ENTERIC COATED PITOLISANT FORMULATIONS AND METHODS OF USE

Abstract
The present disclosure relates generally to dosage forms (e.g., oral dosage forms) and pharmaceutical compositions comprising a pharmaceutically active agent comprising pitolisant or a pharmaceutically acceptable salt, solvate, or hydrate thereof, an enteric coating, and optionally one or more pharmaceutically acceptable excipients; as well as methods of using the dosage forms and pharmaceutical compositions, and methods of making the same.p
Description
BACKGROUND

Pitolisant formulations are useful to treat many diseases and disorders, particularly sleep disorders such as excessive daytime sleepiness (EDS) and cataplexy. For example, WAKIX® (pitolisant monohydrochloride) is a prescription medicine approved for the treatment of EDS or cataplexy in adults with narcolepsy.


Existing pitolisant formulations are usually administered with a titrated dose in the first two weeks, beginning with around 8.9 mg/day that is doubled after one week, and then doubled again after a second week to reach a dose of 35.6 mg/day, which may be lowered depending on the patient's tolerance.


SUMMARY

The present disclosure relates generally to dosage forms (e.g., oral dosage forms) and pharmaceutical compositions comprising a pharmaceutically active agent comprising pitolisant, which is represented by the structure of Formula (I):




embedded image


or a pharmaceutically acceptable salt, solvate, or hydrate thereof, an enteric coating (also referred to as a delayed-release coating), and optionally one or more pharmaceutically acceptable excipients. Also disclosed are methods of using the dosage forms and pharmaceutical compositions (e.g., for treating a disease or disorder), as well as methods of making the same. The dosage form may further comprise an anti-moisture barrier.


An advantage of the dosage forms and pharmaceutical compositions of the present disclosure is that they can have reduced or no gastric side-effects, such as nausea, emesis, and upset stomach, when administered (e.g., orally). For example, a subject that receives by oral administration a dosage form or pharmaceutical composition disclosed herein may experience minimal or no side-effects such as nausea, upset stomach, or emesis, relative to the when the subject is orally administered an equivalent non-enterically coated dosage form or pharmaceutical composition, such as WAKIX®.


Another advantage of the dosage forms and pharmaceutical compositions of the present disclosure is they can result in improved patient compliance in the subjects receiving them, relative to patient compliance typically seen in subjects receiving a non-enterically coated dosage forms or pharmaceutical compositions, such as WAKIX®. This is due in part to the reduced side-effects experienced by subjects receiving the dosage forms or pharmaceutical compositions of the present disclosure, e.g., following oral administration. As the side-effects experienced by subjects receiving the dosage forms or pharmaceutical compositions disclosed herein may be minimal or non-existent, subjects are more likely to comply with the titration period and/or prescribed dosage regimen, and will be more likely to continue at the maximum recommended dosage, therefore receiving full clinical benefit with minimal or no side-effects.


Without wishing to be bound by theory, it is believed that the dosage forms and pharmaceutical compositions disclosed herein are bioequivalent to equivalent dosage forms comprising the same pharmaceutically active agent, without an enteric coating, such as WAKIX®. For example, orally administering a dosage form or pharmaceutical composition disclosed herein to a subject can provide a substantially similar (i.e., about the same) exposure of pitolisant compared to when orally administering an equivalent dose of WAKIX®, as determined by measuring the area under the curve (AUC) of pitolisant. Also, orally administering a dosage form or pharmaceutical composition disclosed herein can provide a substantially similar Cmax of pitolisant, compared to when orally administering an equivalent dose of WAKIX®. Further, orally administering a dosage form or pharmaceutical composition disclosed herein can provide a substantially similar Tmax of pitolisant, compared to when orally administering an equivalent dose of WAKIX®. This is surprising, as dosage forms comprising an enteric coating are normally expected to have a relatively delayed release of the pharmaceutically active agents, compared to their non-enterically coated counterparts, which is expected to result in non-bioequivalence e.g., due to delayed absorption of the pharmaceutically active agent and/or altered pharmacokinetics such as a blunted Cmax, longer Tmax, or altered AUC. Yet, dosage forms and pharmaceutical compositions of the present disclosure comprise an enteric coating and can be substantially bioequivalent to their non-enterically coated counterparts, such as WAKIX®, e.g., they do not exhibit substantially different Cmax, Tmax, and/or exposure as determined by AUC.


In some aspects the present disclosure relates to an oral dosage form comprising: a core; and an enteric coating that surrounds the core, wherein the core comprises pitolisant or a pharmaceutically acceptable salt, solvate, or hydrate thereof; and optionally one or more pharmaceutically acceptable excipients. The core may comprise pitolisant monohydrochloride (e.g., crystalline pitolisant monohydrochloride).


The enteric coating can comprise a polymer. The polymer may comprise an ionizable functional group, such as a carboxylic acid group. The enteric coating can comprise a polymer such as a cellulosic material (e.g., an alkylcellulose), an acrylic polymer, an acrylate polymer, a methacrylic polymer, a methacrylate polymer, or a methacrylate copolymer (e.g., an anionic methacrylate copolymer). For example, the enteric coating may comprise a EUDRAGIT® polymer, such as e.g., EUDRAGIT® L 100-55, or the enteric coating may comprise ACRYL-EZE®. It will be understood that an enteric coating may also be referred to as a delayed-release coating.


The enteric coating may further comprise a plasticizer, such as polyethylene glycol (PEG), e.g., PEG8000, or triethyl citrate.


The dosage forms of the present disclosure may comprise an anti-moisture barrier. The anti-moisture barrier can be positioned between the core and the enteric coating. For example, the dosage form may comprise a core tablet that is coated with the anti-moisture barrier (to give an anti-moisture barrier coated tablet), which itself is further coated with an enteric coating (to give an enteric-coated tablet). The anti-moisture barrier may comprise a polymer, such as polyvinyl alcohol (PVA) or a cellulose-based polymer such as HPMC. The anti-moisture barrier may comprise an OPADRY® polymer, such as OPADRY® amb II (e.g., purple OPADRY® amb II, or clear OPADRY® amb II).


The dosage form can comprise between about 1 mg and about 25 mg pitolisant or a pharmaceutically acceptable salt, solvate, or hydrate thereof (e.g., between about 3 mg and about 7 mg, or between about 18 mg and about 22 mg). For example, the dosage form can comprise about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 7 mg, about 8 mg, about 9 mg, about 10 mg, about 11 mg, about 12 mg, about 13 mg, about 14 mg, about 15 mg, about 16 mg, about 17 mg, about 18 mg, about 19 mg, about 20 mg, about 21 mg, about 22 mg, about 23 mg, about 24 mg, or about 25 mg of pitolisant or a pharmaceutically acceptable salt, solvate, or hydrate thereof. The pitolisant or pharmaceutically acceptable salt, solvate, or hydrate thereof, may be contained within the core of the dose form, and may not be present in the enteric coating or in the anti-moisture barrier. For example, a dosage form of the present disclosure may comprise about 5 mg of pitolisant or a pharmaceutically acceptable salt, solvate, or hydrate thereof (e.g., 5 mg of pitolisant monohydrochloride). For example, a dosage form of the present disclosure may comprise about 20 mg of pitolisant or a pharmaceutically acceptable salt, solvate, or hydrate thereof (e.g., 20 mg of pitolisant monohydrochloride).


The dosage form can comprise a pharmaceutically acceptable excipient. For example, the pharmaceutically acceptable excipient may be microcrystalline cellulose, crospovidone, talc, magnesium stearate, or colloidal silica (e.g., anhydrous colloidal silica). For example, a dosage form of the present disclosure can comprise a core, wherein the core comprises one or more of microcrystalline cellulose, crospovidone, talc, magnesium stearate, or colloidal silica (e.g., anhydrous colloidal silica).


The dosage form of the present disclosure can comprise crystalline pitolisant monohydrochloride. The dosage form may further comprise a pharmaceutically acceptable excipient. In some embodiments, the dosage form comprises a core, wherein the core comprises (e.g., consists essentially of, e.g., consists of) pitolisant monohydrochloride, microcrystalline cellulose, crospovidone, talc, magnesium stearate, and colloidal silica (e.g., anhydrous colloidal silica).


The core of the dosage form disclosed herein can be surrounded by an anti-moisture barrier that comprises (e.g., consists essentially of, e.g., consists of) a PVA-based polymer, e.g., OPADRY® amb II. The core surrounded by the anti-moisture barrier may further be surrounded by an enteric coating that comprises (e.g., consists essentially of, e.g., consists of) a copolymer of methacrylic acid and ethyl acrylate, e.g., EUDRAGIT® L 100-55 or ACRYL-EZE®.


The pitolisant or pharmaceutically acceptable salt, solvate, or hydrate thereof included in a dosage form of the present disclosure can have an X-ray diffractogram that comprises characteristic peaks (2θ) at 11.2°, 19.9°, 20.7°, and 34.1° (±0.2°). For example, the pitolisant or pharmaceutically acceptable salt, solvate, or hydrate thereof can have an X-ray diffractogram that comprises characteristic peaks (2θ) at 11.2°, 15.4°, 16.3°, 16.9°, 17.8°, 19.9°, 20.7°, 21.0°, 21.8°, 22.6°, 24.5°, 24.6°, 25.0°, 25.5°, 26.3°, 28.3°, 30.3°, 34.1°, 35.8°, 40.0°, and 46.0° (±0.2°). An exemplary X-ray diffractogram is provided in FIG. 1. In some embodiments, the pitolisant or pharmaceutically acceptable salt, solvate, or hydrate thereof present in a dosage form or pharmaceutical composition of the present disclosure has an X-ray diffractogram substantially the same as that provided in FIG. 1.


An oral dosage form of the present disclosure may be a tablet.


A dosage form of the present disclosure may be bioequivalent to a dosage form comprising pitolisant or a pharmaceutically acceptable salt, solvate, or hydrate thereof, in about the same amount, that does not comprise an enteric coating (e.g., WAKIX®).


For example, orally administering a dosage form of the present disclosure to a subject can provide a Cmax of pitolisant in the subject that is substantially the same as the Cmax of pitolisant obtained following orally administering an equivalent dosage form without an enteric coating (e.g., WAKIX®) to the subject. As another example, orally administering a dosage form of the present disclosure once daily for a period of about 7 days can provide a steady state Cmax of pitolisant that is substantially the same as the steady state Cmax of pitolisant following orally administering an equivalent dosage form without an enteric coating (e.g., WAKIX®) once daily for a period of about 7 days.


Orally administering a dosage form of the present disclosure to a subject can provide an AUC of pitolisant that is substantially the same as the AUC of pitolisant obtained following orally administering an equivalent dosage form without an enteric coating (e.g., WAKIX®) to a subject. As another example, orally administering a dosage form of the present disclosure once daily for a period of about 7 days can provide a steady state AUC of pitolisant that is substantially the same as the steady state AUC of pitolisant following orally administering an equivalent dosage form without an enteric coating (e.g., WAKIX®) once daily for a period of about 7 days.


A dosage form of the present disclosure, when orally administered to a subject, can provide a Tmax of pitolisant that is substantially the same as the Tmax of pitolisant obtained following orally administering an equivalent dosage form without an enteric coating (e.g., WAKIX®).


The present disclosure also relates to methods of treating a disease or disorder, comprising orally administering to a subject in need thereof an oral dosage form of the present disclosure. The disease or disorder can be a sleep disorder. For example, the disease or disorder can be excessive daytime sleepiness (EDS), cataplexy, narcolepsy, sleep apnea (e.g., obstructive sleep apnea), sleep induced apnea, or diurnal somnolence. In some embodiments, the disease or disorder is excessive daytime sleepiness (EDS). In some embodiments, the disease or disorder is cataplexy. In some embodiments, the method involves treating a disease or disorder in a subject that has narcolepsy (e.g., an adult with narcolepsy).


The present disclosure further relates to a dosage form (e.g., oral dosage form) described herein for use in the treatment of a disease or disorder, optionally, wherein the disease or disorder is excessive daytime sleepiness (EDS), cataplexy, narcolepsy, sleep apnea (e.g., obstructive sleep apnea), sleep induced apnea, or diurnal somnolence. The disease or disorder may be in a subject with narcolepsy (e.g., an adult subject with narcolepsy).


The present disclosure also relates to use of an oral dosage form described herein for the manufacture of a medicament for the treatment of a disease or disorder, optionally, wherein the disease or disorder is excessive daytime sleepiness (EDS), cataplexy, narcolepsy, sleep apnea (e.g., obstructive sleep apnea), sleep induced apnea, or diurnal somnolence. The disease or disorder may be in a subject with narcolepsy (e.g., an adult subject with narcolepsy).


The present disclosure also relates to a method of making an oral dosage form described herein. The method can comprise the steps of: (a) blending pitolisant or a pharmaceutically acceptable salt, solvate, or hydrate thereof with one or more pharmaceutically acceptable excipients (e.g., microcrystalline cellulose, crospovidone, talc, magnesium stearate, colloidal silica (e.g., anhydrous colloidal silica) or a combination thereof), to provide a blend; (b) tableting the blend (e.g., using a tableting press) to provide a core tablet; (c) optionally, coating the core tablet with an anti-moisture barrier, to provide an anti-moisture coated tablet; and (d) coating the core tablet of step (b) or the anti-moisture barrier coated tablet of step (c) with an enteric coating, to provide an enteric coated tablet.


The anti-moisture barrier optionally applied in step (c) may comprise a polymer, such as polyvinyl alcohol (PVA) or HPMC, e.g., an OPADRY® polymer, e.g., OPADRY® amb II. The enteric coating applied in step (d) may also comprise a polymer, such as a copolymer of methacrylic acid and ethyl acrylate, e.g., EUDRAGIT® L 100-55 or ACRYL-EZE®. The enteric coating applied in step (d) may also comprise a plasticizer, such as triethyl citrate or PEG (e.g., PEG8000).





BRIEF DESCRIPTION OF DRAWINGS


FIG. 1 is an X-ray diffraction pattern of a polymorph of crystalline pitolisant monohydrochloride suitable for use in a dosage form of the present disclosure.



FIG. 2 is a plot depicting mass evolution during compression of Batch 1 core tablets.



FIG. 3 is a plot depicting hardness evolution during compression of Batch 1 core tablets.



FIG. 4 is an overlay of plots depicting stability dissolution profiles (means) of Batch 1-1 (10% GR), Batch 1-2 (12% GR), and Batch 1-3 (15% GR), each of which are exemplary enteric-coated dosage forms containing 20 mg pitolisant HCl, in baskets at 100 rpm.



FIG. 5 is an overlay of plots depicting stability dissolution profiles (means) of Batches 3-6, each of which are exemplary enteric-coated dosage forms containing 20 mg pitolisant HCl, in baskets at 100 rpm.



FIG. 6 is an overlay of plots depicting stability dissolution profiles (means) of Batch 7-1 (10% GR), Batch 7-2 (13% GR), Batch 7-3 (15% GR), and Batch 7-4 (17% GR), each of which are exemplary enteric-coated dosage forms containing 20 mg pitolisant HCl.



FIG. 7 is an overlay of plots depicting stability dissolution profiles (means) of Batch 8, an exemplary enteric-coated dosage form containing 20 mg pitolisant HCl, at release and after 1 month at 5° C., 25° C./60% RH, or 40° C./75% RH.



FIGS. 8A and 8B are plots showing the dissolution profile of an exemplary enteric-coated dosage form (clinical batch LC23120A) in two sub-lots: sub-lot 1 (FIG. 8A) and sub-lot 2 (FIG. 8B).



FIG. 9 is a plot showing the dissolution profile of an exemplary enteric-coated dosage form (clinical batch LC23120A) at release.



FIG. 10 is a plot showing the dissolution profile of an exemplary enteric-coated dosage form after 3 months under different stability testing conditions, compared with the same batch at release (clinical batch LC23120A).





DETAILED DESCRIPTION

The present disclosure relates generally to dosage forms (e.g., oral dosage forms) and pharmaceutical compositions comprising a pharmaceutically active agent comprising pitolisant, which is represented by Formula (I):




embedded image


or a pharmaceutically acceptable salt, solvate, or hydrate thereof, an enteric coating, and optionally one or more pharmaceutically acceptable excipients, and optionally an anti-moisture barrier coating.


The dosage forms and pharmaceutical compositions of the present disclosure, which comprise an enteric coating, have many advantages over existing pitolisant formulations such as WAKIX®. In particular, they can be tolerated more by subjects, owing to less severe or no side-effects (e.g., gastric side effects, such as stomach upset, nausea, and emesis), relative to non-enterically coated pitolisant dosage forms or pharmaceutical compositions, such as WAKIX®. Consequently, subjects receiving the dosage forms and pharmaceutical compositions of the present disclosure demonstrate better patient compliance relative to subjects receiving pitolisant formulations that do not comprise an enteric coating (e.g., WAKIX®). Another advantage of the dosage forms and pharmaceutical compositions of the present disclosure is that they are bioequivalent to WAKIX®, a pitolisant-containing dosage form approved for the treatment of EDS and cataplexy in adult subjects with narcolepsy, which is unexpected.


Definitions

The articles “a” and “an” are used herein to refer to one or more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element.


The term “about” when referring to a measurable value such as an amount, a temporal duration, and the like, is meant to encompass variations of ±20% or less, or in some instances±15% or less, or in some instances±10% or less, or in some instances±5% or less, or in some instances±1% or less, or in some instances±0.1% or less, from the specified value, as such variations are appropriate.


The phrase “and/or” as used herein should be understood to mean “either or both” of the elements so conjoined, i.e., elements that are conjunctively present in some cases and disjunctively present in other cases. Multiple elements listed with “and/or” should be construed in the same fashion, i.e., “one or more” of the elements so conjoined. Other elements may optionally be present other than the elements specifically identified by the “and/or” clause, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, a reference to “A and/or B,” when used in conjunction with open-ended language such as “comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.


The terms “administer,” “administering,” or “administration,” as used herein refer to implanting, absorbing, ingesting, injecting, inhaling, or otherwise introducing a compound, dosage form, or pharmaceutical composition.


The terms “comprise,” “comprises,” and “comprising” are used herein in a non-exclusive sense, except where the context requires otherwise. Likewise, the term “include” and its grammatical variants are intended to be non-limiting, such that recitation of items in a list is not to the exclusion of other like items that can be substituted or added to the listed items.


The term “effective amount” or a “therapeutically effective amount” as used herein refers to an amount of a compound, a dosage form, or a pharmaceutical composition, described herein which is sufficient to achieve a desired result under the conditions of administration. For example, an effective amount of a compound, dosage form, or pharmaceutical composition disclosed herein for treating excessive sleep disorder (EDS), e.g., in a subject with narcolepsy, is an amount that can reduce the effects of the EDS, and/or reduce or eliminate the severity of a symptom associated with the EDS. A skilled clinician can determine appropriate dosing based on a variety of considerations including the severity of the disease, the subject's age, weight, general health and other considerations. A dosage form or pharmaceutical composition disclosed herein may be administered to provide an amount of about 0.01 mg to about 250 mg (e.g., about 0.1 mg to about 100 mg) of a pharmaceutically active agent (e.g., pitolisant or a pharmaceutically acceptable salt, solvate, or hydrate thereof), e.g., about 1 mg, about 2 mg, about 3 mg, about 4 mg, about 5 mg, about 10 mg, about 15 mg, about 20 mg, about 30 mg, or about 40 mg. In some aspects, a dosage form or pharmaceutical composition of the present disclosure is administered to provide about 5 mg pitolisant hydrochloride. In some aspects, a dosage form or pharmaceutical composition of the present disclosure is administered to provide about 20 mg pitolisant hydrochloride.


The term “pharmaceutically acceptable excipient” as used herein refers to a non-toxic material that may be formulated with a compound disclosed herein to provide a pharmaceutical composition. Preferably, the pharmaceutically acceptable excipient is inert and does not interfere with the pharmacological activity of a compound which it is formulated with. Pharmaceutically acceptable excipients useful in the manufacture of the pharmaceutical compositions disclosed herein are any of those well known in the art, and include without limitation, diluents, dispersing agents, granulating agents, surface active agents, emulsifiers, disintegrating agents (sometimes referred to herein as disintegrants), binding agents (sometimes referred to herein as binders), flowing agents, preservatives, buffering agents (sometimes referred to herein as buffers), lubricating agents (sometimes referred to herein as lubricants), glidants, adjuvants, fillers, wetting agents, suspending agents, solvents, dispersion media, ion exchangers, salts, electrolytes, waxes, coloring agents, and/or oils, and the like.


