one or more pharmaceutical lubricants comprise magnesium stearate, stearic acid or a mixture thereof.
In further aspect, the lubricant is magnesium stearate.
Glidants are frequently used in tablet formulations to improve flow. In this specification the terms “glidant” and “glidants” are intended to be interpreted in the context of pharmaceutical formulation science. Suitable glidants according to the present subject matter can be selected from but not limited to magnesium oxide, colloidal silicon dioxide, pyrogenic silica and hydrated sodium silicoaluminate.
In one aspect glidant is colloidal silicon dioxide.
As used herein, the word “comprising” describes components that must be present, but leaves open the possibility that other unspecified components may also be present within the scope of the relevant term.
In one aspect there is provided a pharmaceutical tablet comprising the pharmaceutical composition as defined herein.
In one embodiment there is provided a pharmaceutical tablet comprising a pharmaceutical composition in the form of tablet core, wherein the tablet core is coated with a layer of coating. In one embodiment the coating is a film coating. When the tablet has a film coating, the film coating may be applied using conventional methods. A coating can be used to provide protection against, for example, moisture ingress or degradation by light, to colour the formulation or to modify or control the release of lasmiditan hemisuccinate from the formulation.
In one embodiment, the pharmaceutical composition is a pharmaceutical tablet composition (for oral administration).
In one embodiment, the pharmaceutical composition is a pharmaceutical tablet composition suitable for oral administration to a human.
In one embodiment, the pharmaceutical composition is a pharmaceutical tablet composition suitable for oral administration for the treatment of migraine.
In one embodiment, the pharmaceutical tablet comprises 100 mg lasmiditan free base, equivalent to 115.65 mg of lasmiditan hemisuccinate respectively.
In one embodiment, the pharmaceutical tablet comprises 50 mg lasmiditan free base, equivalent to 57.824 mg of lasmiditan hemisuccinate respectively.
In a further aspect, there is provided the use of a pharmaceutical composition, as defined herein, for the manufacture of a medicament.
In one embodiment, there is provided the use of a pharmaceutical composition, as defined herein, for the manufacture of a medicament for the treatment of migraine.
In one aspect, there is provided a pharmaceutical composition, as defined herein, for use as a medicament.
In one embodiment, a pharmaceutical composition is a pharmaceutical tablet. In one embodiment, there is provided a method of acute treatment of migraine with or without aura in adults in need thereof, which method comprises the oral administration of the pharmaceutical tablet(s), as needed, to the patient.
According to present subject matter, lasmiditan hemisuccinate tablet is prepared by dry manufacturing process or a non-aqueous wet manufacturing process.
Embodiments provided herein may be more fully understood by reference to the following examples. These examples are meant to be illustrative of pharmaceutical composition and dosage form provided herein, but is not in any way limiting.
Example 1
Table 1 - Pharmaceutical composition of Lasmiditan hemisuccinate
Figure imgf000013_0001
Figure imgf000014_0001
Example 2
Table 2 - Pharmaceutical composition of Lasmiditan hemisuccinate.
Figure imgf000014_0002
Procedure
1. Lasmiditan hemisuccinate, microcrystalline cellulose (Avicel PH 112), povidone, croscarmellose sodium (Ac-di-sol) and sodium lauryl sulfate were sifted and mixed properly to get uniform blend.
2. Magnesium stearate was sifted and a blend obtained in step 1 was lubricated using magnesium stearate in step 2.
3. The lubricated blend of step 2 was compressed into tablets. Core tablets were coated using Opadry dispersion with inlet temperature of 60-65°C and product temperature 38-44°C.
Above example 2 can be prepared using a dry manufacturing process, e.g. direct compression process, dry granulation process, roller compaction etc.
Dissolution tests The dissolution described herein was performed according to the general procedure of the United States Pharmacopoeia using Apparatus II (Paddle), at 75 rpm with 500 ml of 0.01N HC1 media at a temperature of 37° C. The samples were withdrawn at 5, 10, 15, 20 and 30 minutes.
The dissolution profile of tablets having the Example 2 formulation was tested in 0.01N HC1 dissolution media. The results are presented in Table 3.
