Methods

Information

  • Patent Grant
  • 11957791
  • Patent Number
    11,957,791
  • Date Filed
    Friday, August 30, 2019
    4 years ago
  • Date Issued
    Tuesday, April 16, 2024
    17 days ago
Abstract
The present disclosure relates to solid oral dosage forms comprising lumateperone, in free, or pharmaceutically acceptable salt form, optionally in combination with one or more additional therapeutic agents, processes for manufacture thereof and methods of use in the treatment or prophylaxis of disease.
Description
TECHNICAL FIELD

The present disclosure relates to solid oral dosage forms comprising lumateperone, in free, or pharmaceutically acceptable salt form, optionally in combination with one or more additional therapeutic agents, processes for manufacture thereof and methods of use in the treatment or prophylaxis of disease.


BACKGROUND OF THE INVENTION

The substituted heterocycle fused gamma-carbolines lumateperone (4-((6bR,10aS)-3-methyl-2,3,6b,9,10,10a-hexahydro-1H-pyrido[3′,4′: 4,5]pyrrolo[1,2,3-de]quinoxalin-8(7H)-yl)-1-(4-fluorophenyl)-1-butanone) is known to be a serotonin receptor (5-HT2A), dopamine receptor (D1 and/or D2), and serotonin transporter (SERT) ligand, which is useful in treating a variety of central nervous system disorders.


Lumateperone antagonizes the serotonin-2A (5-HT2A) receptor, and/or modulates dopamine receptor signaling at the level of key intra-cellular phosphoproteins. This compound is principally known to be useful for the treatment of positive and negative symptoms of schizophrenia, depression (especially acute depression and bipolar depression), anxiety and traumatic disorders (including acute anxiety and post-traumatic stress disorder), and dementias (including Alzheimer's disease and the symptoms associated therewith). At dopamine D2 receptors, this compound has dual properties and acts as both a post-synaptic antagonist and a pre-synaptic partial agonist of the D2 receptor. It also stimulates phosphorylation of glutamatergic NMDA NR2B, or GluN2B, receptors in a mesolimbic specific manner. It is believed that this regional selectivity in the brain areas thought to mediate the efficacy of antipsychotic drugs, together with the serotonergic, glutamatergic, and dopaminergic interactions, may result in antipsychotic efficacy for positive, negative, affective and cognitive symptoms associated with schizophrenia. The compound also exhibits serotonin reuptake inhibition, providing antidepressant activity for the treatment of schizoaffective disorder, co-morbid depression, and/or as a stand-alone treatment for major depressive disorder. Lumateperone is also useful for the treatment of bipolar disorder and other psychiatric and neurodegenerative disorders, particularly behavioral disturbances associated with dementia, autism and other CNS diseases. These features may be able to improve the quality of life of patients with schizophrenia and enhance social function to allow them to more fully integrate into their families and their workplace. Lumateperone displays differential dose-dependent effects, selectively targeting the 5-HT2A receptor at low doses, while progressively interacting with the D2 receptor at higher doses. As a result, at lower doses, it is useful in treating sleep, aggression and agitation. At a high dose, it can treat acute exacerbated and residual schizophrenia, bipolar disorders, and mood disorders.


Lumateperone, having the formula:




embedded image



is a novel therapeutic agent with potent (Ki=0.5 nM) 5-HT2A receptor antagonism, activity as a mesolimbic/mesocortical-selective dopamine receptor protein phosphorylation modulator consistent with presynaptic D2 receptor partial agonism and postsynaptic D2 receptor antagonism (Ki=32 nM) in vivo, high D1 receptor affinity (Ki=52 nM), and inhibition of the serotonin transporter (SERT) (Ki=26-62 nM, using different assays for SERT activity). Lumateperone is in Phase III clinical development as a treatment for schizophrenia, bipolar depression and agitation in dementia, including Alzheimer's Disease.