For example, a pharmaceutically acceptable excipient may be alumina, a phosphate (e.g., calcium phosphate, dicalcium phosphate, tricalcium phosphate, disodium hydrogen phosphate, potassium hydrogen phosphate), a sulfate (e.g., calcium sulfate), a cellulose (including, e.g., cellulose derivatives, microcrystalline cellulose (including spray-dried microcrystalline cellulose), silicified microcrystalline cellulose, hydroxypropylmethylcellulose, methylcellulose, carboxymethylcellulose or salts thereof (e.g., sodium carboxymethylcellulose, or calcium carboxymethylcellulose)), kaolin, bentonite, VEEGUM®, lactose (e.g., anhydrous lactose, spray dried lactose, hydrated lactose), mannitol, sorbitol, sucrose, inositol, compressible sugar, trehalose, xylitol, acacia, gelatin, glucose, maltodextrin, starch (e.g., corn starch, potato starch), sodium starch glycolate, starch derivatives, an amino acid (e.g., glycine or leucine), magnesium carbonate, polyvinylpyrrolidone (PVP, povidone) (e.g., crosslinked PVP, crospovidone), polyvinyl alcohol, tragacanth, polyethylene glycol, polymethacrylates, mineral clay powders, croscarmellose, poloxamer, fatty acids or salts thereof (e.g., lauric acid, sodium lauryl sulfate, stearic acid, calcium stearate, magnesium stearate, aluminum stearate, oleic acid), hydrogenated vegetable oils, talc, titanium dioxide, glyceryl behenate, silicon dioxide (e.g., colloidal silicon dioxide), a silicate salt (e.g., magnesium trisilicate), lecithin, serum protein (e.g., human serum albumin), sorbic acid, potassium sorbate, a metal cation salt (e.g., a sodium salt, such as sodium chloride, a potassium salt, such as potassium chloride, a magnesium salt, such as or magnesium chloride, a zinc salt, such as zinc chloride), water, dimethylacetamide, protamine sulfate, a polyacrylate, wool fat, ethylenediaminetetraacetic acid (EDTA), a cyclodextrin (e.g., CAPTISOL®), KOLLIDON® CL, CELLACTOSE®, LUDIPRESS®, polysorbates (e.g., TWEEN®, e.g., TWEEN® 20 or TWEEN® 80), and combinations thereof.


The term “pharmaceutically acceptable salt” as used herein refers to salts of a compound prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the respective compound. When compounds relating to the present disclosure contain relatively basic functionalities (e.g., as in Formula (I)), acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable solvent (e.g., an inert solvent). For example, the neutral form of pitolisant may be contacted with gaseous hydrochloric acid to provide pitolisant monohydrochloride, which can be present in a dosage form disclosed herein. Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, monohydrogencarbonic acid, phosphoric acid, monohydrogenphosphoric acid, dihydrogenphosphoric acid, sulfuric acid, monohydrogensulfuric acid, hydriodic acid, or phosphorous acids and the like, as well as the salts derived from organic acids like acetic acid, propionic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, fumaric acid, lactic acid, mandelic acid, pamoic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid, methanesulfonic acid, oxalic acid, and the like. Also included are salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galacturonic acids and the like. Other pharmaceutically acceptable salts known to those of skill in the art are suitable for pharmaceutical compositions relating to the present disclosure.


The term “solvate” as used herein refers to forms of a compound that are associated with a solvent, usually by a solvolysis reaction. This physical association may include hydrogen bonding. Conventional solvents include water, methanol, ethanol, acetic acid, dimethyl sulfoxide (DMSO), tetrahydrofuran (THF), diethyl ether, and the like. Compounds of the present disclosure may be prepared, e.g., in crystalline form, and may be solvated. Suitable solvates include pharmaceutically acceptable solvates and further include both stoichiometric solvates and non-stoichiometric solvates. In certain instances, the solvate will be capable of isolation, for example, when one or more solvent molecules are incorporated in the crystal lattice of a crystalline solid. “Solvate” encompasses both solution-phase and isolable solvates. Representative solvates include hydrates, ethanolates, and methanolates.


The term “hydrate” as used herein refers to a compound which is associated with water. Typically, the number of the water molecules contained in a hydrate of a compound is in a definite ratio to the number of the compound molecules in the hydrate. Therefore, a hydrate of a compound may be represented, for example, by the general formula R·xH2O, wherein R is the compound and wherein x is a number greater than 0. A given compound may form more than one type of hydrate, including, e.g., monohydrates (x is 1), lower hydrates (x is a number greater than 0 and smaller than 1, e.g., hemihydrates (R·0.5H2O)), and polyhydrates (x is a number greater than 1, e.g., dihydrates (R·2H2O) and hexahydrates (R·6H2O)).


The term “subject” as used herein refers to any animal, such as any mammal, including but not limited to, humans, non-human primates, rodents, dogs, and the like. Non-human primates include chimpanzees, cynomolgus monkeys, spider monkeys, and macaques (e.g., Rhesus). Rodents include mice, rats, woodchucks, ferrets, rabbits, and hamsters. Domestic and game animals include cows, horses, pigs, deer, bison, buffalo, feline species (e.g., domestic cat), canine species (e.g., dog, fox, wolf), avian species, and fish. In some embodiments, the subject is a mammal (e.g., a human, a rat, or a mouse). The subject can be male or female. The subject may be of any age, including an elderly human subject (e.g., 65 years or older), a human subject that is not elderly (e.g., less than 65 years old), or a human pediatric subject (e.g., less than 18 years old). In preferred aspects, the subject is a human.


As used herein, the terms “treat,” “treatment,” “treating,” or grammatically related terms, refer to a method of reducing the effects of a disease or disorder. As is readily appreciated in the art, full eradication of the disease, disorder, or symptoms thereof is preferred but not a requirement for treatment. Desirable effects of treatment include, but are not limited to, preventing occurrence or recurrence of the disease or disorder, alleviation of symptoms, diminishment of any direct or indirect pathological consequences of the disease or disorder, or other improvement of any sign, symptom, or consequence of the disease or disorder, such as prolonged survival, less morbidity, and/or a lessening of side effects.


Throughout this disclosure, various embodiments can be presented in a range format (e.g., from X-Y). It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the present disclosure. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 5, from 1 to 4, from 1 to 3, from 2 to 6, from 2 to 4, from 3 to 6, etc., as well as individual numbers within that range, e.g., 1, 2, 2.8, 3, 3.6, 4, 5, 5.4, and 6. As another example, a range such as 95-99% includes 95%, 96%, 97%, 98%, or 99% and all subranges such as 96-99%, 96-98%, 96-97%, 97-99%, 97-98%, etc. This applies regardless of the breadth of the range.


All publications (e.g., scientific journal articles, patent publications, and the like) cited in this disclosure are incorporated by reference in their entirety. To the extent the material incorporated by reference contradicts or is inconsistent with this specification, the specification will supersede any such material. The citation of any references herein is not an admission that such references are prior art to the present disclosure. Various terms relating to aspects of the description are used throughout the specification and claims. Such terms are to be given their ordinary meaning in the art unless otherwise indicated. Other specifically defined terms are to be construed in a manner consistent with the definitions provided herein.


Compounds (e.g., pharmaceutically active agents) disclosed herein may also comprise one or more isotopic substitutions. For example, hydrogen (H) may be in any isotopic form, including 1H, 2H (D or deuterium), 3H (T or tritium); carbon (C) may be in any isotopic form, including 12C, 13C, and 14C; oxygen (O) may be in any isotopic form, including 16O and 18O; nitrogen (N) may be in any isotopic form, including 14N and 15N; and chlorine (Cl) may be in any isotopic form, including 35Cl and 37Cl.


Various embodiments of the compounds, dosage forms, pharmaceutical compositions, and methods herein are described in further detail below, and additional definitions may be provided throughout the specification.


Dosage Forms and Pharmaceutical Compositions

Disclosed herein are dosage forms (e.g., oral dosage forms) and pharmaceutical compositions comprising a pharmaceutically active agent and an enteric coating, and optionally one or more pharmaceutically acceptable excipients. The pharmaceutically active agent is pitolisant, represented by Formula (I):




embedded image


or a pharmaceutically acceptable salt, solvate, or hydrate thereof.


The dosage form or pharmaceutical composition can comprise a therapeutically effective amount of the pharmaceutically active agent. For example, the dosage form or pharmaceutical composition may comprise between about 1 mg and about 200 mg of the pharmaceutically active agent, e.g., between about 1 mg and about 100 mg, between 1 mg and about 80 mg, between 1 mg and about 60 mg, between about 1 mg and about 50 mg, between about 10 mg and about 25 mg, or between about 1 mg and about 10 mg, of the pharmaceutically active agent, e.g., about 3 mg, about 4 mg, about 5 mg, about 6 mg, about 8 mg, about 10 mg, about 12 mg, about 15 mg, about 20 mg, about 25 mg, about 30 mg, about 40 mg, or about 50 mg, of the pharmaceutically active agent.


It will be understood that when the pharmaceutically active agent is a pharmaceutically acceptable salt of Formula (I), the amount of the pharmaceutically active agent in the dosage form or pharmaceutical composition will be slightly higher than the equivalent amount of free base. For example, a dosage form or pharmaceutical composition disclosed herein comprising 5 mg of pitolisant hydrochloride will comprise about 4.45 mg of pitolisant (freebase). In another example, a dosage form or pharmaceutical composition disclosed herein comprising 20 mg of pitolisant hydrochloride comprises about 17.8 mg of pitolisant (freebase). In some embodiments, a dosage form or pharmaceutical composition disclosed herein comprises about 5 mg of pitolisant monohydrochloride, or about 4.45 mg pitolisant (freebase). In some embodiments, a dosage form or pharmaceutical composition disclosed herein comprises about 20 mg of pitolisant monohydrochloride, or about 17.8 mg pitolisant (freebase).


The dosage forms of the present disclosure can be tablets, caplets, capsules, suspensions, granules, powders, or the like. Preferably, the dosage forms of the present disclosure are enteric coated tablets, enteric coated caplets, enteric coated capsules, or the like. The tablet (e.g., enteric coated tablet) may be a round, biconvex tablet. The tablet may be engraved, e.g., with a number, letter(s), or both.


The dosage forms or pharmaceutical compositions of the present disclosure can further comprise one or more pharmaceutically acceptable excipients, such as diluents, dispersing agents, granulating agents, surface active agents, emulsifiers, disintegrating agents (sometimes referred to herein as disintegrants), binding agents (sometimes referred to herein as binders), preservatives, buffering agents, lubricating agents (sometimes referred to herein as lubricants), glidants, adjuvants, fillers, wetting agents, suspending agents, solvents, dispersion media, ion exchangers, salts, electrolytes, waxes, and/or oils, and the like.


Non-limiting examples of pharmaceutically acceptable excipients suitable for use in the dosage forms or pharmaceutical compositions disclosed herein include alumina, a phosphate (e.g., calcium phosphate, dicalcium phosphate, tricalcium phosphate, disodium hydrogen phosphate, potassium hydrogen phosphate), a sulfate (e.g., calcium sulfate), cellulose, cellulose derivatives, microcrystalline cellulose (including spray-dried microcrystalline cellulose), silicified microcrystalline cellulose, hydroxypropylmethylcellulose, methylcellulose, carboxymethylcellulose or salts thereof (e.g., sodium carboxymethylcellulose, or calcium carboxymethylcellulose), kaolin, bentonite, VEEGUM®, lactose (e.g., anhydrous lactose, spray dried lactose, hydrated lactose), mannitol, sorbitol, sucrose, inositol, compressible sugar, trehalose, xylitol, acacia, gelatin, glucose, maltodextrin, starch (e.g., corn starch, potato starch), sodium starch glycolate, starch derivatives, an amino acid (e.g., glycine or leucine), magnesium carbonate, polyvinylpyrrolidone (PVP, povidone) (e.g., crosslinked PVP, crospovidone), polyvinyl alcohol, tragacanth, polyethylene glycol, polymethacrylates, mineral clay powders, croscarmellose, poloxamer, fatty acids or salts thereof (e.g., lauric acid, sodium lauryl sulfate, stearic acid, calcium stearate, magnesium stearate, aluminum stearate, oleic acid), hydrogenated vegetable oils, talc, titanium dioxide, glyceryl behenate, silicon dioxide (e.g., colloidal silicon dioxide), a silicate salt (e.g., magnesium trisilicate), lecithin, serum protein (e.g., human serum albumin), sorbic acid, potassium sorbate, a metal cation salt (e.g., a sodium salt, such as sodium chloride, a potassium salt, such as potassium chloride, a magnesium salt, such as or magnesium chloride, a zinc salt, such as zinc chloride), water, dimethylacetamide, protamine sulfate, a polyacrylate, wool fat, ethylenediaminetetraacetic acid (EDTA), a cyclodextrin (e.g., CAPTISOL®), KOLLIDON® CL, CELLACTOSE®, LUDIPRESS®, polysorbates (e.g., TWEEN®, e.g., TWEEN® 20 or TWEEN® 80), and combinations thereof.


In a preferred aspect, a dosage form (e.g., oral dosage form) or pharmaceutical composition of the present disclosure comprises a combination of pharmaceutically acceptable excipients. For example, a dosage form or pharmaceutical composition of the present disclosure may comprise one or more, or all of, the following pharmaceutically acceptable excipients: microcrystalline cellulose (e.g., Vivapur 102), crospovidone (e.g., Polyplasdone XL), talc, magnesium stearate (e.g., Kemilub EM-F-V), and colloidal silica. The pharmaceutically acceptable excipient may be present only in the core of the dosage form.


Each pharmaceutically acceptable excipient can be present in the dosage form or pharmaceutical composition in any suitable amount. For example, a pharmaceutically acceptable excipient can be present in the dosage form or pharmaceutical composition in an amount of between about 1% and about 95% by weight of the dosage form or pharmaceutical composition, e.g., between about 60% to 90%, or between about 80% to about 90%, by weight of the dosage form or pharmaceutical composition.


For example, the pitolisant may be present in the core of the dosage form in an amount of about 14 wt % to about 18 wt % (e.g., about 16 wt %), and the pharmaceutically acceptable excipients combined may account for the remainder of the weight of the core of the dosage form. One pharmaceutically acceptable excipient may be a diluent (e.g., microcrystalline cellulose) that is present in the core of the dosage form in an amount of about 60% to about 70% by weight (e.g., about 65 wt %). Another pharmaceutically acceptable excipient may be a disintegrant (e.g., crospovidone) that is present in the core of the dosage form in an amount of about 6% to about 10% by weight (e.g., about 8 wt %). Another pharmaceutically acceptable excipient may be a glidant (e.g., talc) that is present in the core of the dosage form in an amount of about 6% to about 10% by weight (e.g., about 8 wt %). Another pharmaceutically acceptable excipient may be a lubricant (e.g., magnesium stearate) that is present in the core of the dosage form in an amount of about 1% to about 4% by weight (e.g., about 2.4 wt %). Another pharmaceutically acceptable excipient may be a flowing agent (e.g., colloidal silica) that is present in the core of the dosage form in an amount of about 0.1% to about 2% by weight (e.g., about 0.8 wt %).


Preferably, the dosage forms or pharmaceutical compositions of the present disclosure are administered orally.


Preparation of the dosage forms (e.g., oral dosage forms) or pharmaceutical composition of the present disclosure can include conventional methods, such as blending, filling, compressing (e.g., direct compression, compression of dry, wet or sintered granules), coating (e.g., coating in a spray process), extrusion, granulation (e.g., wet or dry granulation), pelleting (e.g., direct pelleting), binding, powder layering (e.g., onto active ingredient-free beads, or neutral cores or particles of pharmaceutically active agent), and rounding off.


For example, a dosage form of the present disclosure may be prepared with following steps: (a) preparing a core blend comprising pitolisant, and optionally one or more pharmaceutically acceptable excipients, e.g., by blending the pharmaceutically active agent (e.g., crystalline powder) with one or more pharmaceutically acceptable excipients (e.g., binder, disintegrant, glidant, etc.). Additional sieving steps may be employed. The next step may comprise (b) forming a core tablet with the blend, e.g., by tableting using a press. Optionally, the core tablet may be coated with an anti-moisture barrier coating disclosed herein, e.g., using a spray process. Finally, the core tablet, which may be coated with an anti-moisture barrier, can be further coated with an enteric coating material disclosed herein, e.g., using a spray process. The coating steps may comprise spraying the anti-moisture barrier material or the enteric coated material as a solution, suspension, or dispersion (e.g., an organic solution, or aqueous dispersion).


Enteric Coating

The dosage forms (e.g., oral dosage forms) and pharmaceutical compositions disclosed herein comprise an enteric coating. The enteric coating may be a material that is pH-dependent. For example, a dosage form or pharmaceutical composition disclosed herein may remain substantially intact in the stomach where the pH is low (about 1.0 to 3.5), and release the pharmaceutically active agent after entering the small intestine, due to disintegration of the enteric coating in the relatively higher pH of the small intestine (about 5.5 to 7.0). In some aspects of the present disclosure, the enteric coating may disintegrate at a higher pH, e.g., to substantially release the pharmaceutically active agent in more distal portions of the small intestine where the pH is even higher (e.g., about 6.0 to 7.5). A pharmaceutical composition or dosage form of the present disclosure may substantially release the pharmaceutically active agent in the duodenum, in the jejenum, or in both the duodenum and jejenum.


The enteric coating can be substantially resistant to disintegration in the stomach or gastric fluid, e.g., at a pH of between about 1.0 and about 3.5. The enteric coating can disintegrate in the intestine (e.g., small intestine) to expose the pharmaceutically active agent, e.g., after the dosage form or pharmaceutical composition has entered the intestine. To that end, the enteric coating may substantially disintegrate in the intestine (e.g., small intestine) or intestinal fluid, or at a pH of greater than 5 (e.g., a pH of greater than 5.5, greater than 6.0, greater than 6.5, greater than 7.0, or greater than 7.5).


An enteric coating of a dosage form or pharmaceutical composition of the present disclosure may be acid resistant (gastric resistant) and prevent more than 10 percent of the pharmaceutically active agent to be released when the dosage form is in the stomach, in a hydrochloride solution (pH 1.2), in gastric fluid, or simulated gastric fluid, for at least 1 hour (e.g., 2 hours), e.g., releasing less than 10%, less than 8%, less than 6%, less than 4% less than 3%, less than 2%, less than 1% by weight, or none of the pharmaceutically active agent. The hydrochloride solution, gastric fluid, or simulated gastric fluid may be at a temperature of about 37° C. The dosage form or pharmaceutical composition, or the enteric coating thereof, may substantially disintegrate after the dosage form or pharmaceutical composition is in the intestine (e.g., small intestine), or is exposed to intestinal fluid, simulated intestinal fluid, or a solution, e.g., buffer (e.g., phosphate buffer), with a pH of greater than 5 (e.g., a pH of greater than 5.5, 6.0, 6.5, 7.0, 7.5, or higher).


A dosage form or pharmaceutical composition according to the present disclosure may be a delayed release dosage form or pharmaceutical composition according to the United States Pharmacopeia (USP) 701 Procedure and Criteria for Delayed-Release Tablets and Capsules (tablets or capsules that are formulated with acid-resistant or enteric coatings).


An enteric coating of a dosage form or pharmaceutical composition disclosed herein may comprise a polymer. For example, the polymer of the enteric coating may be a polymer comprising an ionizable functional group (e.g., a carboxylic acid group), e.g., a functional group that is not ionized in the stomach or gastric fluid, or in a solution with a pH of between about 1.0 and about 3.5. The functional group may be a group that is ionized in the intestine (e.g., small intestine) or intestinal fluid, or in a solution with a pH of greater than 5 (e.g., a pH of greater than 5.5, greater than 6.0, greater than 6.5, greater than 7.0, or greater than 7.5).


The enteric coating may comprise an acrylic polymer, such as acrylic acid and methacrylic acid copolymers, methacrylate copolymers (e.g., copolymers of methyl methacrylate, butyl methacrylate and dimethylethyl methacrylate, copolymers of methyl methacrylate, ethyl acrylate and trimethylammoniumethyl methacrylate, copolymers of methyl methacrylate and ethyl acrylate); a cellulosic material, such as an alkylcellulose, e.g., ethylcellulose, AQUACOAT®, SURELEASE®, hydroxypropyl methylcellulose phthalate, hydroxypropyl methylcellulose acetate succinate, cellulose acetate trimellitate, cellulose acetate phthalate, or the like; polyvinylpyrrolidones (PVP), polyvinyl alcohols, polyvinyl alcohol-polyethylene glycol graft copolymers, a KOLLICOAT® polymer, starch, starch derivatives, polyvinyl acetate (PVAc), polyvinyl acetate phthalate (PVAP, or COATERIC®), a KOLLIDON® polymer, vinyl acetate-vinylpyrrolidone copolymer (e.g., KOLLIDON® VA64), vinyl acetate: crotonic acid copolymer (VAC:CRA), polyethylene glycols (e.g., polyethylene glycols with a molecular weight above 1000 g/mol), chitosan, crosslinked and/or non-crosslinked polyacrylic acid, sodium alginate, pectin, methacrylic acid ester copolymers, ethoxyethyl methacrylates, cyanoethyl methacrylate, poly(methacrylic acid), methacrylic acid alkylamide copolymer, poly(methyl methacrylate), polymethacrylates, poly(methyl methacrylate) copolymer, polyacrylamide, aminoalkyl methacrylate copolymer, poly(methacrylic acid anhydride), glycidyl methacrylate copolymers, ammonio methacrylate copolymers, or the like; shellac, zein, or the like; a combination thereof. In some embodiments, the enteric coating comprises an anionic polymer, e.g., an anionic methacrylate copolymer. It will be understood that the polymer may not be anionic at sufficiently low pH (e.g., in gastric fluid) but becomes ionized upon increasing pH (e.g., in intestinal fluid).