Table 3- Dissolution data of Lasmiditan hemisuccinate 100 mg Tablet
(Example 2) composition
Figure imgf000015_0001
Examples 3 to 10
Table 4 - Pharmaceutical composition of Lasmiditan hemisuccinate
Figure imgf000015_0002
Procedure
1. The intragranular material was sifted and mixed properly to get uniform blend.
2. Magnesium stearate was sifted and a blend obtained in step 1 was lubricated using magnesium stearate in step 2.
3. The lubricated blend of step 2 was compressed into tablets. Core tablets were coated using Opadry dispersion with inlet temperature of 60-65°C and product temperature of 38-44°C.
The dissolution profile of tablet formulations in Examples 3 to 10 were tested in 0. IN HC1 dissolution media. The results are presented in Table 5.
Table 5- Dissolution data of Lasmiditan hemisuccinate tablet composition
(Example 3 to 10)
Figure imgf000016_0001
Table 6- Stability data of Lasmiditan tablet composition of Ex.5
Figure imgf000016_0002
Figure imgf000017_0001
Example 11
Table 7 - Pharmaceutical composition of Lasmiditan hemisuccinate
Figure imgf000017_0002
Procedure
1. Sodium lauryl sulfate and microcrystalline cellulose (Avicel PH 112) were passed through Quadro comil with 0.610mm screen.
2. Step 1 excipients were mixed with lasmiditan and pass through Quadro comil with 0.610mm screen.
3. L-HPC LH-11 was mixed with Step 2 excipients and pass through Quadro comil with 0.610mm screen.
4. Magnesium stearate was mixed with mixture of step 3 excipients.
5. Lubricated blend of step 4 was compressed into tablets. Core tablets were coated using Opadry dispersion with inlet temperature of 60-65°C and product temperature of 38-44°C.
Table 8- Dissolution data of Lasmiditan hemisuccinate Tablet (Example 11) composition
Figure imgf000017_0003
Figure imgf000018_0001
Example 12
Table 9 - Pharmaceutical composition of Lasmiditan hemisuccinate
Figure imgf000018_0002
Procedure:
1. Lasmiditan hemisuccinate, microcrystalline cellulose (Avicel PH 112), L- HPC LH-11 and sodium lauryl sulphate were passed through suitable sieve and mixed properly to get uniform blend.
2. The material obtained in step 1 was subjected to roller compaction and granules were prepared.
3. Extragranular L-HPC LH 11 was sieve and mixed with step 2 granules.
4. Step 3 material was lubricated using magnesium stearate and compressed into tablets. Core tablets were coated using Opadry dispersion with inlet temperature of 60-65°C and product temperature of 38-44°C. Table 10- Dissolution data of Lasmiditan hemisuccinate Tablet (Example 12) composition
Figure imgf000019_0001
Example 13
Table 11 - Pharmaceutical composition of Lasmiditan hemisuccinate
Figure imgf000019_0002
Procedure:
1. Lasmiditan hemisuccinate, microcrystalline cellulose (Avicel PH 112), PVPK-30, croscarmellose sodium and sodium lauryl sulphate were passed through suitable sieve and mixed properly to get uniform blend.
2. Magnesium stearate was sieved and mixed with step 1 material. 3. Slugs were prepared using lubricated blend of Step 2.
4. The material obtained in step 3 was milled and granules were obtained.
5. Extragranular microcrystalline cellulose (Avicel PH 112) and croscarmellose sodium were sieved and mixed with step 4 granules.
6. Step 5 material was lubricated using magnesium stearate and compressed into tablets. Core tablets were coated using Opadry dispersion with inlet temperature of 60-65°C and product temperature of 38-44°C.
Example 14
Table 12 - Pharmaceutical composition of Lasmiditan hemisuccinate
Figure imgf000020_0001
Procedure
1. Lasmiditan hemisuccinate, sodium lauryl sulfate, microcrystalline cellulose (Avicel PH 112) and L HPC LH 11/ croscarmellose sodium were passed through suitable sieve and mixed properly to get uniform blend.
2. Step 1 material was granulated with solvent (IPA/Ethanol).
3. Wet mass was dried at suitable temperature until LOD <2% achieved.
4. Dried granules were sifted through #24 sieve and oversized granules were passed through 1mm sieve. Milled granules were sifted through #24 sieve.
5. Sifted and milled granules were mixed in blender for 5 min. 6. Magnesium stearate was mixed with step 5 granules in blender for 5 min.
7. Lubricated blend of step 6 was compressed into tablet at target weight i.e. 230 mg.
8. Core tablets were coated using Opadry dispersion with inlet temperature of 60-65°C and product temperature of 38-44°C.
Table 13- Dissolution data of Lasmiditan hemisuccinate Tablet (Example 14) composition
Figure imgf000021_0001