Lumateperone and related compounds have been disclosed in U.S. Pat. Nos. 6,548,493, 7,238,690, 6,552,017, 6,713,471, U.S. RE39680, and U.S. RE39679 (each of which are incorporated herein by reference) as novel compounds useful for the treatment of disorders associated with 5-HT2A receptor modulation such as anxiety, depression, psychosis, schizophrenia, sleep disorders, sexual disorders, migraine, conditions associated with cephalic pain, and social phobias. PCT/US08/03340 and U.S. Pat. No. 7,081,455, incorporated by reference herein, also disclose methods of making substituted heterocycle fused gamma-carbolines and uses of these gamma-carbolines as serotonin agonists and antagonists useful for the control and prevention of central nervous system disorders such as addictive behavior and sleep disorders. WO 2009/145900 and U.S. Pat. No. 8,598,119, and WO 2013/155506 and US 2015/0080404, each incorporated herein by reference, disclose the use of specific substituted heterocycle fused gamma-carbolines for the treatment of a combination of psychosis and depressive disorders as well as sleep, depressive and/or mood disorders in patients with psychosis or Parkinson's disease and for the treatment or prophylaxis of disorders associated with dementia, particularly behavioral or mood disturbances such as agitation, irritation, aggressive/assaultive behavior, anger, physical or emotional outbursts and psychosis and sleep disorders associated with dementia. WO 2009/114181 and U.S. Pat. No. 8,648,077, each incorporated herein by reference, disclose methods of preparing toluenesulfonic acid addition salt crystals of particular substituted heterocycle fused gamma-carbolines, e.g., toluenesulfonic acid addition salt of 4-((6bR,10aS)-3-methyl-2,3,6b,9,10,10a-hexahydro-1H-pyrido[3′,4′: 4,5]pyrrolo[1,2,3-de]quinoxalin-8(7H)-yl)-1-(4-fluorophenyl)-1-butanone.


WO 2011/133224 and U.S. Pat. No. 8,993,572, each incorporated herein by reference, disclose prodrugs/metabolites of substituted heterocycle fused gamma-carboline for improved formulation, e.g., extended/controlled release formulation. This application discloses that heterocycle fused gamma-carboline N-substituted with a 4-fluorophenyl(4-hydroxy)butyl moiety are shown to have high selectivity for the serotonin transporter (SERT) relative to the heterocycle fused gamma-carboline containing 4-fluorophenylbutanone.


WO 2009/145900 (and U.S. Pat. No. 8,598,119, incorporated herein by reference) teaches that selected substituted heterocycle fused gamma-carboline compounds have nanomolar affinity for the serotonin reuptake transporter (SERT) and so are selective serotonin reuptake inhibitors.


It has also recently been found that lumateperone may be particularly effective in treating acute depression and acute anxiety owing to its rapid onset of action compared to existing antidepressants, as disclosed in PCT/US2019/035845 (incorporated herein by reference in its entirety). This is believed to be due to its signaling through a neurotransmitter system separate from the traditional monoamine signaling systems. Lumateperone provides a dopamine D1 receptor-dependent enhancement of NMDA and AMPA currents coupled with activation of the mTOR (e.g., mTORC1) signaling pathway.


BRIEF SUMMARY OF THE INVENTION

The present disclosure provides solid oral dosage forms comprising lumateperone in free or pharmaceutically acceptable salt form. In some embodiments, the dosage form is a tablet. In some embodiments the dosage form further comprises one or more additional therapeutic agents. These dosage forms are useful for the treatment or prophylaxis of a variety of central nervous system disorders.







DETAILED DESCRIPTION

Lumateperone is a novel therapeutic agent with potent (Ki=0.5 nM) 5-HT2A receptor antagonism, activity as a mesolimbic/mesocortical-selective dopamine receptor protein phosphorylation modulator consistent with presynaptic D2 receptor partial agonism and postsynaptic D2 receptor antagonism (Ki=32 nM) in vivo, high D1 receptor affinity (Ki=52 nM), and inhibition of the serotonin transporter (SERT) (Ki=26-62 nM, using different assays for SERT activity). Lumateperone is in Phase III clinical development as a treatment for schizophrenia, bipolar depression and agitation in dementia, including Alzheimer's Disease.