The enteric coating may comprise a EUDRAGIT® polymer, such as a EUDRAGIT® L polymer, e.g., EUDRAGIT® L 30 D-55, EUDRAGIT® L 100-55, EUDRAGIT® FL 30 D-55, EUDRAGIT® L 100, EUDRAGIT® L 12,5, or a EUDRAGIT® S polymer such as EUDRAGIT® S 100, EUDRAGIT® S 12,5, EUDRAGIT® FS 30 D, EUDRAGIT® FS 100, a EUDRAGIT® RL polymer such as EUDRAGIT® RL PO, EUDRAGIT® RL 100, EUDRAGIT® RL 30D, EUDRAGIT® RL 12,5, a EUDRAGIT® RS polymer such as EUDRAGIT® RS PO, EUDRAGIT® RS 100, EUDRAGIT® RS 30 D, or EUDRAGIT® RS 12,5, a EUDRAGIT® NM polymer such as EUDRAGIT® NM 30, or a combination thereof.


The enteric coating may comprise ACRYL-EZE®, such as ACRYL-EZE® clear.


The enteric coating may also comprise a plasticizer, a stabilizer (e.g., oleic acid, or a polysorbate e.g., TWEEN®80), a lubricant, a glidant, a release agent, or a pigment (e.g., talc, titanium dioxide, or magnesium stearate). Examples of suitable plasticizers include sebacates (e.g., dibutyl sebacate), propylene glycol, polyethylene glycols (e.g., PEG8000), phthalates (e.g., diethyl phthalate or dibutyl phthalate) or phthalate esters, citric acid esters (e.g., triethyl citrate, tibutyl citrate), triacetin, acetylated monoglycerides, castor oil, propylene glycol, and polyethylene glycols. Examples of release agents include glycerol monostearate or other suitable fatty acid derivatives, silica derivatives, and talc. Preferred stabilizers are PEG8000 and triethyl citrate.


The enteric coating may comprise a material, or may be an enteric coating, disclosed in US 2004/0028737, US 2005/0271778, WO 2005/044240, WO 2007/006353, WO 2008/135090, each of which are incorporated herein by reference in their entireties.


Anti-Moisture Barrier

The dosage forms (e.g., oral dosage forms) and pharmaceutical compositions disclosed herein can comprise an anti-moisture barrier coating. The anti-moisture barrier is distinct from the enteric coating, and the anti-moisture barrier can be present in the dosage form or pharmaceutical composition in addition to the enteric coating. For example, the anti-moisture barrier coating may be positioned around the core of a dose form and underneath the enteric coating layer (i.e., between the outer surface of the core and the inner surface of the enteric coating layer). The anti-moisture barrier may comprise any suitable material, such as material that prevents moisture from the environment readily entering the core of the dose form prior to its intended dissolution (e.g., upon administration.


The anti-moisture barrier may comprise a polymer. Suitable polymers include polyvinyl alchohols and hypromellose (hydroxypropylmethylcellulose). An exemplary anti-moisture barrier material of the present disclosure is OPADRY® amb II (e.g., purple OPADRY® amb II, or clear OPADRY® amb II).


Pharmaceutically Active Agent

The pharmaceutically active agent of the dosage form (e.g., oral dosage form) or pharmaceutical composition disclosed herein is pitolisant (1-{3-[3-(4-chlorophenyl) propoxy]propyl}-piperidine), or a pharmaceutically acceptable salt, solvate, or hydrate thereof. Pitolisant is represented by Formula (I):




embedded image


The pharmaceutically active agent may be Formula (I) in freebase form, or may be any pharmaceutically acceptable salt of Formula (I), e.g., a pharmaceutically acceptable salt disclosed herein. For example, the pharmaceutically active agent can be a hydrochloride salt or an oxalate salt of Formula (I).


In preferred aspects, the pharmaceutically active agent is a hydrochloride salt of Formula (I), e.g., (1-{3-[3-(4-chlorophenyl) propoxy]propyl}-piperidine monohydrochloride; pitolisant monohydrochloride), represented by Formula (Ia):




embedded image


Without wishing to be bound by theory, it is believed that pitolisant can activate receptors within the stomach, such as histamine H3 receptors, which may cause gastric side effects associated with existing pitolisant formulations such as WAKIX®. The inventors discovered that by formulating pitolisant dosage forms or pharmaceutical compositions with an enteric coating, the gastric side effects can be substantially reduced or eliminated. Without wishing to be bound by theory, it is also believed that despite the enteric coating, which prevents release of pitolisant from the dosage form in the stomach or gastric fluid, there is not a blunting of Cmax or Tmax, relative to comparative formulations such as WAKIX®, and the overall exposure (as measured by AUC) is relatively the same between the dosage forms of the present disclosure and WAKIX®. In other words, despite their enteric coatings, the dosage forms and pharmaceutical compositions disclosed herein may be substantially bioequivalent to WAKIX®.


The dosage forms and pharmaceutical compositions disclosed herein may comprise the monohydrochloride salt of Formula (I), which has a relatively high aqueous solubility, e.g., compared to other known pharmaceutically acceptable salts of Formula (I) (e.g., pitolisant oxalate). The aqueous solubility of the pharmaceutically active agent may be about 4 g/mL at 23° C.


The pharmaceutically active agent (e.g., Formula (I), or Formula (Ia)) may be crystalline.


The pharmaceutically active agent can have a water content of 6% (±0.5%) or less, e.g., less than 5%, less than 4%, less than 3%, less than 2%, or less than 1%, by weight.


The pharmaceutically active agent can have characteristic IR peaks at 1112, 1101, 2936, 2868, 1455, 2647, 2551, 1492, and 802 cm−1 (±5).


The pharmaceutically active agent can be a crystalline form of pitolisant hydrochloride.


The pharmaceutically active agent can be the crystalline form of pitolisant hydrochloride disclosed in U.S. Pat. No. 8,207,197, which is incorporated herein by reference in its entirety.


The pharmaceutically active agent can be pitolisant hydrochloride that has an X-ray powder diffraction pattern with characteristic peaks (2θ) at: 11.2°, 19.9°, 20.7° and) 34.1° (±0.2°), as obtained using X-Ray diffraction technique described in U.S. Pat. No. 8,207,197, using a Nonius Kappa Charge Coupled Device system at −158° C., operating at a wavelength of 0.71073 Å. The pharmaceutically active agent can have an X-ray powder diffraction pattern as shown in FIG. 1.


The pharmaceutically active agent may be a compound disclosed in U.S. Pat. Nos. 8,207,197, 8,354,430, 8,486,947, 7,138,413, 7,910,605, or U.S. Pat. No. 7,169,928, each of which are incorporated herein by reference in their entireties.


Pharmacokinetics

Without wishing to be bound by theory, it is believed that the oral dosage forms and pharmaceutical compositions disclosed herein are substantially bioequivalent and/or have the substantially same bioavailability as their non-enterically coated counterparts, such as WAKIX®. In other words, and without wishing to be bound by theory, it is believed that the presence of the enteric coating of the dosage forms or pharmaceutical compositions disclosed herein does not substantially affect their pharmacokinetics and/or bioavailability, and following administration, a similar biological effect, bioavailability, and/or pharmacokinetics are achieved as would be expected by administering an equivalent dosage form or pharmaceutical composition without an enteric coating (e.g., containing the same pharmaceutically active agent and pharmaceutically acceptable excipient(s), in substantially the same amounts), such as WAKIX®.


For example, it is believed that oral administration of one or more dosage forms or pharmaceutical compositions disclosed herein to provide about 35.6 mg/day of pitolisant (freebase), can achieve a steady state Cmax of pitolisant of between about 49.2 ng/ml and about 126 ng/ml, e.g., about 73 ng/mL. Similarly, it is believed that oral administration of one or more dosage forms or pharmaceutical compositions disclosed herein to provide about 35.6 mg/day of pitolisant (freebase), can achieve a steady state AUC of pitolisant of between about 518 ng*hr/mL and about 1468 ng*hr/mL, e.g., about 812 ng*hr/mL. It is further believed that steady state AUC and Cmax is reached by day 7, following once daily oral administration of one or more dosage forms or pharmaceutical compositions disclosed herein.


The dosage forms (e.g., oral dosage forms) or pharmaceutical compositions disclosed herein can have substantially the same oral bioavailability as an equivalent dosage form or pharmaceutical composition without the enteric coating (e.g., WAKIX®). For example, without wishing to be bound by theory, it is believed that administering (e.g., orally administering) a dosage form or pharmaceutical composition disclosed herein can achieve an oral absorption of greater than 50%, e.g., about 75%, about 85%, about 90%, or more. It is further believed that administering (e.g., orally administering) a dosage form or pharmaceutical composition disclosed herein can achieve a Tmax of pitolisant of between about 2 hours and about 5 hours (e.g., about 3.5 hours).


In some embodiments, the rate and/or extent of absorption of pitolisant following administration of a dosage form or pharmaceutical composition disclosed herein does not show a significant difference when compared to administering the same molar dose of the therapeutic in a dosage form that does not comprise an enteric coating, such as WAKIX®.


In some embodiments, an oral dosage form or pharmaceutical composition disclosed herein is bioequivalent to WAKIX®, as determined by the Bioequivalence Guidance 94D-0401 provided by the U.S. Food and Drug Administration (FDA).


In some embodiments, an oral dosage form or pharmaceutical composition disclosed herein has substantially the same bioavailability as WAKIX®.


Methods of Treatment

The present disclosure further relates to a method for the treatment of a disease or disorder, comprising administering a dosage form (e.g., oral dosage form) or pharmaceutical composition disclosed herein to a subject in need thereof. The disease or disorder may be a sleep disorder (e.g., excessive daytime sleepiness (EDS), cataplexy, narcolepsy, sleep apnea (e.g., obstructive sleep apnea), sleep induced apnea, diurnal somnolence, central nervous system disorder (e.g., epilepsy, Alzheimer's disease, Parkinson's disease, dementia (e.g., dementia with Lewy bodies and/or vascular dementia), attention disorders, wakefulness disorders, memorization disorders, cognitive deficits (e.g., in aged persons), psychiatric pathologies, depressive and asthenic states, vertigo, and motion sickness, obesity, psychosomatic disorders, respiratory disorders, allergic conditions, inflammatory conditions, cardiac conditions, gastrointestinal conditions, conditions of the urogenital system, conditions of the cutaneous system, stress, migraine, headache, pain, psychotropic disorders, asthma, bronchitis, rhinitis, tracheitis, gastric ulcers, duodenal ulcers, ulcerative colitis, Crohn's disease, irritable bowel syndrome, cystitis, metritis, urinary incontinence, fecal incontinence, urticaria, itching, arthritis, conjunctivitis, premenstrual syndrome, prostatic inflammations, genital disorders, rheumatic conditions, ocular conditions, sialorrhea, convulsion, depression, disorders of the hypothalam hypophyseal system, disorders of the cerebral circulation, and disorders of the immune system.


In preferred aspects, the disease or disorder is a sleep disorder. For example, the disease or disorder can be excessive daytime sleepiness (EDS). The EDS can be in subjects (e.g., adult subjects) with narcolepsy.


The present disclosure further relates to a method for the prevention of undesirable side effects associated with using antipsychotic or antidepressant agents (e.g., aripiprazole, clozapine, olanzapine, risperidone, quetiapine, sertindole, mirtazapine, amitryptiline, and paroxetine), comprising administering a dosage form or pharmaceutical composition of the present disclosure to a subject in need thereof. Non-limiting examples of undesirable side effects associated with using antipsychotic or antidepressant agents includes weight gain, somnolence, and cognitive impairment.


The present disclosure further relates to a method for (i) inducing an extended state of wakefulness; (ii) improving cognitive processes; (iii) reducing food intake; and/or (iv) normalizing vestibular reflexes, comprising administering a dosage form or pharmaceutical composition disclosed herein to a subject in need thereof.


A dosage form (e.g., oral dosage form) or pharmaceutical composition disclosed herein may be taken once daily, twice daily, or more often. More than one dosage form can be administered at once to achieve a desired dose. Preferably, the dosage form (or dosage forms, when multiple dosage forms are used) or pharmaceutical composition is/are taken once daily. For example, one or more dosage forms disclosed herein may be administered orally once daily, e.g., in the morning upon wakening. One or more dosage forms or pharmaceutical compositions disclosed herein may be taken with a frequency (e.g., once daily) so that the total amount of pharmaceutically active agent administered is within the range of from about 10 mg to about 50 mg per day, e.g., about 15 mg to about 40 mg per day. One or more dosage forms or pharmaceutical compositions disclosed herein may be taken with a frequency (e.g., once daily) so that the total amount of pitolisant (in terms of freebase) administered is within the range of from about 17.8 mg to about 35.6 mg per day. For example, a subject may be administered orally two dosage forms once daily, where each dosage form comprises 4.45 mg pitolisant (in terms of freebase), to achieve a daily dose of 8.9 mg pitolisant (in terms of freebase). A subject may be administered orally one dosage form once daily, where the dosage form comprises 17.8 mg pitolisant (in terms of freebase), to achieve a daily dose of 17.8 mg pitolisant (in terms of freebase). A subject may be administered orally two dosage forms once daily, where each dosage form comprises 17.8 mg pitolisant (in terms of freebase), to achieve a daily dose of 35.6 mg pitolisant (free base).


EXAMPLES

In order that the invention described herein may be more fully understood, the following examples are set forth. These examples are offered to illustrate the dose forms (e.g., enteric-coated dose forms), methods of use, and methods of making, and are not to be construed in any way as limiting their scope.


The compounds and dose forms provided herein can be prepared from readily available starting materials using modifications to the specific protocols set forth below that would be well known to those of ordinary skill in the art. It will be appreciated that where typical or preferred process conditions are given, other process conditions can also be used unless otherwise stated. Optimum reaction conditions may vary with the particular reactants or solvents used, but such conditions can be determined by those skilled in the art by routine optimization.


Materials and Methods

Materials used in the examples set forth below were purchased from commercial suppliers. Pitolisant hydrochloride (API) was obtained from Corden Pharma Chenove (MA-22-013). Pitolisant hydrochloride may also be prepared according to the methods described in U.S. Pat. No. 8,207,197, which is incorporated herein by reference in its entirety. Microcrystalline cellulose (Vivapur 102) (diluent) was obtained from JRS Pharma (ME-19-093; ME-19-077). Crospovidone (Polyplasdone XL) (disintegrant) was obtained from BASF (0002356295). Talc (Luzenac pharma) (glidant) was obtained from IMI Fabi (TE142). Magnesium stearate (Kemilub EM-F-V) (lubricant) was obtained from Peter Greven (241231). Anhydrous colloidal silica (Aerosil 200 Pharma) (flowing agent) was obtained from Evonik (150061214). OPADRY® amb II clear (anti-moisture barrier agent) was obtained from Colorcon (TKL53663). ACRYL-EZE® Clear (gastro resistant coating agent) was also obtained from Colorcon (TKL64755). Top mill red 240.17 (coloring agent) was obtained from Biogrund (9030806-L). Water for injection (solvent) was obtained from Lavoisier (19KK18GC) (purified water can also be used). Triethyl citrate (TEC) and PEG8000 (plasticizers) were obtained from Colorcon (DTR527487 and DTR550141).


Weighing scales used included OGAL075, OGAL023, OGAL367, AND OGAL351-1. Blending used Soneco blender OGAL112, Soneco container 100 L OGAL108, and 800 μm manual sieve OGAL211-IV3. Compression used a tableting machine FETTE P1200 OGAL035-1, Turret B OGAL035-2, Punches and die D7.5R9.2, D7.5 B132 I/S/M, engraving “20.” Coating used Manesty OGAL117-1, 330 mm perforated pan OGAL117-4, nozzle 0.8 mm, pump OGAL236, agitator OAMA092, adequate beaker, tubes 4.8-2.4, and magnetic stirrer.


Example 1. Preparation of Exemplary Pitolisant Core Tablets (Batch 1 Core)
Manufacturing Formula

Core tablets were prepared using the manufacturing formula described in Table 1.









TABLE 1







Manufacturing formulae for core tablets for Batch 1.
















Theoretical
Actual




Unit formula
Formula
quantity
quantity


Raw material
Batch No.
(mg/unit)
(%)
(g)
(g)















Pitolisant HCl
MA-22-013
20
16
1440
1440.1


Microcrystalline
ME-19-093
81
64.8
5832
5832


cellulose
ME-19-077


Crospovidone
0002356295
10
8
720
720


Talc
TE142
10
8
720
720


Colloidal silica
150061214
1
0.8
72
72


Magnesium
241231
3
2.4
216
216


stearate







Total

125
100
9000
9000.1









Blending Step

Prior to tableting the manufacturing formula was subjected to sieving and blending steps. Table 2. Provides the manufacturing parameters used during these steps to provide the final blend for tableting.









TABLE 2







Blending parameters












Required



Blending step
Parameters
Value
Batch 1





Sieving 1
Sieve aperture
800 μm
800 μm


½ cellulose +


crospovidone +


talc +


pitolisant HCl +


½ cellulose


Blending 1
Blending time
3 min 38 sec
3 min 38 sec


Sieved material
Blending speed
 22 rpm
 22 rpm



Rotation number
80
80


Sieving 2
Sieve aperture
800 μm
800 μm


Aerosil +


magnesium


sterate


Lubrication
Blending time
3 min 38 sec
3 min 38 sec


Blending 1 +
Blending speed
 22 rpm
 22 rpm


aerosil +
Rotation number
80
80


magnesium
Filling level

50%


stearate









Manufacturing Formula

The final blend was tested for its flowability and density (Table 3). For comparison, a reference batch blend (Batch 2) was measured. Despite the appearance of poor flowability in Batch 1, there were no flowing issues during the compression step described below.









TABLE 3







IPC on final blends











Batch Number
Batch 1
Batch 2















Sample size (g)
50.105




Flowability



Flowing time (s)
Infinite1
Infinite



Flowing speed (g/s)
NA
0



Tapped volume (mL)



V0
128
118



V10
118
114



V500
96
94



V1250
94
92



V2500
92
/



V10-V500
22
20



Density (g/mL)



D0
0.39
0.42



D10
0.42
0.44



D500
0.52
0.53



D1250
0.53
0.54



D2500
0.54
/



Carr index
28
22



Hausner ratio
1.39
1.28



Flow character
Poor
Passable



LOD (%)
3.9
4.69







NA: not applicable;




1Blend flows when regularly tapping on funnel







Tableting

Batch 1 tablets were manufactured on a rotative tableting press using 5 punches D7.5R9.2 engraved with “20.” Main compression force and filling volume were adapted to reach 50N hardness tablets and 125.0 mg tablet mass. Tableting parameters are provided in Table 4.









TABLE 4







Tableting parameters and IPC results











Batch 1



Specifications
D7.5R9.2 “20”













Tableting actual parameters




Tableting speed (tab/min)
/
90 000 tblt/h




(=18750




tblt/h with




5 punches)


Compression force (kN)
/
6.2


Pre-compression force (kN)
/
1.5


Filling height (mm)
/
5.98


Distributor speed (%)
/
20


IPC results on n = 20 tablets


Mean tablet mass (mg)
125.0 ± 3.0%
125.0



(121.3-128.8)


Min-Max mean tablet
T1: 7.5%
124.1-126.0


mass (mg)
(115.6-134.4)


Min-Max individual
T2: 15%
121.1-128.2


tablet mass (mg)
(106.3-143.8)


CV (%)
/
0.95


Mean tablet hardness (N)
/
54


Min-Max mean hardness
/
53-55


Min-Max individual hardness
/
45-63


CV (%)
/
6.04


Mean tablet thickness (mm)
/
3.08


Min-Max mean thickness
/
3.04-3.10


Min-Max individual thickness
/
3.02-3.13


CV (%)
/
0.35


Mean tablet diameter (mm)
/
7.58


Min-Max mean diameter
/
7.57-7.59


Min-Max individual diameter
/
7.51-7.66


CV (%)
/
0.45


Aspect

Round white




tablets




with one face




marked “20”


Bulk density (g/mL)
/
0.73









Disintegration and Friability Tests

The Batch 1 tablets were then subjected to disintegration and friability tests, the results of which are provided in Table 5.









TABLE 5







Disintegration and Friability Test Results of Batch 1 tablets.











Representative



Settings
sample














Disintegration time [min-s]
Min
1 min 50 sec
3 min 13 sec


Specification: <15 min
Max
3 min 33 sec
4 min 11 sec


Friability [%]

0.1%
0.0 (=0.05%)


Specification: ≤1.0%









Mass evolution during compression was tested, the results of which are provided in FIG. 2. Tablet mass is very stable, and remains below 1.5%. Increase in RSD over time was attributed to lack of sufficient powder in the feeding system.