Claims

CLAIMS:
1. A pharmaceutical composition comprising a therapeutically effective amount of Lasmiditan or a pharmaceutically acceptable salt thereof, at least one pharmaceutical diluent and one or more pharmaceutical acceptable excipients.
2. The pharmaceutical composition as claimed in claim 1, wherein the pharmaceutical composition is prepared by a dry manufacturing process.
3. The pharmaceutical composition as claimed in claim 2, wherein the dry manufacturing process comprises direct compression process or dry granulation process.
4. The pharmaceutical composition as claimed in claim 1, wherein the manufacturing process comprises non-aqueous granulation process.
5. The pharmaceutical composition as claimed in claim 1, wherein one or more pharmaceutical acceptable excipients are selected from disintegrant, binder, surfactant, lubricant and glidant.
6. The pharmaceutical composition as claimed in claim 1, wherein the pharmaceutical composition comprises 35 to 65 % w/w of the at least one pharmaceutical diluent based on the total weight of the composition.
7. The pharmaceutical composition as claimed in claim 6, wherein the pharmaceutical composition comprises 0 to 7 % w/w of the disintegrant, 0 to 7 % w/w of the binder, 0 to 5 % w/w of the surfactant, 0 to 3 % w/w of the lubricant and 0 to 1% w/w of glidant based on the total weight of composition.
PCT/IB2021/059337 2020-10-14 2021-10-12 Pharmaceutical composition of lasmiditan and process of preparation thereof WO2022079591A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN202021044785 2020-10-14
IN202021044785 2020-10-14

Publications (1)

Publication Number Publication Date
WO2022079591A1 true WO2022079591A1 (en) 2022-04-21

Family

ID=81207756

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2021/059337 WO2022079591A1 (en) 2020-10-14 2021-10-12 Pharmaceutical composition of lasmiditan and process of preparation thereof

Country Status (1)

Country Link
WO (1) WO2022079591A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN100352817C (en) * 2002-03-29 2007-12-05 伊莱利利公司 Pyridinoylpiperidines as 5-HTLF agonists
US20130072524A1 (en) * 2010-04-02 2013-03-21 Jean-Francois Carniaux Compositions And Methods Of Synthesis Of Pyridinolypiperidine 5-HT1F Agonists

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN100352817C (en) * 2002-03-29 2007-12-05 伊莱利利公司 Pyridinoylpiperidines as 5-HTLF agonists
US20130072524A1 (en) * 2010-04-02 2013-03-21 Jean-Francois Carniaux Compositions And Methods Of Synthesis Of Pyridinolypiperidine 5-HT1F Agonists

Similar Documents

Publication Publication Date Title
US20210093575A1 (en) Immediate release formulations and dosage forms of gamma-hydroxybutyrate
CA2798178C (en) Immediate release formulations and dosage forms of gamma-hydroxybutyrate
US20070104785A1 (en) Tablets of linezolid form iii and processes for their preparation
EP4076402A1 (en) Dosage form comprising amorphous solid solution of empagliflozin with polymer
US20110189274A1 (en) Stable Pharmaceutical Compositions Of Montelukast Or Its Salts Or Solvates Or Hydrates
WO2017208136A1 (en) Pharmaceutical composition of dapagliflozin co-crystal
US20090155369A1 (en) Pharmaceutical composition containing levodopa, entacapone and carbidopa
WO2016079687A1 (en) Oral pharmaceutical composition of teriflunomide
CN114867469A (en) Oral pharmaceutical composition comprising carbamate compound and preparation method thereof
AU2017309302A1 (en) Solid pharmaceutical composition comprising amorphous sofosbuvir
KR20080011278A (en) Solid oral dosage form of valopi citabine (val-mcyd) and method of preparing it
WO2022079591A1 (en) Pharmaceutical composition of lasmiditan and process of preparation thereof
WO2017037645A1 (en) Stable pharmaceutical formulations of teriflunomide
CA2893480C (en) Pharmaceutical formulation of n-[5-[2-(3,5-dimethoxyphenyl)ethyl]-2h-pyrazol-3-yl]-4-[(3r,5s)-3,5-dimethylpiperazin-1-yl]benzamide
WO2006013444A1 (en) Preparations of stable pharmaceutical compositions of nateglinide and processes for their preparation
WO2020003196A1 (en) Pharmaceutical composition of axitinib
EP2801352B1 (en) Orally disintegrating formulations of Lacosamid
EP4091604B1 (en) Granules containing posaconazole
WO2023227997A1 (en) Pharmaceutical composition containing combination of azilsartan and chlorthalidone and process of preparation thereof
WO2023073513A1 (en) Pharmaceutical composition containing combination of meloxicam and rizatriptan and process of preparation thereof
WO2005016315A1 (en) Pharmaceutical compositions of nateglinide and a high amount of a water-soluble filler
WO2019138346A1 (en) Pharmaceutical composition of osimertinib
WO2022029798A1 (en) Pharmaceutical compositions comprising ribociclib
WO2022173402A1 (en) A film coated tablet comprising naltrexone hydrochloride
WO2024068758A1 (en) Palatable orodispersible formulation of vortioxetine

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: 21879628

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: 21879628

Country of ref document: EP

Kind code of ref document: A1