The present disclosure provides a solid oral dosage form (Dosage Form 1), comprising lumateperone:




embedded image



in free or pharmaceutically acceptable salt form (e.g., in tosylate salt form), optionally wherein the dosage form is an immediate release dosage form. For example, Dosage Form 1 may be as follows:

    • 1.1. Dosage Form 1, wherein the dosage form comprises lumateperone in free base form (e.g., in free base solid amorphous dispersion form);
    • 1.2. Dosage Form 1, wherein the dosage form comprises lumateperone in pharmaceutically acceptable salt or co-crystal form;
    • 1.3. Dosage Form 1, wherein the dosage form comprises lumateperone in tosylate salt form, e.g., in one or more of mono-tosylate salt form, di-tosylate salt form, and tri-tosylate salt form;
    • 1.4. Dosage Form 1.3, wherein the dosage form comprises a combination of lumateperone in mono-tosylate salt form and lumateperone in di-tosylate salt form;
    • 1.5. Any of Dosage Forms 1 or 1.1-1.3, wherein the Dosage Form comprises lumateperone in mono-tosylate salt form;
    • 1.6. Dosage Form 1.5, wherein the lumateperone mono-tosylate is in solid crystal form, e.g., having the physical and chemical properties as disclosed in U.S. Pat. No. 8,648,077, such as one or more of the XRPD spectrum, IR spectrum, and/or DSC/TGA spectrum as disclosed therein;
    • 1.7. Dosage Form 1.5, wherein the lumateperone mono-tosylate is in solid crystal form, wherein the crystal exhibits an X-ray powder diffraction pattern comprising at least two peaks having 2-theta values selected from the group consisting of 5.68°, 12.11°, 16.04°, 17.03°, 18.16°, 19.00°, 21.67°, 22.55°, 23.48° and 24.30°, each of said peaks ±0.2°, e.g., wherein the X-ray powder diffraction data is collected on a diffractometer operating with a copper anode with a nickel filter;
    • 1.8. Dosage Form 1.5, wherein the lumateperone mono-tosylate is in solid crystal form, wherein the crystal exhibits an X-ray powder diffraction pattern comprising at least five peaks having 2-theta values selected from the group consisting of: 5.68°, 12.11°, 16.04°, 17.03°, 18.16°, 19.00°, 21.67°, 22.55°, 23.48° and 24.30°, each of said peaks ±0.2°, e.g., wherein the X-ray powder diffraction data is collected on a diffractometer operating with a copper anode with a nickel filter;
    • 1.9. Dosage Form 1.5, wherein the lumateperone mono-tosylate is in solid crystal form, wherein the crystal exhibits an X-ray powder diffraction pattern comprising the following peaks having 2-theta values: 5.6811°, 8.5140°, 11.3750°, 12.1088°, 13.3354°, 15.7948°, 16.0419°, 16.4461°, 17.0309°, 17.2606°, 17.5531°, 18.1581°, 18.9968°, 19.8889°, 20.7510°, 21.6724°, 22.25463°, 23.4815°, 23.7411°, 24.3006°, 25.9394°, 27.2321°, 28.3782°, 28.9055°, 29.6695°, 31.6106°, 32.2950°, 34.8530°, 37.5435°, 39.4972°, 40.2502° and 40.8303°, each of said peaks ±0.2°, e.g., wherein the X-ray powder diffraction data is collected on a diffractometer operating with a copper anode with a nickel filter;
    • 1.10. Any of Dosage Forms 1.3-1.5, wherein the lumateperone tosylate, e.g., the lumateperone mono-tosylate, is in solid amorphous form or is in the form of a solid amorphous dispersion.