Tablet hardness was also evaluated during compression, and the results are provided in FIG. 3. Tablet hardness was as stable as the tablet mass.


The Batch 1 core tablets complied in terms of disintegration time and friability results. Tablet mass and hardness were very stable throughout the process.


Example 2. Preparation of Exemplary Enteric-Coated Pitolisant (Batch 1)

The exemplary core tablet of Example 1 (Batch 1 core) was coated, first with an anti-moisture barrier coating, followed by a gastric resistant coating.


The coasting was performed in perforated pan using a spray nozzle of 0.8 mm. A first layer of OPADRY® amb II (3%) was performed (sub-layer). A coloring agent was also added in this layer. Then the gastroresistant coating was applied using ACRYL-EZE®. The plasticizers used were TEC (12%). The suspensions for the coatings were prepared using a magnetic bar and beaker to stir.


Anti-Moisture Barrier Coating Step (Batch 1)

A suspension of OPADRY® amb II was prepared according to the manufacturing formulae provided in Table 6. Suspension preparation details are provided in Table 7.









TABLE 6







Manufacturing formulae for Amb II coating.













Unit
Theoretical
Actual




formula
quantity
quantity


Raw material
Batch No.
(mg / unit)
(g)
(g)














Core
Batch 1
125
800
800


OPADRY ® amb II
TKL53663
3.75
48
48


clear


Top mill red
9030806-L
0.1
1.28
0.39497


Water for injection
19KK18GC

279.25
279.71


Total
/
128.85
/
/






115% solids, in excess (x2)














TABLE 7







Suspension preparation (OPADRY ® amb II)









Step
Parameters
Batch 1













OPADRY ® amb II and top
Stirring speed
450 to 500
rpm


mill introduction
Duration
45
sec


Stirring
Stirring speed
500
rpm










Stirring time
2 hours 30 mins







Suspension was homogenous and stirring speed was decreased at 300 rpm for coating.






The coating parameters for the anti-moisture barrier layer (OPADRY® amb II) are provided in Table 8.









TABLE 8





Coating parameters for OPADRY ® amb II coating



















Pre heat (min)

10



Product temperature (° C.)
Start
21.8




End
45.6



Pan speed (rpm)
Start
5 (intermittent)




End
5 (intermittent)



Actual inlet air temp. (° C.)
Start
65




End
66



Outlet air temp (° C.)
Start
51.2




End
54.6



Air flow rate (m3/h)
Start
300




End
303



Coating time (min)

80



Product temp. (° C.)
Min
45.6




Max
47.4



Pan speed (rpm)
Min
25




Max
28



Actual inlet air temp. (° C.)
Min
64.6




Max
66.9



Outlet air temp (° C.)
Min
51.1




Max
55.6



Air flow rate (m3/h)
Min
293




Max
310



Atomization pressure (bar)

1.4 to 1.2



Spray width (bar)

1.8 to 1.6



Spraying flow rate (g/min)
Min
2.24




Max
3.38



Pump setting (%)

10-11



Drying time (min)

5



Product temp. (° C.)
Start
47.4




End
48.2



Pan speed (rpm)
Start
15




End
15



Actual inlet air temp. (° C.)
Start
66.8




End
66.5



Outlet air temp (° C.)
Start
52.7




End
53.2



Air flow rate (m3/h)
Start
299




End
311



Weight gain (%)

3.0



Cooling time (min)

20



Product temp. (° C.)
Start
48.2




End
25.7



Pan speed (rpm)
Start
5 (intermittent)




End
5 (intermittent)



Actual inlet air temp. (° C.)
Start
66.5




End
33.5



Outlet air temp (° C.)
Start
53.2




End
28.3



Air flow rate (m3/h)
Start
311




End
305










Gastroresistant Coating Step (Batch 1)

A suspension of ACRYL-EZE® was prepared according to the manufacturing formulae provided in Table 9. Suspension preparation details are provided in Table 10.









TABLE 9







Manufacturing formulae for ACRYL-EZE ® coating.













Unit
Theoretical
Actual




formula
quantity
quantity


Raw material
Batch No.
(mg/unit)
(g)
(g)














OPADRY ® amb II
Batch 1 coated
128.9375
767.7
767.7


coated core
tablets (above)


Acryl eze clear
TKL64755
19.34
240.0
240.0


TEC
DTR527487
2.32
28.8
28.8


Purified water
/
/
960.0
960.01


Total
/
150.6
/
/






120% solids, in excess (×2)














TABLE 10







Suspension preparation (ACRYL-EZE ®)









Step
Parameters
Batch 1





PEG 8000 introduction
Stirring speed
400 then 500 rpm



Duration
45 sec + 5 min


Acryl eze introduction
Stirring speed
About 600 rpm



Duration
1 min 30 sec + 5 min


Stirring
Stirring speed
About 500 tpm



Stirring time
2 hours 50 min





Suspension was homogenous and stirring speed was decreased at 300 rpm for coating.






The coating parameters for the gastroresistant layer (ACRYL-EZE®) are provided in Table 11.









TABLE 11





Coating parameters for ACRYL-EZE ® coating



















Pre heat (min)

6



Product temperature (° C.)
Start
21.0




End
34.5



Pan speed (rpm)
Start
5 (intermittent)




End
5 (intermittent)



Actual inlet air temp. (° C.)
Start
43.7




End
44.0



Outlet air temp (° C.)
Start
38.9




End
39.1



Air flow rate (m3/h)
Start
289




End
289



Coating time (min)

170



Product temp. (° C.)
Min
32.5




Max
34.5



Pan speed (rpm)
Min
28




Max
28



Actual inlet air temp. (° C.)
Min
42.7




Max
44.7



Outlet air temp (° C.)
Min
36.5




Max
39.1



Air flow rate (m3/h)
Min
289




Max
303



Atomization pressure (bar)

1.2



Spray width (bar)

1.6



Spraying flow rate (g/min)
Min
2.60




Max
5.11



Pump setting (%)

10-13



Drying time (min)

5



Product temp. (° C.)
Start
33.2




End
36.6



Pan speed (rpm)
Start
15




End
15



Actual inlet air temp. (° C.)
Start
44.7




End
44.3



Outlet air temp (° C.)
Start
37.3




End
37.9



Air flow rate (m3/h)
Start
296




End
304



Weight gain (%)

17.6 (corresponding





to 15.7% of GR)



Cooling time (min)

14



Product temp. (° C.)
Start
36.6




End
25.5



Pan speed (rpm)
Start
5 (intermittent)




End
5 (intermittent)



Actual inlet air temp. (° C.)
Start
44.3




End
29.4



Outlet air temp (° C.)
Start
37.96




End
26.3



Air flow rate (m3/h)
Start
296




End
289










Final IPC was performed on the exemplary coated tablets. The results are provided in Table 12.









TABLE 12





IPC results of exemplary enteric-coated tablets


















Mean tablet mass (mg)
149.0



Min-Max mean tablet mass (mg)
145.1-153.2



CV (%)
1.73



Mean tablet hardness (N)
110



Min-Max mean hardness
 87-127



CV (%)
10.15



Mean tablet thickness (mm)
3.43



Min-Max mean thickness
3.40-3.46



CV (%)
0.60



Mean tablet diameter (mm)
7.78



Min-Max mean diameter
7.70-7.86



CV (%)
0.59










The addition of dye confirmed that the coating was uniform.


Example 3. Preparation of Exemplary Enteric Coated Tablets (Batches 3-6)

Four additional batches were prepared, which are identified in Table 13. For each, the same core tablet prepared in Example 1 was used (Batch 1 core). Different coatings were applied following the protocols set forth below.









TABLE 13







Batches 3-6 comprising different amounts of gastroresistant


layer (GR), anti-moisture barrier layer (amb


II), and plasticizer (PEG or TEC).











F1
F2
F3














Batch 3
15% weight gain GR
3% weight gain ambII
PEG 8%


Batch 4
10% weight gain GR
5% weight gain ambII
PEG 8%


Batch 5
10% weight gain GR
3% weight gain ambII
TEC 12%


Batch 6
15% weight gain GR
5% weight gain ambII
TEC 12%









Anti-Moisture Barrier Coating Step

A 3% suspension of OPADRY® amb II was prepared according to the manufacturing formulae provided in Table 14, and a 5% suspension of OPADRY® amb II was prepared according to the manufacturing formulae provided in Table 15. As both the OPADRY® amb II 3% and 5% coatings were made on the same day, only one solution was made for each. Suspension preparation details are provided in Table 16.









TABLE 14







Manufacturing formulae for 3% OPADRY ® amb II coating.













Unit formula
Theoretical
Actual


Raw material
Batch No.
(mg/unit)
quantity (g)
quantity (g)














Core
Batch 1 core
125
1600
/


OPADRY ®
TKL53663
3.75
96
96


amb II clear


Water for
19KK18GC

544.0
544.11


injection




total
/
128.75
/
/






115% solids, in excess (×2)














TABLE 15







Manufacturing formulae for 5% OPADRY ® amb II coating.













Unit formula
Theoretical
Actual


Raw material
Batch No.
(mg/unit)
quantity (g)
quantity (g)














Core
Batch 1 core
125
1600
/


OPADRY ®
TKL53663
6.25
160
160


amb II clear


Purified water
19KK18GC

906.67
906.71


Total
/
131.25
/
/






115% solids, in excess (×2)














TABLE 16







Suspension preparation (OPADRY ® amb II)










Step
Parameters
Batches 3 and 5
Batches 4 and 6





AmbII
Stirring speed
450 to 500 rpm
300 rpm


introduction
Duration
55 sec
2 min 0 sec


Stirring
Stirring speed
500 rpm
300 rpm



Stirring time
1 hours 5 mins
1 hour 30 min





Suspension was homogenous and stirring speed was decreased at 300 rpm for coating.






The coating parameters for the anti-moisture barrier layer (OPADRY® amb II) applied to Batches 3-6 are provided in Table 17.









TABLE 17







Coating parameters for OPADRY ® amb II coating on Batches 3-6. Batch 1 is included for comparison









Batch













1
3
4
5
6

















Pre heat (min)

10
9
8
8
9


Product
Start
21.8
21.1
20.8
21.9
21.7


temperature
End
45.6
46.9
46.1
47.6
47.5


(° C.)


Pan speed (rpm)
Start
5 (intermittent)
5 (intermittent)
5 (intermittent)
5 (intermittent)
5 (intermittent)



End
5 (intermittent)
5 (intermittent)
5 (intermittent)
5 (intermittent)
5 (intermittent)


Actual inlet air
Start
65
64.3
66.8
65.8
65.7


temp. (° C.)
End
66
65.5
65.1
66.2
65.9


Outlet air temp
Start
51.2
51.6
54.7
51.8
52.8


(° C.)
End
54.6
52.2
51.8
52.9
52.6


Air flow rate
Start
300
321
304
304
304


(m3/h)
End
303
305
303
306
300


Coating time

80
70
100
80
110


(min)


Product temp.
Min
45.6
45.7
46.0
46.4
46.3


(° C.)
Max
47.4
47.1
47.3
47.6
47.5


Pan speed (rpm)
Min
25
25
25
25
25



Max
28
28
28
28
25


Actual inlet air
Min
64.6
65.7
64.8
64.9
64.3


temp. (° C.)
Max
66.9
66.9
66.6
66.2
65.9


Outlet air temp
Min
51.1
51.5
51.5
51.5
51.4


(° C.)
Max
55.6
52.6
52.6
52.9
52.6


Air flow rate
Min
293
295
295
291
295


(m3/h)
Max
310
313
308
306
307


Atomization

1.4 to 1.2
1.2
1.2
1.2
1.2


pressure (bar)


Spray width

1.8 to 1.6
1.6
1.6
1.6
1.6


(bar)


Spraying flow
Min
2.24
2.58
2.68
2.75
2.66


rate (g/min)
Max
3.38
3.30
3.44
3.43
3.54


Pump setting

10-11
10
10-11
10-11
10-11


(%)


Drying time

5
5
5
5
5


(min)


Product temp.
Start
47.4
46.5
47.3
46.4
46.7


(° C.)
End
48.2
50.5
49.4
50.5
49.9


Pan speed (rpm)
Start
15
15
15
15
15



End
15
15
15
15
15


Actual inlet air
Start
66.8
65.7
64.8
65.2
64.5


temp. (° C.)
End
66.5
66.3
65.2
65.0
64.3


Outlet air temp
Start
52.7
52.2
52.6
51.8
51.7


(° C.)
End
53.2
53.4
53.8
53.2
52.6


Air flow rate
Start
299
302
298
303
307


(m3/h)
End
311
303
298
295
306


Weight gain

3.0
3.0
5.0
3.1
5.0


(%)


Cooling time

20
15
14
14
18


(min)


Product temp.
Start
48.2
50.2
49.4
50.5
49.9


(° C.)
End
25.7
28.7
29.7
29.3
28.9


Pan speed (rpm)
Start
5 (intermittent)
5 (intermittent)
5 (intermittent)
5 (intermittent)
5 (intermittent)



End
5 (intermittent)
5 (intermittent)
5 (intermittent)
5 (intermittent)
5 (intermittent)


Actual inlet air
Start
66.5
66.3
65.2
65.0
64.3


temp. (° C.)
End
33.5
36.5
37.2
37.6
35.9


Outlet air temp
Start
53.2
53.4
53.8
53.2
52.6


(° C.)
End
28.3
31.5
33.5
32.4
32.8


Air flow rate
Start
311
303
298
295
306


(m3/h)
End
305
303
303
296
295









Gastroresistant Coating Step

Suspensions of ACRYL-EZE® were prepared for each of Batches 3-6 according to the manufacturing formulae provided in Tables 18-21. Suspension preparation details are provided in Table 22.









TABLE 18







Manufacturing formulae for ACRYL-EZE ® coating for Batch 3.













Unit
Theoretical
Actual




formula
quantity
quantity


Raw material
Batch No.
(mg/unit)
(g)
(g)














amb II coated
Batch 3 coated
128.75
800
784.1


core
tablets (above)


ACRYL-EZE ®
TKL64755
19.31
240
240


clear


PEG8000
DTR550141
1.545
19.2
19.2


Purified water
/
/
960.0
959.91


Total
/
149.61
/
/






120% solids, in excess (×2)














TABLE 19







Manufacturing formulae for ACRYL-EZE ® coating for Batch 4.













Unit
Theoretical
Actual




formula
quantity
quantity


Raw material
Batch No.
(mg/unit)
(g)
(g)














amb II coated
Batch 4 coated
131.25
800
763.6


core
tablets (above)


ACRYL-EZE ®
TKL64755
13.125
160
160


clear


PEG8000
DTR550141
1.05
12.8
12.8


Purified water
/
/
640
640.41


Total
/
145.425
/
/






120% solids, in excess (×2)














TABLE 20







Manufacturing formulae for ACRYL-EZE ® coating for Batch 5.













Unit
Theoretical
Actual




formula
quantity
quantity


Raw material
Batch No.
(mg/unit)
(g)
(g)














amb II coated
Batch 5 coated
128.75
800
765


core
tablets (above)


ACRYL-EZE ®
TKL64755
12.875
160
160


clear


TEC
DTR527487
1.545
19.2
19.2


Purified water
/
/
640.0
640.11


Total
/
143.17
/
/






120% solids, in excess (×2)














TABLE 21







Manufacturing formulae for ACRYL-EZE ® coating for Batch 6.













Unit
Theoretical
Actual




formula
quantity
quantity


Raw material
Batch No.
(mg/unit)
(g)
(g)














amb II coated
Batch 6 coated
131.25
800
776.4


core
tablets (above)


ACRYL-EZE ®
TKL64755
19.6875
240
240


clear


TEC
DTR527487
2.3625
28.8
28.8


Purified water
/
/
960
960.21


Total
/
153
/
/






120% solids, in excess (×2)














TABLE 22







Batches 3-6 suspension preparation (ACRYL-EZE ®)












Step
Parameters
Batch 3
Batch 4
Batch 5
Batch 6





PEG 8000
Stirring speed (rpm)
500
300
350
350


introduction
Duration
40 sec + 5 min
20 sec + 5 min
5 min
5 min


ACRYL-EZE ®
Stirring speed (rpm)
About 500
300 to 600
500 to 600
500 to 600


introduction
Duration (min)
2 min + 5 min
2.5 min + 5 min
1 min + 4 min
2.25 min + 5 min


Stirring
Stirring speed (rpm)
About 400
About 500
About 500
About 500



Stirring time
3 h 0 min
1 h 10 min
1 h 0 min
3 h 0 min





Suspension was homogenous and stirring speed was decreased at 250/300 rpm for coating.






The coating parameters for the gastroresistant layer (ACRYL-EZE®) applied to Batches 3-6 are provided in Table 23.









TABLE 23







Coating parameters for ACRYL-EZE ® coating of Batches 3-6. Batch 1 is included for comparison









Batch













1
3
4
5
6

















Pre heat

6
7
8
8
7


(min)


Product
Start
21.0
20.9
20.4
20.9
20.5


temperature
End
34.5
37.3
34.6
35.1
34.7


(° C.)


Pan speed
Start
5
5
5
5
5


(rpm)

(intermittent)
(intermittent)
(intermittent)
(intermittent)
(intermittent)



End
5
5
5
5
5




(intermittent)
(intermittent)
(intermittent)
(intermittent)
(intermittent)


Actual inlet
Start
43.7
44.9
45.0
45.0
44.9


air temp.
End
44.0
45.1
45.0
44.1
44.4


(° C.)


Outlet air
Start
38.9
37.2
37.4
38.1
36.9


temp (° C.)
End
39.1
38.5
38.1
37.7
37.7


Air flow
Start
289
303
297
302
298


rate (m3/h)
End
289
295
301
301
310


Coating

170
170
120
120
180


time (min)


Product
Min
32.5
32.1
32.3
32.3
32.0


temp. (° C.)
Max
34.5
37.3
34.6
35.1
34.7


Pan speed
Min
28
28
28
27
28


(rpm)
Max
28
28
28
28
28


Actual inlet
Min
42.7
43.7
44.3
44.1
44.3


air temp.
Max
44.7
45.2
45.0
44.7
45.3


(° C.)


Outlet air
Min
36.5
36.8
45.0
44.7
37.0


temp (° C.)
Max
39.1
38.5
36.9
36.8
37.8


Air flow
Min
289
294
293
296
296


rate (m3/h)
Max
303
308
303
307
310


Atomization

1.2
1.2
1.2
1.2
1.2


pressure (bar)


Spray width

1.6
1.6
1.6
1.6
1.6


(bar)


Spraying
Min
2.60
2.90
3.02
2.86
2.82


flow rate
Max
5.11
5.12
4.97
5.04
4.90


(g/min)


Pump

10-13
9-15
10-14
10-14
10-15


setting (%)


Drying time

5
5
5
5
5


(min)


Product
Start
33.2
33.1
33.5
32.5
32.7


temp. (° C.)
End
36.6
37.1
36.7
34.1
36.3


Pan speed
Start
15
15
15
15
15


(rpm)
End
15
15
15
15
15


Actual inlet
Start
44.7
44.5
44.5
44.5
44.5


air temp.
End
44.3
44.4
44.5
44.5
44.4


(° C.)


Outlet air
Start
37.3
37.0
37.0
37.1
37.2


temp (° C.)
End
37.9
39.1
38.1
38.3
38.8


Air flow
Start
296
306
303
297
300


rate (m3/h)
End
304
292
300
302
306


Weight gain

17.6
16.1
11.0
11.3
17.3


(%)

(corresponding
corresponding
corresponding
corresponding
corresponding




to 15.7% of GR)
to 14.9% GR
to 9.8% GR
to 10.9% GR
to 15.4% GR


Cooling

14
9
13
8
8


time (min)


Product
Start
36.6
37.1
36.7
34.1
36.3


temp. (° C.)
End
25.5
29.8
27.5
27.9
28.7


Pan speed
Start
5
5
5
5
5


(rpm)

(intermittent)
(intermittent)
(intermittent)
(intermittent)
(intermittent)



End
5
5
5
5
5




(intermittent)
(intermittent)
(intermittent)
(intermittent)
(intermittent)


Actual inlet
Start
44.3
44.4
44.5
44.5
44.4


air temp.
End
29.4
37.4
32.0
33.3
34.9


(° C.)


Outlet air
Start
37.96
39.1
38.1
33.8
38.8


temp (° C.)
End
26.3
32.9
29.2
29.3
30.6


Air flow
Start
296
292
300
302
306


rate (m3/h)
End
289
299
301
297
308









Final IPC was performed on the exemplary tablets after anti-moisture barrier coating (OPADRY® amb II) was applied and after applying gastroresistant coating (ACRYL-EZE®) for Batches 3-6. The results are provided in Table 24.









TABLE 24







IPC results of exemplary enteric-coated tablets. Batch 1 is included for comparison.













Batch 1
Batch 3
Batch 4
Batch 5
Batch 6











OPADRY ® amb II-coated tablets












Mean tablet mass (mg)
128.2
127.5
129.2
127.8
129.3


Min-Max mean tablet mass (mg)
125.6-130.0
125.4-129.6
127.7-129.6
125.9-129.3
125.2-130.5


CV (%)
1.09
1.04
1.04
0.97
1.24


Mean tablet hardness (N)
74
73
80
75
79


Min-Max mean hardness
70-83
67-84
70-87
68-81
71-85


CV (%)
5.83
7.77
7.54
5.61
5.91


Mean tablet thickness (mm)
3.15
3.14
3.17
3.14
3.17


Min-Max mean thickness
3.11-3.16
3.11-3.17
3.14-3.19
3.12-3.17
3.15-3.20


CV (%)
0.48
0.72
0.57
0.52
0.54


LOD (%)
3.4
3.34
3.23
3.21
3.04


Disintegration time in HCl
Min 4 m 1 s
Min 3 m 53 s
Min 4 m 22 s
Min 3 m 42 s
Min 4 m 21 s


(minutes (m) and seconds (s)).
Max 4 m 42 s
Max 5 m 49 s
Max 5 m 10 s
Max 4 m 37 s
Max 4 m 41 s







ACRYL-EZE ® coated tablets












Mean tablet mass (mg)
149.0
147.8
143.5
141.6
152.2


Min-Max mean tablet mass (mg)
145.1-153.2
145.3-150.2
140.9-145.2
138.2-144.1
149.5-155.2


CV (%)
1.73
0.95
1.10
1.50
1.04


Mean tablet hardness (N)
110
110
92
91
115


Min-Max mean hardness
 87-127
 94-118
 84-102
62-99
107-125


CV (%)
10.15
6.24
6.35
12.02
5.43


Mean tablet thickness (mm)
3.43
3.41
3.35
3.34
3.46


Min-Max mean thickness
3.40-3.46
3.38-3.45
3.30-3.39
3.30-3.37
3.42-3.48


CV (%)
0.60
0.65
0.96
0.65
0.60


LOD (%)
2.83
3.08
2.89
2.99
2.88









Example 4. Preparation of an Exemplary Enteric Coated Tablets (Batch 7)

Another enteric-coated tablet batch (Batch 7) was prepared according to the following protocol.


This batch was prepared using the same core tablet prepared in Example 1 (Batch 1 core). The first coating was carried out with OPADRY® amb II BEIGE (reference 88A270004) (weight gain 3%) and the second coating was performed with ACRYL-EZE® clear at different percentages (up to 18%). The plasticizer used for this batch was PEG8000 (8%).


Anti-Moisture Barrier Coating Step (Batch 7)

An OPADRY® amb II BEIGE suspension was prepared according to the manufacturing formulae provided in Table 25. Suspension preparation details are provided in Table 26.









TABLE 25







Manufacturing formulae for Batch 7 OPADRY ® amb


II BEIGE coating.













Unit formula
Theoretical
Actual


Raw material
Batch No.
(mg/unit)
quantity (g)
quantity (g)














Core
Batch 1 core
125
800.0
800.3


ambII beige
TKL66169
3.75
48.0
 48.0


88A270004


Purified Water
/
/
272
 272.11


Total
/
128.75
/
/






115% solids, in excess (×2)














TABLE 26







Suspension preparation for Batch 7 OPADRY ® amb


II BEIGE coating.











Step
Parameters
Batch 7







AmbII
Stirring speed
About 500 rpm



introduction
Duration
2 min 0 sec



Stirring
Stirring speed
About 600 rpm




Stirring time
55 min







Suspension was homogenous and stirring speed was decreased at 300 rpm for coating.






The coating parameters for the anti-moisture barrier layer (OPADRY® amb II BEIGE) applied to Batch 7 is provided in Table 27.









TABLE 27







Coating parameters for OPADRY ® amb II coating


on Batch 7. Batch 1 is included for comparison.











Batch
1
7
















Pre heat (min)

10
8



Product
Start
21.8
21.6



temperature (° C.)
End
45.6
47.4



Pan speed (rpm)
Start
5 (intermittent)
4 (intermittent)




End
5 (intermittent)
4 (intermittent)



Actual inlet air
Start
65
64.8



temp. (° C.)
End
66
66.8



Outlet air temp
Start
51.2
52.1



(° C.)
End
54.6
53.6



Air flow rate
Start
300
304



(m3/h)
End
303
299



Coating time

80
85



(min)



Product temp.
Min
45.6
45.4



(° C.)
Max
47.4
47.4



Pan speed (rpm)
Min
25
28




Max
28
28



Actual inlet air
Min
64.6
66.8



temp. (° C.)
Max
66.9
68.3



Outlet air temp
Min
51.1
52.0



(° C.)
Max
55.6
53.8



Air flow rate
Min
293
295



(m3/h)
Max
310
307



Atomization

1.4 to 1.2
1.2



pressure (bar)



Spray width (bar)

1.8 to 1.6
1.6



Spraying flow
Min
2.24
2.54



rate (g/min)
Max
3.38
3.10



Pump setting (%)

10-11
10



Drying time (min)

5
5



Product temp.
Start
47.4
47.0



(° C.)
End
48.2
49.5



Pan speed (rpm)
Start
15
15




End
15
15



Actual inlet air
Start
66.8
66.7



temp. (° C.)
End
66.5
67.7



Outlet air temp
Start
52.7
53.6



(° C.)
End
53.2
53.8



Air flow rate
Start
299
302



(m3/h)
End
311
299



Weight gain (%)

3.0
2.9



Cooling time

20
22



(min)



Product temp.
Start
48.2
47.0



(° C.)
End
25.7
29



Pan speed (rpm)
Start
5 (intermittent)
5 (intermittent)




End
5 (intermittent)
5 (intermittent)



Actual inlet air
Start
66.5
66.7



temp. (° C.)
End
33.5
37.5



Outlet air temp
Start
53.2
53.6



(° C.)
End
28.3
31.4



Air flow rate
Start
311
299



(m3/h)
End
305
299










Gastroresistant Coating Step (Batch 7)

Suspensions of ACRYL-EZE® were prepared for Batch 7 according to the manufacturing formulae provided in Table 28. Suspension preparation details are provided in Table 29.









TABLE 28







Manufacturing formulae for acryl-ease coating for Batch 7.













Unit

Actual




formula
Theoretical
quantity


Raw material
Batch No.
(mg/unit)
quantity (g)
(g)














amb II
Batch 7 coated
128.75
800
787.5


coated core
tablets (above)


ACRYL-EZE ®
ME-22-083
21.8875
272
272  


clear


PEG8000
DTR550141
1.751
21.76
 21.8


Purified water
/
/
1088.0
1087.91


Total
/
152.3885
/
/






120% solids, in excess (×2)














TABLE 29







Suspension preparation details for Batch 7.











Step
Parameters
Batch 7







PEG 8000
Stirring speed (rpm)
About 600



introduction
Duration
30 sec + 7 min



ACRYL-EZE ®
Stirring speed (rpm)
About 600 to 700



introduction
Duration (min)
3 min



Stirring
Stirring speed (rpm)
About 500




Stirring time
2 h 30 min










The coating parameters for the gastroresistant layer (ACRYL-EZE®) applied to Batch 7 is provided in Table 30.









TABLE 30







Coating parameters for ACRYL-EZE ® coating


of Batch 7. Batch 1 is included for comparison









Batch
1
7













Pre heat (min)

6
5


Product
Start
21.0
26.6


temperature (° C.)
End
34.5
34.9


Pan speed (rpm)
Start
5 (intermittent)
5 (intermittent)



End
5 (intermittent)
5 (intermittent)


Actual inlet air
Start
43.7
44.8


temp. (° C.)
End
44.0
44.3


Outlet air temp
Start
38.9
36.8


(° C.)
End
39.1
38.0


Air flow rate
Start
289
300


(m3/h)
End
289
300


Coating time

170
190


(min)


Product temp.
Min
32.5
31.9


(° C.)
Max
34.5
34.9


Pan speed (rpm)
Min
28
28



Max
28
28


Actual inlet air
Min
42.7
42.4


temp. (° C.)
Max
44.7
44.3


Outlet air temp
Min
36.5
36.2


(° C.)
Max
39.1
38.0


Air flow rate
Min
289
295


(m3/h)
Max
303
304


Atomization

1.2
1.2


pressure (bar)


Spray width (bar)

1.6
1.6


Spraying flow
Min
2.60
2.70


rate (g/min)
Max
5.11
4.95


Pump setting (%)

10-13
10-15


Drying time (min)

5
5


Product temp.
Start
33.2
32.7


(° C.)
End
36.6
35.8


Pan speed (rpm)
Start
15
15



End
15
15


Actual inlet air
Start
44.7
42.4


temp. (° C.)
End
44.3
42.4


Outlet air temp
Start
37.3
36.2


(° C.)
End
37.9
37.6


Air flow rate
Start
296
304


(m3/h)
End
304
299


Weight gain (%)

17.6 (corresponding
18.8 (corresponding




to 15.7% of GR)
to 16.9% of GR)


Cooling time

14
8


(min)


Product temp.
Start
36.6
35.8


(° C.)
End
25.5
28.3


Pan speed (rpm)
Start
5 (intermittent)
5 (intermittent)



End
5 (intermittent)
5 (intermittent)


Actual inlet air
Start
44.3
42.4


temp. (° C.)
End
29.4
36.6


Outlet air temp
Start
37.96
37.6


(° C.)
End
26.3
30.0


Air flow rate
Start
296
299


(m3/h)
End
289
303









Example 5. Quality Control Testing

Batches 1 and 3-6 were subjected to quality control testing, including disintegration, dissolution, content uniformity. Batches 3, 5, and 6 were also subjected to stability testing for appearance, average mass, water content, assay and purity.


Disintegration-Batch 1

Disintegration studies were performed on n=6 tablets from a representative sample of Batch 1 with 10% GR (Batch 1-1); 12% GR (Batch 1-2); and 15% GR coating (Batch 1-3). Studies were performed in two steps, first in HCl 0.1M for 2 hours (without disks), then in phosphate buffer at pH 6.8 for 1 hour (with disks), according to the Ph.Eur. 2.9.1/USP <711>.


Results of the studies are provided in Table 31. All disintegration results were compliant.









TABLE 31







Disintegration results for Batches 1-1, 1-2, and 1-3













Batch 1-1
Batch 1-2
Batch 1-3



Specification
10% GR
12% GR
15% GR















First in HCl
No tablet shows
comply
comply
Comply


0.1M for 2 h
signs of either


(without disks)
disintegration



(apart from



fragments of



coating) or



cracks that



would allow



the escape



of contents


Then phosphate
Comply with
7 min
9 min
9 min


buffer pH 6.8 for
Eur. Ph.


1 h (with disks)
2.9.1 <1 hour









Disintegration—Batches 3-6

Disintegration studies were performed on n=6 tablets from a representative sample of Batches 3-6, in two steps, first in HCl 0.1M for 2 hours (without disks), then in phosphate buffer pH 6.8 for 1 hour (with disks), according to the Ph.Eur. 2.9.1/USP <711>. For the Batch 4 disintegration results obtained were not compliant, in acid media, 5 units were open after 120 minutes in HCl 0.1M. For Batch 6, 2 tablets were swollen after 120 minutes in HCl 0.1M. The other batches 3 and 5 were compliant. Results are provided in Table 32









TABLE 32







Disintegration Studies with Batches 3-6













Specification
Batch 3
Batch 4
Batch 5
Batch 6
















First in HCl
No tablet shows
Comply
Do not
Comply
Do not


0.1M for 2 h
signs of either

comply

comply


(without disks)
disintegration (apart

(5 tablets

(2 swollen)



from fragments of

open)



coating) or cracks



that would allow the



escape of contents


Then phosphate
Comply with
8 min
7 min
4 min
8 min


buffer pH 6.8 for
Eur. Ph. 2.9.1


1 h (with disks)









Disintegration—Batch 7

Disintegration studies were performed on n=6 tablets from a representative sample of Batch 7 with 10% GR (7-1), 12% GR (7-2), 15% GR (7-3), and 17% GR (7-4). Studies were performed in two steps, first in HCl 0.1M for 2 hours (without disks), then in phosphate buffer pH 6.8 for 1 hour (with disk), according to the Ph.Eur. 2.9.1/USP <711>.


For all the amount tested of GR coating (from 10% to 17%) of the Batch 7 disintegration results obtained are compliant, in acid media and in phosphate buffer. Results are provided in Table 33.









TABLE 33







Disintegration results for Batches 7-1, 7-2, 7-3, and 7-4













Specification
Batch 3
Batch 4
Batch 5
Batch 6
















First in HCl
No tablet shows
Comply
Comply
Comply
Comply


0.1M for
signs of either


2 h (without
disintegration


disks)
(apart from



fragments of



coating) or



cracks that



would allow



the escape



of contents


Then
Comply with
9 min
10 min
13 min
12 min


phosphate
Eur. Ph. 2.9.1


buffer pH 6.8


for 1 h


(with disks)









Dissolution—Batch 1

Dissolution testing was performed on gastroresistant film coated tablets from a representative sample (n=6 tablets). Results are provided in Tables 34-36, which show a plateau was reached at 100% dissolution and a low variability was observed between each unit. The dissolution results comply with specification at level 1. There are no significant differences in dissolution profiles regardless of the amount of gastroresistant coating (GR) by comparing 10% GR (Batch 1-1); 12% GR (Batch 1-2); and 15% GR coating (Batch 1-3).









TABLE 34







Dissolution Results for Batch 1-1


Dissolution in HCl 0.1N/Ph 6.8 (75/25) batch 1-1 (10% GR), 20 mg pitolisant hydrochloride


















Time
Vessel
Vessel
Vessel
Vessel
Vessel
Vessel
Mean
Std.





(min)
1
2
3
4
5
6
(n = 6)
Dev.
RSD %
min
max





















0
0
0
0
0
0
0
0
0
0
0
0


60
0
0
0
0
0
0
0
0
0
0
0


120
0
0
0
0
0
0
0
0
0
0
0


125
0
0
0
0
0
0
0
0
0
0
0


130
4.7
80.4
77.9
75.4
79.3
82.9
67
30.5
45.66
5
83


135
92.8
96.4
93.5
94.4
97.3
94.7
95
1.7
1.78
93
97


140
98.8
99
97.7
97.0
100.2
97.2
98
1.2
1.24
97
100


150
99.3
99
98.0
97.4
100.5
97.2
99
1.3
1.28
97
100


165
99.3
99.4
98.2
97.6
100.6
97.4
99
1.2
1.26
97
101


180
99.5
99.7
98.4
97.4
100.9
97.3
99
1.4
1.41
97
101


195
99.3
99.7
98.5
97.5
100.8
97.6
99
1.3
1.31
97
101
















TABLE 35







Dissolution Results for Batch 1-2


Dissolution in HCl 0.1N/Ph 6.8 (75/25) batch 1-2 (12% GR), 20 mg pitolisant hydrochloride


















Time
Vessel
Vessel
Vessel
Vessel
Vessel
Vessel
Mean
Std.





(min)
1
2
3
4
5
6
(n = 6)
Dev.
RSD %
min
max





















0
0
0
0
0
0
0
0
0
0
0
0


60
0.6
0.2
0.3
0.2
0.2
1.6
1
0.6
103.79
0
2


120
0
0
1.0
0
0
0.5
0
0.4
169.65
0
1


125
0.9
0.6
0.5
0.5
1.3
0.4
1
0.4
50.37
0
1


130
0
0.3
27.3
0.8
0.8
0.6
5
10.9
220.90
0
27


135
91.4
90.5
92.6
84.9
91.3
87.3
90
2.9
3.25
85
93


140
96.3
97.0
98.2
93.8
96.3
92.6
96
2.1
2.19
93
98


150
97.6
98.6
99.2
96.6
98.1
96.5
98
1.1
1.11
97
99


165
97.6
98.2
99.8
96.7
98.6
97.0
98
1.1
1.17
97
100


180
97.8
98.5
99.7
97.1
98.8
96.5
98
1.2
1.20
97
100


195
97.8
98.6
99.7
98.0
99.1
96.6
98
1.1
1.13
97
100
















TABLE 36







Dissolution Results for Batch 1-3


Dissolution in HCl 0.1N/Ph 6.8 (75/25) batch 1-2 (12% GR), 20 mg pitolisant hydrochloride


















Time
Vessel
Vessel
Vessel
Vessel
Vessel
Vessel
Mean
Std.





(min)
1
2
3
4
5
6
(n = 6)
Dev.
RSD %
min
max





















0
0
0
0
0
0
0
0
0
0
0
0


60
0
0.3
0.5
0
1.1
0.6
0
0.4
99.33
0
1


120
0.5
0
0
0.3
0.7
0
0
0.3
122.24
0
1


125
0.5
0
0
0.6
0
0.9
0
0.4
116.60
0
1


130
7.6
57.8
4.9
3.8
10.9
0.8
14
21.6
150.76
1
58


135
91.8
91
87.5
94.2
90.7
93.1
91
2.3
2.53
87
94


140
95.6
95.4
93.5
98.9
96.6
96.8
96
1.8
1.85
94
99


150
97
97.1
96.7
99.9
98.0
99.4
98
1.4
1.41
97
100


165
97.6
97.7
96.3
101.2
98.3
100.5
99
1.9
1.90
96
101


180
97.5
97.5
96.8
101.5
99.0
100.9
99
2.0
1.99
97
101


195
98
98.4
97.1
101.7
98.8
101.9
99
2.04
2.04
97
102





An overlay of dissolution profiles for Batches 1-1 to 1-3 is provided in FIG. 4.






Dissolution—Batches 3-6

Dissolution testing was performed on gastroresistant film coated tablets from a representative sample (n=6 tablets) from batches 3-6. Results are provided in Tables 37-40. The dissolution results comply with the specifications except for Batch 4, all units exceed 10% dissolved after 120 minutes.









TABLE 37







Dissolution Results for Batch 3


Dissolution in HCl 0.1N/Ph 6.8 (75/25) Batch 3, 20 mg pitolisant hydrochloride


















Time
Vessel
Vessel
Vessel
Vessel
Vessel
Vessel
Mean
Std.





(min)
1
2
3
4
5
6
(n = 6)
Dev.
RSD %
min
max





















0
0
0
0
0
0
0
0
0
0
0
0


60
0
0
0
0
0
0.1
0
0.1
244.95
0
0


120
0.2
0.1
0
0
0
0.0
0
0.1
168.99
0
0


125
2.3
0
0
0
0
0.0
0
1.0
244.95
0
2


130
18.7
20.6
0
0.8
67.9
20.1
21
24.7
115.67
0
68


135
87.3
87.3
85.8
90.6
90.3
90.6
89
2.1
2.36
86
91


140
93.7
96.1
94.5
97.4
97.2
96.8
96
1.5
1.58
94
97


150
96.5
100.4
97.2
99.8
100.7
99.3
99
1.7
1.76
96
101


165
97.8
100.9
98.2
99.3
101.0
99.8
99
1.4
1.36
98
101


180
97.5
101.1
98.1
98.9
101.3
99.4
99
1.5
1.55
98
101


195
97.8
101.0
97.8
100.3
102.6
100.0
100
1.9
1.88
98
103
















TABLE 38







Dissolution Results for Batch 4


Dissolution in HCl 0.1N/Ph 6.8 (75/25) Batch 4, 20 mg pitolisant hydrochloride


















Time
Vessel
Vessel
Vessel
Vessel
Vessel
Vessel
Mean
Std.





(min)
1
2
3
4
5
6
(n = 6)
Dev.
RSD %
min
max





















0
0
0
0
0
0
0
0
0
0
0
0


60
77.6
10.5

0.4
103.2
7.5
40
47.2
118.46
0
103


120
90.6
46.1

28.8
105.9
49.5
64
32.5
50.69
29
106


125
86.6
46.6

30.1
99.4
49.0
62
29.3
46.98
30
99


130
88.3
49.5

71.5
100.5
52.6
72
22.1
30.56
49
100


135
91.6
55.3

89.3
101.5
81.7
84
17.4
20.81
55
101


140
96.5
88.0

96.4
101.4
93.3
95
4.9
5.20
88
101


150
97.8
99.4

98.6
101.6
98.4
99
1.5
1.52
98
102


165
98.0
100.6

98.9
101.4
98.9
100
1.4
1.38
98
101


180
97.2
100.3

98.6
101.5
98.9
99
1.6
1.65
97
101


195
97.9
100.4

98.9
101.1
99.1
99
1.3
1.28
98
101
















TABLE 39







Dissolution Results for Batch 5


Dissolution in HCl 0.1N/Ph 6.8 (75/25) Batch 5, 20 mg pitolisant hydrochloride


















Time
Vessel
Vessel
Vessel
Vessel
Vessel
Vessel
Mean
Std.