    • 1.11. Dosage Form 1.10, wherein the lumateperone tosylate, e.g., the lumateperone mono-tosylate, is in the form of a solid amorphous dispersion comprising amorphous lumateperone tosylate in admixture with one or more excipients, e.g., stabilizing excipients.
    • 1.12. Dosage Form 1.11, wherein the dosage form comprises one or more excipients which stabilize the amorphous from of ITI-007 tosylate to prevent conversion of the amorphous form to the crystal form.
    • 1.13. Dosage Form 1.11 or 1.12, wherein the one or more excipients are selected from the group consisting of cellulose acetate, cellulose acetate phthalate, methacrylate/methyl acrylate copolymer, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, hydroxypropyl methyl cellulose acetate succinate (HPMC-AS), hydroxypropyl methyl cellulose phthalate (HPMC-P), polyvinyl acetate, polyvinyl pyrrolidone, polyvinyl pyrrolidone/vinyl acetate copolymer, and polyethylene glycol/polyvinyl acetate/polyvinylcaprolactam copolymer.
    • 1.14. Any of Dosage Forms 1.11-1.13, wherein the dosage form further comprises an anti-oxidant, e.g., selected from one or more of tocopherol, butylated hydroxytoluene (BHT), propyl gallate (OPG), ascorbic acid, butylated hydroxyanisole (BHA), tert-Butylhydroquinone (TBHQ), carotenoids, glutathione, sodium metabisulfite, sodium ethylenediaminetetraacetate, cysteine, methionine, sesamol, and citric acid.
    • 1.15. Any of Dosage Forms 1.11-1.14, wherein the dosage form further comprises a surfactant, e.g., an anionic, cationic, zwitterionic or neutral surfactant.
    • 1.16. Any of Dosage Forms 1.5-1.15, wherein the Dosage Form further comprises toluenesulfonic acid, e.g., in a molar ratio of about 1:1 to 1:2 with respect to the lumateperone mono-tosylate, e.g., 1:1 to 1:1.5 molar ratio, or 1:1 to 1:2 molar ratio, or about a 1:1 molar ratio;
    • 1.17. Dosage Form 1 or any of 1.1-1.16, wherein the Dosage Form comprises the lumateperone, in free and/or pharmaceutically acceptable salt form in a total unit amount equivalent to 0.01 to 120 mg of lumateperone free base, e.g., 0.01 to 100 mg, 0.01 to 75 mg, 0.01 to 50 mg, 0.01 to 30 mg, 0.01 to 20 mg, 0.1 to 20 mg, 5 to 20 mg, 10 to 20 mg, 10 to 30 mg, 20 to 30 mg, 20 to 50 mg, 30 mg to 50 mg, 50 to 100 mg, 1 to 75 mg, or 1 to 60 mg, or 1 to 40 mg, or 1 to 20 mg, 1 to 10 mg, 25 to 35 mg, or 35 to 45 mg, or about 6 mg, about 14 mg, or about 28 mg, or about 42 mg;
    • 1.18. Dosage Form 1 or any of 1.1-1.17, further comprising one or more pharmaceutically acceptable diluents or carriers (i.e., excipients);
    • 1.19. Dosage Form 1.18, wherein the one or more pharmaceutically acceptable diluents or carriers comprises one or more of (a) diluent/filler (e.g., cellulose or microcrystalline cellulose (e.g., silicified microcrystalline cellulose), mannitol, lactose monohydrate, dicalcium phosphate, or isomalt), (b) binder (e.g., hydroxypropyl cellulose, hydroxypropyl methyl cellulose, copovidone), (c) disintegrant (e.g., sodium starch glycolate, crospovidone or croscarmellose sodium), (d) lubricant (e.g., magnesium stearate or glyceryl monostearate), (e) glidant (e.g., silicon dioxide or talc), (f) effervescent, (g) polymer, (h) plasticizer, (i) drying agent or desiccant, (j) humectant (e.g., polyol), (k) wetting agent, (1) anti-oxidant (e.g., BHT, citric acid, propyl gallate, ascorbic acid or sodium metabisulfite), (m) thickening agent (e.g., gelling agent), (n) surfactant, (o) buffer, (p) sweetener or flavor, and (q) dye or colorant;