(min)
1
2
3
4
5
6
(n = 6)
Dev.
RSD %
min
max





















0
0
0
0
0
0
0
0
0
0
0
0


60
0
0
0
0
0
0
0
0
0
0
0


120
2.6
5.3
0
2.7
4.9
2.9
3
1.9
62.37
0
5


125
3.0
5.7
0.2
3.2
5.6
3.5
4
2.0
57.29
0
6


130
5.7
8.3
86.4
5.5
8.4
6.3
20
32.4
159.98
6
86


135
57.3
82.2
96.0
51.0
79.1
86.8
75
17.5
23.24
51
96


140
99.2
94.4
99.1
93.9
99.8
97.3
97
2.6
2.64
94
100


150
101.4
95.3
99.5
97.3
101.1
98.5
99
2.3
2.37
95
101


165
101.7
95.7
99.9
97.8
100.9
98.6
99
2.2
2.23
96
102


180
102.3
95.5
99.9
97.6
101.3
98.3
99
2.5
2.53
95
102


195
101.8
95.6
99.8
98.1
101.3
98.7
99
2.3
2.30
96
102
















TABLE 40







Dissolution Results for Batch 6


Dissolution in HCl 0.1N/Ph 6.8 (75/25) Batch 6, 20 mg pitolisant hydrochloride


















Time
Vessel
Vessel
Vessel
Vessel
Vessel
Vessel
Mean
Std.





(min)
1
2
3
4
5
6
(n = 6)
Dev.
RSD %
min
max





















0
0
0
0
0
0
0
0
0
0
0
0


60
0
0
0
0
0
0
0
0
0
0
0


120
1.2
0
0.5
0
0
0
0
0.5
172.88
0
1


125
1.3
0
0.7
0.6
0.3
0.3
1
0.5
88.48
0
1


130
2.0
6.0
1.7
2.0
1.0
1.0
2
1.9
82.50
1
6


135
8.9
57.4
8.3
35.8
38.4
38.4
30
18.8
62.85
8
57


140
80.1
87.8
82.3
89.4
94.2
94.2
87
5.2
5.97
80
94


150
98.1
99.1
97.3
97.6
100.9
100.9
99
1.3
1.34
97
101


165
99.2
100.2
98.8
98.5
101.2
101.2
100
1.0
1.00
99
101


180
99.3
100.0
99.1
98.7
101.3
101.3
100
0.9
0.90
99
101


195
99.7
100.4
99.2
98.8
101.7
101.7
100
1.0
1.02
99
102









An overlay of dissolution profiles for Batches 4-6 is provided in FIG. 5.


Dissolution—Batch 7

The dissolution testing was performed on Gastro resistant film coated tablets from a representative sample (n=6) of Batch 7 tablets with 10% GR (7-1), 12% GR (7-2), 15% GR (7-3), and 17% GR (7-4). The dissolution results comply with the specifications except for the batch 7-1 (10% GR coating). Results are provided in Tables 41-44.









TABLE 41







Dissolution results for Batch 7-1 (10% GR coating)


Batch 7-1 10% pelliculage GR


















Time
Vessel
Vessel
Vessel
Vessel
Vessel
Vessel
Mean
Std.





(min)
1
2
3
4
5
6
(n = 6)
Dev.
RSD %
min
max





















0
0
0
0
0
0
0
0
0.0
0




60
0.330
1.397
0.359
0.362
0.503
1.496
1
0.6
74.28
0
1


120
0.000
35.865
0.108
21.997
6.320
32.163
16
16.1
100.08
0
36


125
0.248
42.809
0.000
23.496
6.459
57.651
22
24.1
110.68
0
58


130
11.676
78.021
52.928
29.655
10.614
70.140
42
29.2
69.26
11
78


135
88.527
86.510
90.772
42.745
53.726
80.194
74
20.4
27.62
43
91


140
100.873
97.151
96.916
57.966
93.254
98.274
91
16.2
17.90
58
101


150
102.990
99.623
100.209
93.617
98.762
102.192
100
3.3
3.33
94
103


165
102.897
99.677
100.068
96.520
99.232
102.381
100
2.3
2.31
97
103


180
101.878
98.199
99.756
96.967
98.799
102.268
100
2.1
2.10
97
102


195
102.306
99.427
99.728
97.316
99.013
101.862
100
1.9
1.86
97
102
















TABLE 42







Dissolution results for Batch 7-2 (13% GR coating)


Batch 7-2 13% pelliculage GR


















Time
Vessel
Vessel
Vessel
Vessel
Vessel
Vessel
Mean
Std.





(min)
1
2
3
4
5
6
(n = 6)
Dev.
RSD %
min
max





















0
0
0
0
0
0
0
0
0.0
0




60
0.683
0.317
0.547
0.396
0.450
0.303
0
0.1
32.39
0
1


120
0.333
0.291
0.385
0.139
0.157
0.091
0
0.1
51.30
0
0


125
0.000
0.439
0.170
0.404
0.000
0.282
0
0.2
89.09
0
0


130
0.482
28.818
0.000
0.000
0.730
85.997
19
34.6
178.88
0
86


135
93.276
87.871
87.429
87.422
85.624
94.409
89
3.6
4.02
86
94


140
95.002
100.846
92.925
93.319
96.436
97.447
96
2.9
3.07
93
101


150
99.618
104.687
98.686
97.171
102.489
99.831
100
2.7
2.71
97
105


165
100.443
105.159
99.735
97.691
102.468
100.148
101
2.6
2.55
98
105


180
100.127
104.752
100.011
97.254
102.248
99.776
101
2.5
2.53
97
105


195
99.708
104.671
99.437
97.566
102.860
99.500
101
2.6
2.60
98
105
















TABLE 43







Dissolution results for Batch 7-3 (15% GR coating)


Batch 7-3 15% coating GR


















Time
Vessel
Vessel
Vessel
Vessel
Vessel
Vessel
Mean
Std.





(min)
1
2
3
4
5
6
(n = 6)
Dev.
RSD %
min
max





















0
0
0
0
0
0
0
0
0.0
0




60
0.230
0.234
0.236
0.214
0.285
0.381
0
0.1
23.70
0
0


120
0.116
0.252
0.114
0.112
0.176
0.142
0
0.1
36.05
0
0


125
0.000
0.000
0.000
0.000
0.214
0.119
0
0.1
164.10
0
0


130
72.705
34.035
0.000
0.000
0.141
0.000
18
30.1
169.17
0
73


135
90.468
71.036
83.882
90.381
88.095
85.255
85
7.3
8.57
71
90


140
93.913
87.777
82.354
97.968
96.927
93.516
92
5.9
6.46
82
98


150
99.414
100.288
97.496
101.513
99.777
99.613
100
1.3
1.31
97
102


165
99.630
101.014
97.887
101.411
100.193
99.588
100
1.3
1.25
98
101


180
98.346
100.248
96.985
101.368
99.612
99.523
99
1.5
1.53
97
101


195
98.972
100.403
96.721
101.510
99.838
99.270
99
1.6
1.62
97
102
















TABLE 44







Dissolution results for Batch 7-4 (17% GR coating)


Batch 7-4 17% coating GR


















Time
Vessel
Vessel
Vessel
Vessel
Vessel
Vessel
Mean
Std.





(min)
1
2
3
4
5
6
(n = 6)
Dev.
RSD %
min
max





















0
0
0
0
0
0
0
0
0.0
0




60
0.189
0.199
0.654
0.209
0.422
0.230
0
0.2
58.84
0
1


120
0.000
0.000
0.310
0.107
0.146
0.105
0
0.1
102.82
0
0


125
0.000
0.000
0.157
0.000
0.000
0.000
0
0.1
244.95
0
0


130
0.000
1.980
13.827
34.699
0.292
0.000
8
13.9
164.44
0
35


135
92.967
92.161
86.572
92.421
91.916
96.507
92
3.2
3.46
87
97


140
97.337
99.148
93.791
96.815
98.291
100.584
98
2.3
2.38
94
101


150
99.420
101.356
101.774
99.374
101.923
102.275
101
1.3
1.28
99
102


165
99.801
101.962
102.151
99.616
102.593
102.862
101
1.4
1.40
100
103


180
100.190
102.254
102.404
99.995
102.997
103.323
102
1.4
1.40
100
103


195
100.302
102.548
102.859
101.174
103.192
103.472
102
1.2
1.22
100
103









An overlay of dissolution profiles for Batches 7-1-7-4 is provided in FIG. 6.


Content Uniformity—Batch 1-3

Content of uniformity studies were performed on 10 units with isocratic mode for Batch 1-3 (15% GR). The acceptance value (AV) was calculated, according the Eur. Ph. 2.9.40/USP<905> on n=10 units. The content uniformity complied with the specification of Ph.Eur. 2.9.40/USP <905> (≤15.0). Results obtained are provided in Table 45, and match the observation made on dissolution results. There was no variability between tablets and a target at 100%. The content uniformity comply with the specification of Ph.Eur. 2.9.40/USP <905> (≤15.0).









TABLE 45







Content uniformity results for batch 1-3 (15% GR).













Batch 1-3 (20 mg



Content (%)
Specification
pitolisant HCl)















Tablet 1
Eur. Ph. 2.9.40
98.8



Tablet 2
USP <905>
101.9



Tablet 3
AV ≤15.0 with
97.9



Tablet 4
L1 = 15.0
100.5



Tablet 5
L2 = 25.0
99.4



Tablet 6

98.9



Tablet 7

99.7



Tablet 8

100.1



Tablet 9

101.6



Tablet 10

99.2



Mean value (%)

99.8



SD

1.2



Acceptance value (AV)

3.0










Content Uniformity—Batches 3-6

The content of uniformity was performed on 10 units with gradient mode for enteric coated tablets of Batches 3, 5, and 6. The acceptance value (AV) was calculated, according the Eur. Ph. 2.9.40/USP<905> on n=10 units. The content uniformity results presented in Table 46 comply with the specification of Ph.Eur. 2.9.40/USP <905> (≤15.0).









TABLE 46







Content uniformity results for batches 3-6 (20 mg pitolisant HCl).












Content (%)
Specification
Batch 3
Batch 4
Batch 5
Batch 6















Tablet 1
Eur. Ph. 2.9.40
100.0
99.0
96.6
98.5


Tablet 2
USP <905>
93.3
99.8
99.3
100.0


Tablet 3
AV ≤15.0 with
98.3
96.9
99.5
96.6


Tablet 4
L1 = 15.0
97.4
100.6
96.2
99.9


Tablet 5
L2 = 25.0
98.8
97.6
95.7
99.6


Tablet 6

99.4
98.3
98.1
99.8


Tablet 7

100.0
101.9
98.4
101.4


Tablet 8

96.2
98.3
98.3
99.1


Tablet 9

99.0
98.0
99.5
96.2


Tablet 10

100.8
99.4
97.8
99.3


Mean

98.3
99.0
97.9
99.0


value (%)


SD

2.2
1.5
1.4
1.6


Acceptance

5.5
3.6
3.8
3.8


value (AV)









Appearance Studies

The appearance study was performed on n=10 tablets taken from a representative sample, by visual inspection, using Batches 3, 5, and 6. The results are provided in Table 47.









TABLE 47







Appearance for Batches 3, 5, and 6












Specification
Batch 3
Batch 5
Batch 6















Appearance
White to almost white
complies
complies
complies



tablet, no damage,



no sticking trace









Average Mass Studies

Average mass studies were performed on n=10 tablets from a representative sample, by weighing, using Batches 3, 5, and 6. The results are provided in Table 48.









TABLE 48







Average mass results for batches 3, 5, and 6












Specification
Batch 3
Batch 5
Batch 6

















Average
Run and record
147.3
142.0
151.6



mass
for information










Water Content Studies

The water content of a representative sample was performed (n=3) by volumetric Karl Fischer analysis. Batches 3, 5, and 6 were studied. The results are provided in Table 49.









TABLE 49







Water content for Batches 3, 5, and 6












Specification
Batch 3
Batch 5
Batch 6















Mean water
Run and record
3.8
3.8
3.4


content (n = 3)
for information









Assay and Purity

Assay and related substances testing was performed on n=2 samples, with Batches 3, 5, and 6, using HPLC according to the parameters provided in Table 50. A gradient method was developed in order to optimize a co-elution between API (pitolisant) and triethyl citrate (TEC), an excipient present in some formulations.









TABLE 50





HPLC parameters
















Parameters
Conditions





Instrument
HPLC system with UV detection, a heater column



oven


Column
Luna II C18 Column, 250*4.6 mm, 5.0 μm particle



size


Mobile phase
Mobile phase A: triethylamine 0.6% adjusted at pH



6.0 ± 0.05



Mobile phase B: acetonitrile


Wavelength
220 nm


Flow rate
1.4 mL/min


Column
38° C. ± 2° C.


temperature


Autosampler
20° C. ± 3° C.


temperature


Injection volume
25 μL


Injection mode
Syringe draw rate mode normal


Needle cleaning
Acetonitrile/water (50 v/50 v)


solvent















Mobile phase
Mobile phase



Time (min)
A (%)
B (%)





Gradient
0
65
35



17
50
50



30
45
55



31
65
35



36
65
35











Retention time
Approx. min


of pitolisant


Run time
36 min









Assay and related substances results complied with specifications. The target of 100% assay was reached and individual impurity as well as total impurities remained <LQ. The assay and purity results are provided in Table 51.









TABLE 51







Assay and purity results.












Specification
Batch 3
Batch 5
Batch 6










Assay (%)











Sample 1
95-105
98 (98.3)
97 (97.0)
98 (98.0)


(% LC)






Sample 2

98 (98.0)
98 (98.1)
97 (96.7)


(% LC)






Mean assay

98 (98.2)
98 (97.5)
97 (97.3)


(% LC)











Related substances (%)











Each impurity
BF4 ≤0.2%
<LQ
<LQ
<LQ



BP2.941
ND
ND
ND



(N-oxide ≤0.2%






Unknown
RRT 0.60:
RRT 0.60:
RRT 0.60:



impurity ≤0.2%
0.1 (0.05)
0.1 (0.05)
0.1 (0.05)


Total
Sum of
0.1
0.1
0.1


degradation
impurities





products
(except BF6)






≤1.0%





LQ: limit of quantification (0.05%).






Summary of Quality Control Studies

For Batch 1 (amb II 3%, TEC 12%) all the results were compliant regardless of the amount of ACRYL-EZE® coating (10%, 12%, or 15%). Batch 4 (ACRYL-EZE® 10%, amb II 5%, PEG 8%) disintegration and dissolution results obtained were not compliant (disintegration: in acid media, 5 units were open after 120 mins; dissolution: all units exceed 10% dissolved after 120 min). On the other hand, Batches 3, 5, and 6 were compliant. In acid medium, the best formula obtained was Batch 3 (ACRYL-EZE® 15%, amb II 3%, PEG 8%). For Batches 3-6 the content uniformity tests were compliant, CU results showed good homogeneity of the batch. The results are summarized in Table 52 below.









TABLE 52







Initial analysis of Batches 3-6 summary.











Initial analysis
Batch 3
Batch 4
Batch 5
Batch 6





Experience
Experience 2
Experience 3
Experience 5
Experience 8


ACRYL-EZE ® 93A19346 clear
15%
10%
10%
15%


OPADRY ® amb II 88A190022 clear
 3%
 5%
 3%
 5%


Plasticizer type
PEG-8%
PEG-8%
TEC-12%
TEC-12%












Disintegration
Results in HCl 0.1M for 2 hours
Complies
Not
Complies
Complies


(n = 6)
without disks:

compliant





After 1 hour
NA
/
NA
NA



After 2 hours
NA
5 open
NA
2 tablets







swollen



in Buffer Stage (pH 6.8) for 1 hour
8 min
7 min
4 min
8 min



(with disks)







[Maximum in Min]






Dissolution
Results (% dissolved)
Complies
Not
Complies
Complies





compliant





in acid (HCl 0.1M) after 120
0 (6)
64 (5)
3 (6)
0 (6)



minutes
[0-0]
[29-106]*
[0-5]
[0-1]



Average (n) [min-max]







in Buffer Stage (pH 6.8)
99 (6)
99 (5)
99 (6)
99 (6)



Average (n) [min-max]
[96-101]
[98-102]*
[95-101]
[97-101]


CU
AV (n = 10)
5.5
3.6
3.8
3.8



Mean (%)
98.3
99.0
97.9
99.0


Water content
Mean (%)
3.8
Not
3.8
3.4





performed











According to these initial analysis results, batches 3, 5, and 6 were put in stability studies. On the basis of Batch 3, additional trials were performed.


Batch 7 was also analyzed with the colored OPADRY® amb II for supportive batch (BEIGE) and to study the variation of ACRYL-EZE® coating quantity. Disintegration studies with Batch 7 showed a compliance of all these batches to the specification with an increase of the disintegration time the more ACRYL-EZE® coating there is in the formulation.


Example 6. 1-Month Stability Test of Enteric-Coated Dose Form of Pitolisant HCl

Following a similar protocol as described in Examples 1 and 2, a cGMP batch of enteric coated 20 mg pitolisant HCl tablets was produced (Batch 8), as well as enteric coated 5 mg pitolisant HCl tablets (Batch 9).


The formula for Batch 8 and 9 are provided in Table 53.









TABLE 53





Exemplary enteric-coated dose forms with 4.45 mg and 17.8 mg pitolisant HCl
















Batch 8 (20 mg pitolisant HCl)
Batch 9 (5 mg pitolisant HCl)












Unit formula

Unit Formula



Raw Material
(17.8 mg)

(4.45 mg)



Internal phase
mg/unit
% w/w
mg/unit
% w/w





Pitolisant HCl
20
16
5
16


Microcrystalline cellulose
81
64.8
20.25
64.8


(Vivapur 102)






Crospovidone
10
8
2.5
8


(Polyplasdone XL)






Talc
10
8
2.5
8


Magnesium stearate
3
2.4
0.75
2.4


(Kemub EM-F-V)






Colloidal silica
1
0.8
0.25
0.8


Core-TOTAL
125
100
31.25
100











Coating
Coating














Opadry AMB II1
3.75

0.9375



ACRYL-EZE ® clear2
19.3125

4.828125



PEG3
1.545

0.38625



Sub-Total
24.6075

6.151875



Total
149.61

37.4025






115% suspension, weight gain = 5%;




220% suspension, weight gain = 15%;




3PEG at 8% of ACRYL-EZE ®.







A representative sample of Batch 8 tablets were subjected to 1 month stability testing. The results are provided in Table 54.









TABLE 54







1 month stability testing of enteric-coated Batch 8 tablets.










Batch Number
Batch 8
Initial
1 month















Packaging:
Description
Clinical
5° C.
25° C./60%
40° C./75%


30 tablets/20 mL HDPE

batch;

RH
RH


bottle and PP caps with

stability





desiccant (2.4 g)














TEST
ACCEPTANCE CRITERIA
RESULTS












Appearance
Light purple round
Complies
Complies
Complies
Complies



shape tablet engraved







with “C”






Identification
Complies with reference
Complies
Complies
Complies
Complies


(pitolisant







hydrochloride)







Uniformity of dosage
L1 = 15.0
Complies
NT
NT
NT


units
L2 = 25.0






Water content by KF (%)
Report
3.9%
3.9%
3.8%
3.6%


In-vitro dissolution (%)
Acid stage: 2 h-no
Complies
Complies
Complies
Complies



individual unit >10%







Buffer stage:
100% (n = 6)
100 (6)
99 (6)
100 (6)



Stage 1 no unit ≤ + 5%
[98-102]
[98-103]
[98-100]
[97-101]



(80%) after 30 min







Stage 2 and 3: see Eur.







Ph. 2.9.3./USP <711>






Assay (pitolisant) (%)
95-105%
99%
99%
99%
99%


Degradation products
Report ≤0.05%

<LD
<LD
<LD


by LC-UV (%)







BF4
≤0.2%
<LD





BF2.941
≤0.3%
<LD





Any unknown impurity
≤0.2%
<LQ





Total impurities
≤1.0%
<LQ





% Microbiological







Quality
NMT 103 CFU/g
<500 CFU/g
NT
NT
NT


TAMC
NMT 102 CFU/g
 <10 CFU/g





TYMC
Absence in 1 g







Escherichia
coli


Absence in







1 g









Additioal data is provided in FIG. 7, showing the stability dissolution profiles (means) of exemplary enteric-coated dose form (Batch 8) over 1 month, at 5° C., 25° C./60% RH, and at 40° C./75% RH.


Example 7. Manufacture of Exemplary Enteric-Coated Dosage Forms

Gastroresistant (GR) film coated tablets for the supply of a phase I clinical study (NF2 project) were manufactured. Tablets were packaged into 20 mL HDPE bottle (30 tablets per bottle) closed with PP cap fitted with desiccant 2.4 g. A batch size of 5000 g core tablets was manufactured (4519.9 g were actually obtained).


At first, two sub-lots of coating were performed with Opadry AMBII Purple. Following slight difference in color between sub-lot 1 and 2, a third sub-lot was performed as an evaluation of color tone for information purposes but was not pursued. It was inconclusive why differences in color tone were obtained when the preparation and parameters were similar between the sub-lots. The differences in tone of color were cosmetic and did not impact the critical quality attributes of the gastric resistant tablet.)


The following raw materials were used: crospovidone (Polyplasdone XL) (supplier batch number: 0002618775); Anhydrous colloidale silica (Aerosil 200 pharma) (supplier batch number: M22120016), Opadry amb II Purple, 88A200015 (supplier batch number: DT743323); Acryl eze 93A19346 clear (supplier batch number: DT738120), and cellulose microcrystalline (Vivapur 102) (supplier batch number: 56102212111). Following manufacturing, all of these raw materials were released and found compliant to the required quality standard. There was no impact on clinical batch.


Manufacturing Formula

The actual manufacturing formula of core tablets (active batch, LC23120) is described below in Table 55. For readjustment, pitolisant HCl assay and water content (98.19%) were taken into account (compensation on cellulose). The equipment used is described above in Materials and Methods.









TABLE 55







Manufacturing formula for LC23120 clinical batch.












Theoretical
Actual



Manufacturer
quantity
quantity


Raw material
batch No.
(g)
(g)













Pitolisant HCl
 001231789
814.75
814.07


Colloidal silica (aerosil 200 pharma)
 M22120016
40.00
39.93


Crospovidone (polyplasdone XL)
 0002618775
400.00
400.4


Microcrystalline cellulose
56102212111
3225.25
3225.2


(vivapur 102)





Talc (Luzenac 00)
 M21040260
400.00
400.3


Magnesium stearate
   139721
120.00
120.48


(Kemilub EM-F-V)





TOTAL
/
5000.00
5000.38









The manufacturing parameters used during blending step are provided in Table 56. The blend was sampled 5 times for blend uniformity (2 top, 1 middle, 2 bottom; about 625 mg each). Filling level was 30%, which is suitable for good mixing.









TABLE 56







Manufacturing parameters for active batch LC23120.












Required
Actual


Blending step
Parameters
value
value





Raw materials sieving
Sieve aperture
800 μm
800 μm


(about 50% cellulose +





crospovidone + talc +





API + about 50% cellulose)





Blend 1 (about 50%
Blending time
3.63*
3.63


cellulose + crospovidone





+talc + API + about
Blending speed
22
22


50% cellulose)





Raw materials sieving
Sieve aperture
800 μm
800 μm


(silica and magnesium





stearate)





Blend 2 (silica +
Blending time
3.63*
3.63


magnesium stearate
Blending speed
22
22



Filling level
For
30%




information






*3.63 (min, cent) corresponding to 03 min 38 s at 22 rpm, i.e. 80 rotations.






The yield of blending step is presented in Table 57 and is very good (98.5%).









TABLE 57







Blending step yield.











Verum batch



Attributes
LC23120














Actual used quantity (g)
5000.38



Obtained quantity (g)
4926.7



Waste quantity (g)
0



Step yield (%)
98.5










Flowability and density of the final blend (25 g) was tested. While flowability was characterized as poor, there was no issue during tableting. Relevant parameters are provided in Table 58.









TABLE 58







Flowability and density data.











Final blend




LC23120














Sample size [g]
24.9



Flowability




Flowing time [s]
02



Flowing speed [g/s]
12.5



Tapped volume [mL]




V0
64



V10
60



V50
48



 0
46



V12
/



50
/



V25




00




V10- V500




Density [g/mL]




D0
0.39



D10
0.42



D50
0.52



 0
0.54



D12
/



50




D2500




Carr index
28



Hausner index
1.39



Flow character
Poor



LOD [%]
3.74










Tableting Step

The tableting was performed on a rotative tableting press (Fette P1200), using 8 punches.


Table 59 shows tableting parameters and obtained IPC results on tablet mass, hardness and thickness. Results of friability and disintegration tests are showed in Table 60.









TABLE 59







Tableting parameters and IPC results.











Core tablets



Specifications
LC23120










Tableting actual parameters









Tableting speed (tab/h)
90 000 to confirm
90 000


Filling height (mm)
/
 7.17


Feeder speed (rpm)
20 rpm to confirm
 40*


Mean compression force (kN)
About 5 ± 1 KN (to confirm)
5.8 kN


Srel (%)

5.0%


Mean pre-comp. force (kN)
About 1.5 ± 0.5 KN (to confirm)
 1.5


Tableting run duration (min)
/
 72







IPC results









Mean tablet mass (mg)
125.0 ± 4.0% (120.0-130.0)
124.9


Min-Max mean tablet mass (mg)
T1: 125.0 ± 7.5% (115.6-134.4)
123.3-125.8


Min-Max individual tablet mass (mg)
T2: 125.0 ± 15.0% (106.3-143.8)
121.2-128.2


CV (%)

 0.80


Mean tablet hardness (N)
About 55N (for information)
 56


Min-Max mean hardness

  53-60


Min-Max indiv. Hardness

  49-66


CV (%)

 4.20


Mean tablet thickness (mm)
For information
 3.20


Min-Max mean thickness

 3.15-3.23


Min-Max indiv. Thickness

 3.13-3.25


CV (%)

 1.08





*At 15 minutes, the feeder speed was increased from 20 to 40 rpm.













TABLE 60







Disintegration and friability tests results.











Core tablets LC23120













Start
Middle
End





Disintegration time [min/s]
Min
04 min 59 s
02 min 57 s
04 min 45 s


Specification: ≤15 min (target around 04 min)
Max
05 min 35 s
05 min 21 s
05 min 07 s


Friability [%]
100 rots.
0.0%
0.0%
0.0%


Specification: ≤0.3% (target: ≤0.1%)






LOD [%] for information
/
4.08
3.68
3.72









The results of disintegration and friability IPC tests comply with the specifications. LOD is quite high, which is known with this product.


Tableting yield was more than 90%, which is very good (see Table 61A). Core tablets were split in three sub-lots for coating: 1004.8 g for sub-lot 1, 1004.8 g for sub-lot 2 and 1000.6 g for sub-lot 3 (which was ultimately not pursued further).









TABLE 61A







Tableting and global assessment.











Core tablet



Item
LC23120














Implemented quantity (g)
4926.7



Obtained quantity (g)
4519.9



Core tablet mean mass (mg)
124.8



Number of units obtained
36 217



Waste quantity (g)
352.8



Tableting yield (%)
91.7










Coating Step

The coating was performed in 330 mm perforated pan using spray nozzle of 0.8 mm. Two sub-lots were film coated in two steps: a first layer of Opadry amb II was performed (sub layer) with weight gain at 3%. Then, the gastro resistant coating was performed with acryleze with weight gain at 15%. The suspensions were prepared in excess (×2.5 for ambII, ×2.0 for acryl eze) at 15% or 20% solids for ambII and acryl eze, respectively (PEG corresponding at 8% of acryl eze). Two sub-lots of 1000 g (1004.8 g for sub-lots 1 and 2) were performed. Details are in Tables 61B and Table 62.









TABLE 61B







Suspension preparation (amb II)














Actual
Actual




Theoretical
quantity
quantity




quantity
(g)
(g)


Raw material
Batch No.
(g)
Sub-lot 1
Sub-lot 2














Opadry amb II purple
ME-23-033
75.00
75.10
75.00


88A200015






Water for injection
ME-22-064
425.00
425.3
426.6


TOTAL
LC23120
/
/
/
















TABLE 62







Suspension preparation (amb II)














LC23120
LC23120



Step
Parameters
sub-lot 1
Sub-lot 2







Amb II
Stirring speed
750-850 rpm
750-850 rpm



introduction
stirring time
02 min 50 sec
02 min 30 sec



Stirring
Stirring speed
750-850 rpm
750-850 rpm




stirring time
50 min
50 min










Amb II coating lasted 90 minutes for sub-lot 1 and 80 minutes for sub-lot 2. During sub-lot 1, spray gun was clogged after 20 minutes. Spraying was stopped and pan speed was decreased at 15 rpm during gun cleaning. Coating was re-started after 3 minutes. After 50 minutes of coating, pan speed was decreased from 28 to 26 rpm to avoid tablet loss on the side.


No issue was observed during sub-lot 2.


Coating parameters were close to target for both sub-lots and similar between sub-lots 1 and 2—details in Tables 63A and 63B.









TABLE 63A







Coating parameters (amb II).












LC23120
LC23120




sub-lot 1
sub-lot 2












Pre heat (min)
13
9










Product
Start
20.9
21.2


temperature (° C.)
End
45.6
45.0


Pan speed
Start
4.0 [intermittent]
4.0 [intermittent]


(rpm)
End
4.0 [intermittent]
4.0 [intermittent]


Actual inlet air
Start
67.2
67.1


temperature (° C.)
End
65.9
66.3


Outlet air
Start
48.5
50.5


temperature (° C.)
End
53.8
51.6


Air flow rate
Start
316
296


(m3/h)
End
298
308
















TABLE 63B







Coating parameters (amb II) continued.














LC23120
LC23120




Specifications
sub-lot 1
sub-lot 2













Coating time (min)

90
80











Product
Min
45-48
45.6
45.0


temperature (° C.)
Max

47.3
46.0


Pan speed (rpm)
Min
25 ± 5 
26.0
26.0



Max

28.0
26.0


Actual inlet air
Min
66 ± 2 
65.6
66.6


temperature (° C.)
Max

67.7
67.2


Outlet air
Min

52.0
52.0


temperature (° C.)
Max

54.3
52.9


Air flow rate
Min
300 ± 5 
293
292


(m3/h)
Max

308
304










Atomization pressure (bar)
1.2 ± 0.5
1.2
1.2


Spray width (bar)
1.6 ± 0.5
1.6
1.6











Spraying flow rate
Min
3.0 ± 1.0
3.26
3.00


(g/min)
Max

4.07
3.95










Pump setting (%)
10
9-10
10


Drying time (min)

5
5











Product
Start

47.3
46.0


temperature (° C.)
End

50
49.4


Pan speed (rpm)
Start
15
15.0
15.0



End

15.0
15.0


Actual inlet air
Start

66.1
67.6


temperature (° C.)
End

66.0
66.6


Outlet air
Start

52.8
53.4


temperature (° C.)
End

54.6
53.8


Air flow rate
Start

300
299


(m3/h)
End

297
292










Weight gain (%)

3.4
3.0


Cooling time (min)

17
20











Product
Start
<35
50.0
49.5


temperature (° C.)
End

34.9
34.9


Pan speed (rpm)
Start
4
4.0 [intermittent]
4.0 [intermittent]



End

4.0 [intermittent]
4.0 [intermittent]


Actual inlet air
Start

65.9
66.3


temperature (° C.)
End

41.8
37.9


Outlet air
Start

52.2
54.0


temperature (° C.)
End

36.4
36.3


Outlet air temperature
Start

299
302


(° C.)
End

309
308









For acryl eze coating, suspensions were prepared according to Tables 64 and 65.









TABLE 64







Suspension preparation (acryl eze).














Actual
Actual




Theoretical
quantity (g)
quantity (g)


Raw material
Batch No.
quantity (g)
Sub-lot 1
Sub-lot 2














Acryl eze clear
ME-22-083
300.00
300.4
300.2


93A19346


PEG 8000
ME-22-073
24.00
23.98
24.11


Water for

1200.00
1200.2
1201.7


injection


TOTAL
LC23120
















TABLE 65







Suspension preparation (acryl eze)












LC23120
LC23120


Step
Parameters
sub-lot 1
Sub-lot 2





Amb II
Stirring speed
1300 rpm
1200-1300 rpm


introduction
stirring time
1 min + 5 min
54 sec + 5 min


Stirring
Stirring speed
1300 rpm
1300-1900 rpm



stirring time
3 + 60 min
02 min 48 s + 73 min









The coating was performed in 330 mm perforated pan using spray nozzle of 0.8 mm. Sub-lot 1 lasted 207 minutes whereas sub-lot 2 lasted 175 minutes. During sub-lot 1, there was an issue with peristaltic pump after 180 minutes of coating. The spraying was stopped to clean the gun. Coating was re-start after few minutes. There was no particular issue during sub-lot 2. Coating parameters were close to target for the two sub-lots and similar between sub-lots 1 and 2, with details in Tables 66A and 66B.









TABLE 66







Coating parameters (acryl eze)











Specifications
LC23120 sub-lot 1
LC23120 sub-lot 2













Pre heat (min)
5
6











Product temperature (° C.)
Start

25.5
23.4



End

35.0
35.6


Pan speed (rpm)
Start
4
4.0 [intermittent]
4.0 [intermittent]



End

4.0 [intermittent]
4.0 [intermittent]


Actual inlet air temperature
Start

44.1
44.0


(° C.)
End

43.9
44.1


Outlet air temperature (° C.)
Start

36.2
36.2



End

37.6
38.1


Air flow rate (m3/h)
Start

306
303



End

300
296










Coating time (min)

207
175











Product temperature (° C.)
Min
<35° C.
32.8
32.5



Max
(target: 33° C.)
34.5
33.9


Pan speed (rpm)
Min
 25 ± 5
26.0
26.0



Max

26.0
26.0


Actual inlet air
Min
 43 ± 2
43.5
43.8


temperature (° C.)
Max

44.3
45.6


Outlet air temperature (° C.)
Min

36.7
36.6



Max

37.5
38.3


Air flow rate (m3/h)
Min
300 ± 5
292
291



Max

308
311










Atomization pressure (bar)
 1.2 ± 0.5
1.2
1.2


Spray width (bar)
 1.6 ± 0.5
1.6
1.6











Spraying flow rate (g/min)
Min
3.0-5.5 ± 0.5 
3.11
3.02



Max

5.31
6.00










Pump setting (%)
10-15
10-14
10-14


Drying time (min)

5
5











Product temperature (° C.)
Start

32.9
33.9



End

36.0
37.2


Pan speed (rpm)
Start
15 
15.0
15.0



End

15.0
15.0


Actual inlet air
Start
 43 ± 2
43.8
45.3


temperature (° C.)
End

44.1
45.3


Outlet air temperature (° C.)
Start

36.8
38.2



End

38.9
39.3


Air flow rate (m3/h)
Start
300 ± 5
307
303



End

292
306










Weight gain (%)

16.4
16.2


Cooling time (min)

8
19











Product temperature (° C.)
Start

36.0
37.2



End

32.0
31.6


Pan speed (rpm)
Start
4
4.0 [intermittent]
4.0 [intermittent]



End

4.0 [intermittent]
4.0 [intermittent]


Actual inlet air
Start

44.1
45.2


temperature (° C.)
End

36.0
33.7


Outlet air temperature (° C.)
Start

38.9
39.4



End

33.1
32.1


Air flow rate (m3/h)
Start
300 ± 5
292
303



End

297
298









The two sub-lots were compliant and had similar IPC results, as shown in Table 67.









TABLE 67







IPC results on film-coated tablets.












LC23120
LC23120


IPC results
Specifications
Sub-lot 1
Sub-lot 2










After ambII coating










After ambII coating





Mean tablet mass (mg)
128.75 ± 5.0% (122.31-135.19)
130.3
129.6


Min-Max individual tablet mass (mg)
T1: 128.75 ± 7.5% (119.09-138.41)
126.6-132.7
125.8-132.4


CV (%)
T2: 128.75 ± 15.0% (109.44-148.06)
1.26
1.40


Mean tablet hardness (N)
For information
69
67


Min-Max indiv. Hardness

64-75
60-73


CV (%)

4.61
5.56


Mean tablet thickness (mm)
For information
3.33
3.32


Min-Max indiv. Thickness

3.27-3.37
3.25-3.35


CV (%)

0.86
0.81


LOD (%)
For information
4.20
3.93







After acryl eze coating










Mean tablet mass (mg)
149.61 ± 5.0% (142.13-157.09)
149.6
148.7


Min-Max individual tablet mass (mg)
T1: 149.61 ± 7.5% (138.39-160.83)
146.1-152.8
146.2-151.6


CV (%)
T2: 149.61 ± 15.0% (127.17-172.05)
1.18
0.90


Mean tablet hardness (N)
For information
128
119


Min-Max indiv. Hardness CV (%)

114-142
114-125




5.40
2.60


Mean tablet thickness (mm)
For information
3.60
3.57


Min-Max indiv. Thickness CV (%)

3.51-3.63
3.50-3.61




0.77
0.88


LOD (%)
For information
3.46
3.72


Disintegration
No open tablet in HCl after 2 H
Compliant
Compliant









Global yield was acceptable (Table 68).









TABLE 68







Coating assessment. Reconciliation was out of specifications due to an anomaly.









Item
Code
LC23120












Remaining suspension (ambII) sub-lot 1 (g)
(7)
155.6


Remaining suspension (ambII) sub-lot 2 (g)
(8)
210.6


Remaining suspension (acryl eze) sub-lot 1 (g)
(9)
562.0


Remaining suspension (acryl eze) sub-lot 2 (g)
(10) 
647.6


Remaining suspension in solids (g)
0.15*[(7) + (8)] + 0.20*[(9) + (10)]
296.85


Other waste (g)

550.4


Total quantity of waste (g)
(1)
847.25


Sampling (g)
(2)
112.9411


Implemented quantity for blend (g)
(3)
5000.38


Implemented quantity for coating (g)
(4)
798.79


Implemented quantity for coating with core tablets (g)
(11) 
2808.39


Obtained quantity sub-lot 1 (g)

1095.8


Obtained quantity sub-lot 2 (g)

1038.4


Total obtained quantity (g)
(5)
2134.2


Remaining of core tablets (g)
(6)
2510.9


Mean mass film coated tablet (mg)

149.15


Obtained quantity (tab)

14309


Coating yield (%)
(5)/(4)*100
76.0


Global yield (%)
[(5) + (6)]/5393.76*100
86.1


Reconciliation (%)
[(5) + (6) + (1) + (2)]/[(11) + (6)]*100
105.4









Batch LC23120 was packaged into 20 mL HDPE bottles (30 tablets per bottle) closed by PP childproof resistant caps with desiccant (2.4 g) (LC23120A1) and then labelled (LC23120A2). One cardboard box of 38 bottles was prepared.


Wakix comparator (manufacturer batch number 3831701) was also packaged into 20 mL HDPE bottles (30 tablets per bottle) closed by PP childproof resistant caps with desiccant (2.4 g) and labelled (LC23177A1). One cardboard box of 33 bottles was prepared.


Pantoprazole comparator (2 labelled blisters of 14 tablets per marketed box; manufacturer batch number 535260) was labelled (LC23206A1). Five boxes of 2 blisters each were prepared.


Lubricated Blend Results

An analytical method for blend uniformity was caried out for sample preparations and quantification. As about 625 mg of powder were sampled, the preparation was identical to preparation of sample solution for 20 mg tablets.


As observed in Table 69, individual assay results on blends are homogeneous and near the target.









TABLE 69







Blend uniformity results on five units from top (n = 2), middle


(n = 1), and bottom (n = 2) of mix.










Specification
LC23120













Blend 1
90.0-110.0% (report as individual results)
99.5


Blend 2
RSD: not more than 5%
99.3


Blend 3

99.8


Blend 4

99.6


Blend 5

100.4


Average

99.7


RSD

0.4









Content Uniformity (Core Tablets)

The content of uniformity was performed on 10 units from start, 10 units from middle and 10 units from end of compression. The acceptance value (AV) was calculated, according the Ph. Eur. 2.9.40/USP <905>, on n=10 units.


The content uniformity results presented in Table 70 comply with the specification of Ph. Eur. 2.9.40/USP <905>, with an acceptance value≤15.0









TABLE 70







Content uniformity results on 10 units from


start, middle, and end of compression.









LC23120












Specification
Start
Middle
End















Tablet 1 (%)
Eur. Ph. 2.9.40
98.217
101.289
101.827


Tablet 2 (%)
USP <905>
98.371
103.788
100.648


Tablet 3 (%)

96.411
102.043
103.335


Tablet 4 (%)

98.478
101.421
101.270


Tablet 5 (%)

96.422
102.273
101.809


Tablet 6 (%)

97.444
101.662
103.551


Tablet 7 (%)

97.396
103.761
102.234


Tablet 8 (%)

97.611
101.605
102.685


Tablet 9 (%)

97.277
101.902
100.584


Tablet 10 (%)

97.010
101.601
102.233


Mean value (%)

97.5
102.1
102.0


SD

0.7
0.9
1.0


Acceptance value

2.8
2.8
2.9









Bulk Analysis—Appearance

The appearance was performed (on n=10 tablets) from a representative sample, by visual inspection, on each of the two sub-lot. A difference of coloration between both sub-lots was observed. Nevertheless, results conform to purple to light purple appearance criteria.









TABLE 71







Appearance results for batch LC23120.











Specifica-





tion
LC23120 sub-lot 1
LC23120 sub-lot 2














Appear-
For
Purple to light purple
Purple to light purple


ance
information
round shaped tablet
round shaped tablet




engraved with “C”
engraved with “C”









Bulk Analysis—Average Mass

For both sub-lot of batch LC23120, the average mass was performed (on n=10 tablets) from a representative sample, by weighing. Sub-lot 1 had a mean mass of 150.2 mg, and sub-lot 2 had a mean mass of 148.2 mg.