    • 1.20. Dosage Form 1.18, wherein the one or more pharmaceutically acceptable diluents or carriers comprises one or more hydrophilic water-soluble or water swellable polymers;
    • 1.21. Dosage Form 1.20, wherein the polymer is selected from the group consisting of natural or modified cellulosic polymers, polymers of ethylene oxide and/or propylene oxide, polymers comprising acrylic acid monomers, natural or modified gums (e.g. xanthan gum), natural or modified starches (e.g., pre-gelatinized starches), or any mixture thereof;
    • 1.22. Dosage Form 1.20, wherein the one or more pharmaceutically acceptable diluents or carriers comprises one or more hydrophobic polymers or poorly water-soluble polymers, for example, a silicone polymer, or polyalkylene polymer (e.g., polyethylene);
    • 1.23. Dosage Form 1.20, wherein the one or more pharmaceutically acceptable diluents or carriers comprises are selected from any of the following: alcohols (ethanol, glycerol, propylene glycol), gums (e.g., acacia, guar, agar, xanthan, tragacanth, karaya, gellan), polysaccharides and polysaccharide derivatives (e.g., starches, dextrans, pectins, alginates, carrageenans, cellulose, cellulose derivatives (e.g., carboxymethyl cellulose, methylcellulose, hydroxyalkyl celluloses (e.g., hydroxyethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methyl cellulose)), gelatins including non-gelling and gelling types (e.g., mammalian gelatins such as bovine gelatin, porcine gelatins, avian gelatins, fish gelatins (e.g., mixed high molecular weight and low molecular weight gelatins), synthetic polymers (e.g., polyvinylpyrrolidones, polyethylene oxide and/or polypropylene oxide polymers and copolymers (e.g., poloxamers, such as poloxamer 188), polyacrylate polymers (e.g., carbopols), polyamide polymers, sugars and sugar alcohols (e.g., dextrose, lactose, galactose, glucose, ribose, sucrose, trehalose, mannitol, maltitol, lactitol, sorbitol, xylitol, erythritol, galactitol, inositol), polypeptides/proteins, amino acids, inorganic or organic acids (e.g., citric acid, lactic acid, malic acid, gluconic acid, benzoic acid, toluenesulfonic acid, phosphoric acid, sulfuric acid, hydrochloric acid, tartaric acid, oxalic acid, cyclamic acid, ascorbic acid, methanesulfonic acid, benzenesulfonic acid, formic acid) and their salts (e.g., sodium, potassium, calcium, magnesium, lithium, ammonium salts of aforementioned acids), inorganic or organic bases (e.g., alkali metal or alkaline earth metal carbonates, bicarbonates, hydroxide, oxides), anionic surfactants (e.g., sodium lauryl sulfate, sodium laureth sulfate, sodium dodecylbenzenesulfonate, sodium lauroyl sarcosinate, sodium stearate), cationic surfactants (e.g., benzalkonium halides, cetylpyridinium halides, cetrimonium halides, benzethonium halides), zwitterionic surfactants (e.g., cocamidoalkyl betaines, such as cocamidopropyl betaine), nonionic surfactants (e.g., fatty alcohol ethoxylates (e.g., polyethylene glycol polydodecyl ethers)), sorbitan esters (e.g., sorbitan monolaurate, sorbitan monooleate, sorbitan monopalmitate, sorbitan monostearate, sorbitan tristearate), polyethoxylated sorbitan esters (e.g., polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80), and antioxidants (e.g., ascorbic acid, citric acid, ascorbyl palmitate, sodium metabisulfite, sodium sulfite, BHT, BHA, TBHQ, propyl gallate, beta-carotene, tocopherols, tocotrienols, citric acid, EDTA);