Bulk Analysis—Water Content

For both sub-lots of batch LC23120, the water content was performed by Karl Fischer (on n=2 tablets) from a representative sample. The water content for sub-lot 1 was 4.2%, and the water content for sub-lot 2 was 4.0%.


Bulk Analysis—Disintegration

Disintegration analysis was performed according to the Eur. Ph. 2.9.1 for sub-lot 1 and 2 of batch LC23120 during IPC (on n=6 tablets) and results were compliant.


The clinical batch LC23120, bulk of gastro resistant film coated tablet containing 20 mg of pitolisant hydrochloride, is composed of two sub-lots (as 2 runs of coating were performed, venue 1 and venue 2). During the manufacturing, it was observed that theses 2 venues had two slightly different colors (purple for venue 1 and light purple for venue 2). For investigations and to evaluate the impact, it was decided to perform disintegration time in HCL testing and QC dissolution testing for these two venues. Nevertheless, disintegration testing for sub-lot 1 and 2 of batch LC23120 were performed again during the initial analysis of the bulk tablets. The disintegration time was performed (on n=6 tablets) from a representative sample, for sub-lot 1 and 2 of batch LC23120, according to the Eur. Ph. 2.9.1. The disintegration results comply with the specifications for sub-lot 1, with no tablets opened in HCl 0.1M and are for information in phosphate buffer pH 6.8. However, disintegration results do not comply with the specifications for sub-lot 2 with 4 tablets opened in HCl 0.1M (see 011/RHS/2023).


During the 011/RHS/2023, the investigations show that the method cause (the GR tablets open in the presence of small amount of water) seems the most probable cause, but cannot be confirmed. The results of the initial analysis are not invalidated. A retest was performed on n=6 tablets for sub-lot 2 of batch LC23120 using careful cleaning and drying of the apparatus grid and the result was compliant. Before the test, the grid was washed, dried (with compressed air) and dipped in HCl during 3 seconds and 3 times (to remove residual water) before dropping the tablet on the grid. For disintegration time testing in HCl 0.1N for 2 hours, none of the tablets had shown sign of either disintegration or cracks that would allow the escape of the contents. Film coating barrier was not altered.









TABLE 72





Disintegration time results for batch LC23120 during IPC, initial analysis and retest.




















LC23120 sub-lot 1
LC23120 sub-lot 2



Specification
(IPC)
(IPC)





Disintegration
First in HCl 0.1M for 2 hours (without
Complies
Complies


time
disks):



No tablet opened



Then phosphate buffer pH 6.8 for 60
NA*
Max: 10 min



minutes (with disks):*

Min: 09 min



For Information

Mean: 10 min















LC23120 sub-lot 1
LC23120 sub-lot 2



Specification
(initial analysis)
(initial analysis)





Disintegration
First in HCl 0.1M for 2 hours (without disks):
Complies
Not Complies


time
No tablet opened

4 tablets opened*



Then phosphate buffer pH 6.8 for 60 minutes
Max: 11 min
Max: 10 min



(with disks):
Min: 08 min
Min: 01 min



For information
Mean: 10 min
Mean: 04 min
















LC23120 sub-lot 2




Specification
(retest 011/RHS/2023)







Disintegration
First in HCl 0.1M for 2 hours (without disks):
Complies



time
No tablet opened




Then phosphate buffer pH 6.8 for 60 minutes
Max: 22 min




(with disks):
Min: 09 min




For Information
Mean: 13 min










Bulk Analysis—Dissolution

The dissolution testing was performed on sub-lot 1 and 2 of bulk film-coated tablets batch LC23120 from a representative sample. Results in Tables 73 and 74.









TABLE 73







Dissolution results for batch LC23120 sub-lot 1.


LC23120 Sub-lot 1 - F/23/053


















Time
Vessel
Vessel
Vessel
Vessel
Vessel
Vessel
Mean
Std.





(min)
1
2
3
4
5
6
(n = 6)
Dev.
RSD %
min
max





















0
0
0
0
0
0
0
0
0
0
0
0


60
0.000
0.000
0.000
0.000
0.000
0.000
0
0.0
0.0
0
0


120
0.000
0.000
0.000
0.000
0.000
0.000
0
0.0
0.0
0
0


125
0.000
0.000
0.000
0.000
0.000
0.000
0
0.0
0.0
0
0


130
3.489
0.000
0.000
0.217
0.000
0.000
1
1.4
228.2
0
3


135
83.742
0.000
53.417
91.988
84.265
84.392
66
35.1
53.0
0
92


140
93.071
91.403
83.425
96.211
92.150
93.577
92
4.3
4.7
83
96


150
102.976
98.708
99.870
100.151
97.262
102.603
100
2.2
2.2
97
103


165
103.684
99.756
101.591
100.253
98.627
103.507
101
2.1
2.0
99
104


180
103.773
99.782
101.055
100.124
98.607
103.257
101
2.0
2.0
99
104


195
103.635
99.326
101.280
100.043
98.618
103.271
101
2.1
2.1
99
104









The dissolution profile for batch LC23120 sub-lot 1 is shown in FIG. 8A.









TABLE 74







Dissolution results for batch LC23120 sub-lot 2.


LC23120 sub-lot 2 - F/23/054


















Time
Vessel
Vessel
Vessel
Vessel
Vessel
Vessel
Mean
Std.





(min)
1
2
3
4
5
6
(n = 6)
Dev.
RSD %
min
max





















0
0
0
0
0
0
0
0
0
0
0
0


60
0.310
0.274
0.281
0.314
0.717
0.311
0
0.2
46.7
0
1


120
0.099
0.104
0.111
0.114
0.194
0.108
0
0.0
29.4
0
0


125
0.000
0.000
0.000
0.000
0.000
0.000
0
0.0
0.0
0
0


130
0.000
0.000
0.000
0.000
0.000
0.000
0
0.0
0.0
0
0


135
74.081
86.526
20.591
81.448
85.947
0.000
58
37.9
65.2
0
87


140
95.345
94.616
95.313
91.135
96.218
85.385
93
4.1
4.4
85
96


150
99.200
98.625
99.787
96.136
101.539
99.658
99
1.8
1.8
96
102


165
99.390
98.975
100.309
96.651
102.081
100.308
100
1.8
1.8
97
102


180
99.215
98.578
100.105
96.284
101.856
100.021
99
1.9
1.9
96
102


195
99.166
98.584
100.059
96.198
102.100
100.373
99
2.0
2.0
96
102









The dissolution profile for batch LC23120 sub-lot 2 is shown in FIG. 8B.


Bulk Analysis—Assay and Related Substances

The assay and related substances testing was performed (on n=2 samples) using sub-lot 1 and 2 tablets of bulk batch LC23120 from a representative sample. Results are given in Table 75.









TABLE 75







Assay and related substances results


for batches LC23120 sub-lots 1 and 2.










LC23120 Sub-lot 1
LC23120 Sub-lot 2











Assay









Sample 1 (% LC)
99
100


Sample 2 (% LC)
99
99


Mean assay (% LC)
99
100







Related substances









BF4
<LD
<LD


BP2.941 (N-oxide)
<LD
<LD


Unknown impurity
<LQ
<LQ


Sum of impurities
<LQ
<LQ









Release Tests on Film Coated Tablets (HDPE Bottle, 30 Units)

The appearance was performed (on n=10 tablets) from a representative sample of the batch, by visual inspection, on each of the two sub-lots and on packaged bulk. The appearance of the packaged product LC23120A1 complies with specification.









TABLE 76







Appearance results for batch LC23120A1.










Specification
LC23120A1













Appear-
Purple to light purple round
Purple to light purple round


ance
shaped tablet engraved with
shaped tablet engraved with



“C”
“C”



Diameter approx: 7 mm
7 mm



Thickness: run and record
4 mm



for information









The average mass, water content, disintegration, identification, assay and related substances, content uniformity, and dissolution were tested on the release batch and were substantively the same as bulk testing values provided above and were within the specification, and compliant.


Dissolution profile for batch LC23120A1 is also provided in FIG. 9.


The microbiology testing was performed on tablets from a representative sample, Results comply with specification.









TABLE 77







Microbiology results.










Specification
LC23120A1















TAMC
≤103 CFU/g
<500 CFU/g



TYMC
≤102 CFU/g
 <10 CFU/g



Specified organism: E. Coli
Absence in 1 g
Absence in 1 g







TAMC: Total aerobic microbial count



TYMC: Total combined yeasts and moulds count






CONCLUSION

In conclusion, blending and tableting steps went well. There was a slight difference in color between sub-lots 1 and 2 but no difference according to disintegration and dissolution testing. With regards to analytical IPC, all results were within the specification. No trend was observed on lubricated blend of mix or between start, middle and end of the compression. Results obtained for assay and uniformity of content unit were at the target. In addition, release results all comply with acceptance criteria. Disintegration was recorded only for information.


This manufacturing delivered compliant tablets: pitolisant hydrochloride (20 mg gastro-resistant film-coated tablets corresponding to pitolisant 17.8 mg as free base).


Example 8. Three-Month Stability Study

The enteric coated dosage forms from Example 7 (batch LC23120A1) were subject to a 3-month stability study. The objectives of the study were to investigate the stability of the product packaged in one packating, and to confirm the shelf-life of the product determined on the supportive batches. Data obtained after 3 months of storage at 5° C., 25° C./60% RH and 40° C./75% RH is summarized below.









TABLE 78







Pitolisant HCl enteric coated dosage forms (made in Example 7).












Unit formula
Centesimal


Raw material
Function
(mg/unit)
formula (%)













Pitolisant HCl (BF2.649)
API
20.00
16.00


Cellulose microcrystalline (Vivapur 102)
diluent
81.00
64.80


Crospovidone (Polyplasdone XL)
disintegrant
10.00
8.00


Talc
Glidant
10.00
8.00


Magnesium stearate (Kemilub EM-F-V)
Lubricant
3.00
2.40


Silica colloidal anhydrous (Aerosil 200 pharma)
Glidant
1.00
0.80


TOTAL
/
125.0
100.00


Film-coating


Purple Opadry ® AMB II1 (88A200015)
Coating agent
3.75
/


ACRYL-EZE ®2 clear (93A19346)
Coating agent
19.3125
/


PEG 80003
Plasticizer
1.545
/


TOTAL
/
149.61
/






115% suspension, weight gain = 3%




220% suspension, weight gain = 15%




3PEG at 8% of acryl eze














TABLE 79







Packaging information.













Dosage


Number of
Amount of tablets


Packaged product
strength
Date of
Type of
units per
needed for


Batch number
(as salt)
packaging
packaging
bottles
stability





LC23120A1
20 mg
Jul. 5th, 2023
HDPE bottle
30
8400





and PP cap





fitted with





2.4 g desiccant
















TABLE 80







Packaging articles characteristics.








Packaging type
Characteristics





HDPE bottle and
20 mL White HDPE bottle and white tamper evident


PP caps
childproof PP caps with desiccant (2.4 g)










Stability Program. Storage Conditions and Stability Testing


The storage of samples is performed according to ICH conditions: Long term conditions 25° C./60% RH; Intermediate conditions 30° C./65% RH; Accelerated conditions 40° C./75% RH; 5° C. for information.









TABLE 81







Storage conditions and stability testing








Storage
Test times (months)


















conditions
0.5
1
2
3
6
9
12
18
24
36
Back-up





5° C.
1
1
[1]
1
1
1
1 + 2
1
1 + 2
1 + 2
1 + 2


25° C./60% RH
1
1
[1]
1
1
1
1 + 2
1
1 + 2
1 + 2
1 + 2


30° C./65% RH

[1]1


[1]1


[1]1


[1]1


[1]1


[1]1


[1 + 2]1





[1 + 2]1



40° C./75% RH
1
1
[1]
1
1 + 2





1 + 2





—: not tested;


[ ]: optional testing;



1only if 40° C./75% RH condition fails







After removal from climatic chambers, all samples were stored at 15-25° C. before and after analysis.


Pitolisant Hydrochloride 20 mg, Gastro Resistant Film-Coated Tablets, Batch LC23120A1 After 3 Months Of Storage at 5° C., 25° C./60% RH and 40° C./75% RH

Appearance: No change in appearance was observed after 3 months at 5° C., 25° C./60% RH and 40° C./75% RH. The appearance complies with the specification.


Mean mass: A slight decrease in mean mass was observed after 3 months at 5° C. and 25° C./60% RH (respectively from 148.0 mg at initial to 146.9 mg, and 147.0 mg). No significant change in mean mass was observed after 3 months at 40° C./75% RH. The mean mass results comply with the specification.


Water content: A slight increase was observed in water content results after 3 months for all conditions, whereas a slight decrease was observed after 2 months at 25° C./60% RH and 40° C./75% RH.


Dissolution: After 3 months at 25° C./60% RH and 40° C./75% RH, a slightly slower dissolution profile was observed at the start of dissolution in buffer stage. At 5° C., no significant change was observed. The dissolution results comply with the specification. An overlay of stability dissolution profiles is provide in FIG. 10.


Disintegration time: After 3 months of storage at 5° C., no significant change was observed for the disintegration time. An increase was observed after 1 month of storage at 25° C./60% RH and 40° C./75% RH. At the 3 month time point, the disintegration was about 3 to 4 minutes longer than the initial timepoint and shows variation tablet to tablet.


Assay/Purity: Regarding the assay, no significant change was observed after 3 months at 5° C., 25° C./60% RH and 40° C./75% RH. The assay results comply with the specification.


Regarding the purity, all the individual impurities as well as the total of impurities remained<0.05%. The purity results comply with the specification.


Microbiology testing: At initial testing, the microbial results were within the specification.


Conclusion: After 3 months storage at 5° C., 25° C./60% RH and 40° C./75% RH in HDPE bottle, the 20 mg formulation of pitolisant hydrochloride gastro-resistant film-coated tablets, corresponding to pitolisant 17.8 mg free base, packaged in 20 mL HDPE bottles (30 tablets per bottle) closed with PP cap fitted with desiccant 2.4 g is stable. Based on the stability data obtained, the current 12 months shelf-life applied to the pitolisant hydrochloride 20 mg gastro-resistant film-coated tablets is compliant and stable across all storage conditions.


EQUIVALENTS

Those skilled in the art will recognize or be able to ascertain using no more than routine experimentation many equivalents to the specific embodiments disclosed herein. Those of ordinary skill in the art will appreciate that various changes or modifications to this description may be made without departing from the spirit or scope of the present disclosure, as defined in the following claims.

Claims
  • 1. An oral dosage form comprising: a core that comprises pitolisant monohydrochloride and one or more pharmaceutically acceptable excipients;an anti-moisture barrier that surrounds the core; andan enteric coating that surrounds the core and the anti-moisture barrier.
  • 2-4. (canceled)
  • 5. The oral dosage form of claim 1, wherein the enteric coating comprises ACRYL-EZE®.
  • 6. The oral dosage form of claim 5, wherein the enteric coating further comprises a plasticizer.
  • 7. The oral dosage form of claim 6, wherein the plasticizer comprises polyethylene glycol (PEG).
  • 8-9. (canceled)
  • 10. The oral dosage form of claim 1, wherein the anti-moisture barrier comprises a polyvinyl alcohol (PVA)-based polymer.
  • 11. The oral dosage form of claim 1, wherein the anti-moisture barrier comprises OPADRY® amb II.
  • 12. The oral dosage form of claim 1, comprising about 20 mg of pitolisant monohydrochloride.
  • 13. The oral dosage form of claim 1, comprising about 5 mg of pitolisant monohydrochloride.
  • 14-15. (canceled)
  • 16. The oral dosage form of claim 1, wherein the one or more pharmaceutically acceptable excipients are selected from the group consisting of microcrystalline cellulose, crospovidone, talc, magnesium stearate, and colloidal silica.
  • 17-22. (canceled)
  • 23. The oral dosage form of claim 1, wherein the pitolisant monohydrochloride is crystalline and has an X-ray diffractogram that comprises characteristic peaks (2θ) at 11.2°, 19.9°, 20.7°, and 34.1° (±0.2°).
  • 24-26. (canceled)
  • 27. The oral dosage form of claim 1, wherein the oral dosage form is a tablet.
  • 28. The oral dosage form of claim 1, wherein the oral dosage form is bioequivalent to another oral dosage form comprising pitolisant monohydrochloride in the same amount, and wherein the another oral dosage form does not comprise an anti-moisture barrier and an enteric coating.
  • 29. The oral dosage form of claim 1, wherein orally administering the oral dosage form to a subject provides a Cmax of pitolisant in the subject that is substantially the same as the Cmax of pitolisant provided by orally administering another oral dosage form to a subject comprising pitolisant monohydrochloride in the same amount, wherein the another oral dosage form does not comprise an anti-moisture barrier and an enteric coating.
  • 30. The oral dosage form of claim 1, wherein orally administering the oral dosage form once daily to a subject for a period of about 7 days provides a steady state Cmax of pitolisant that is substantially the same as the steady state Cmax of pitolisant following orally administering once daily to a subject for a period of about 7 days another oral dosage form comprising pitolisant monohydrochloride in the same amount, wherein the another oral dosage form does not comprise an anti-moisture barrier and an enteric coating.
  • 31. The oral dosage form of claim 1, wherein orally administering the oral dosage form to a subject provides an AUC of pitolisant that is substantially the same as the AUC of pitolisant obtained following orally administering another oral dosage form to a subject comprising pitolisant monohydrochloride in the same amount, wherein the another oral dosage form does not comprise an anti-moisture barrier and an enteric coating.
  • 32. The oral dosage form of claim 1, wherein orally administering the oral dosage form once daily to a subject for a period of about 7 days provides a steady state AUC of pitolisant that is substantially the same as the steady state AUC of pitolisant following orally administering once daily to a subject for a period of about 7 days another oral dosage form comprising pitolisant monohydrochloride in the same amount, wherein the another oral dosage form does not comprise an anti-moisture barrier and an enteric coating.
  • 33. The oral dosage form of claim 1, wherein orally administering the oral dosage form to a subject provides a Tmax of pitolisant that is substantially the same as the Tmax of pitolisant obtained following orally administering to a subject another oral dosage form comprising pitolisant monohydrochloride in the same amount, wherein the another oral dosage form does not comprise an anti-moisture barrier and an enteric coating.
  • 34. A method of treating excessive daytime sleepiness (EDS) or cataplexy, comprising orally administering to a subject in need thereof an oral dosage form of claim 1.
  • 35-38. (canceled)
  • 39. The method of claim 34, wherein the subject has narcolepsy.
  • 40-43. (canceled)
  • 44. An oral dosage form comprising a tablet, wherein the tablet comprises: a core that comprises crystalline pitolisant monohydrochloride and one or more pharmaceutically acceptable excipients;an anti-moisture barrier that surrounds the core, wherein the anti-moisture barrier comprises an OPADRY® polymer; andan enteric coating that surrounds the core and the anti-moisture barrier, wherein the enteric coating comprises ACRYL-EZE® and a plasticizer.
  • 45. The oral dosage form of claim 44, wherein the anti-moisture barrier comprises OPADRY® amb II.
  • 46-57. (canceled)
  • 58. The oral dosage form of claim 44, wherein the plasticizer comprises polyethylene glycol (PEG).
  • 59. The oral dosage form of claim 44, comprising about 20 mg of pitolisant monohydrochloride.
  • 60. The oral dosage form of claim 44, comprising about 5 mg of pitolisant monohydrochloride.
  • 61. The oral dosage form of claim 44, wherein the pitolisant monohydrochloride is crystalline and has an X-ray diffractogram that comprises characteristic peaks (2θ) at 11.2°, 19.9°, 20.7°, and 34.1° (±0.2°).
  • 62. The oral dosage form of claim 1, wherein the oral dosage form is bioequivalent to a WAKIX″ tablet comprising the same amount of pitolisant monohydrochloride.
  • 63. The oral dosage form of claim 44, wherein the oral dosage form is bioequivalent to a WAKIX® tablet comprising the same amount of pitolisant monohydrochloride.
  • 64. The oral dosage form of claim 44, wherein the one or more pharmaceutically acceptable excipients are selected from the group consisting of microcrystalline cellulose, crospovidone, talc, magnesium stearate, and colloidal silica.
  • 65. A method of treating excessive daytime sleepiness (EDS) or cataplexy, comprising orally administering to a subject in need thereof an oral dosage form of claim 44.
  • 66. The method of claim 65, wherein the subject has narcolepsy.
Priority Claims (1)
Number Date Country Kind
24306216.3 Jul 2024 EP regional
RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application No. 63/590,678, filed on Oct. 16, 2023, U.S. Provisional Application No. 63/616,039, filed on Dec. 29, 2023, U.S. Provisional Application No. 63/679,270, filed on Aug. 5, 2024, and European Patent Application No. 24306216.3, filed on Jul. 18, 2024, the entire contents of which are each incorporated herein by reference.

Provisional Applications (3)
Number Date Country
63590678 Oct 2023 US
63616039 Dec 2023 US
63679270 Aug 2024 US
Continuations (1)
Number Date Country
Parent 18917141 Oct 2024 US
Child 19052933 US