    • 1.24. Dosage Form 1 or any of 1.1-1.23, wherein the dosage form comprises or consists of (a) lumateperone tosylate (e.g., mono-tosylate), lactose monohydrate, starch (e.g., pregelatinized starch), cellulose (e.g., microcrystalline cellulose, optionally silicified), hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), copovidone (cross-linked polyvinyl pyrrolidone), sodium starch glycolate, flavors and/or colors and/or antioxidants, or (b) lumateperone tosylate (e.g., mono-tosylate), cellulose (e.g., microcrystalline cellulose, optionally silicified), hydroxypropyl cellulose (HPC), croscarmellose sodium (cross-linked carboxymethyl cellulose sodium); silicon dioxide (e.g., colloidal silicon dioxide), magnesium stearate, flavor and/or colors and/or antioxidants;
    • 1.25. Any of Dosage Forms 1.12-1.24, wherein any one or more of each said pharmaceutically acceptable carriers or diluents are present in an amount of 0.01 to 80% by weight of the Dosage Form, e.g., 0.1 to 60%, or 0.1 to 40%, or 0.1 to 30%, 0.01 to 15%, or 0.01 to 10%, or 0.1 to 20%, or 0.1 to 15% or 0.1 to 10%, or 0.5 to 10%, or 0.5 to 5%, or 1 to 5%, or 2.5 to 5%, or 1 to 3%, or 0.1 to 1%; optionally wherein the Dosage Form comprises from 60 to 90% by weight of diluent/filler, e.g., 70 to 80% diluent/filler;
    • 1.26. Any of Dosage Forms 1.12-1.25, wherein the Dosage Form comprises from 1% to 90% lumateperone, in free and/or in pharmaceutically acceptable salt form (e.g. tosylate), by weight of the composition and measured as the total content of lumateperone in all forms thereof, e.g., 1% to 80%, or 1% to 70%, or 1% to 60%, or 1% to 50%, or 1% to 40%, or 1% to 30%, or 1% to 20% or 1% to 15%, or 1% to 10%, or 1% to 5%, or 5% to 10%, or 10% to 20%, or 20 to 30%, lumateperone, in free and/or pharmaceutically acceptable salt form;
    • 1.27. Any preceding Dosage Form, wherein the Dosage Form comprises about 60 to 90% by weight of diluents/fillers (e.g., cellulose or microcrystalline cellulose (e.g., silicified microcrystalline cellulose), mannitol, lactose monohydrate, dicalcium phosphate, or isomalt), and about 1 to 10% by weight of binders (e.g., hydroxypropyl cellulose, hydroxypropyl methyl cellulose, copovidone), and about 1 to 10% by weight of disintegrants (e.g., sodium starch glycolate, crospovidone or croscarmellose sodium), and about 0.1 to 5% by weight of lubricants (e.g., magnesium stearate or glyceryl monostearate), and about 0.1 to 5% by weight of glidants (e.g., silicon dioxide or talc), and about 0.1 to 5% by weight of anti-oxidants (e.g., BHT, citric acid, propyl gallate, ascorbic acid or sodium metabisulfite);
    • 1.28. Any preceding Dosage Form, wherein the dosage form comprises one or more surface coatings, e.g., polymer surface coatings (e.g., comprising polyvinyl alcohol), optionally wherein the Dosage Form comprises 1-10% by weight of the polymer surface coating(s);
    • 1.29. Any preceding Dosage Form wherein the Dosage Form is a tablet, e.g., a spherical (e.g., round) or approximately spherical (e.g., oval or oblong) tablet;
    • 1.30. Any preceding Dosage Form wherein the Dosage Form is a caplet, e.g., a capsule-shaped tablet;
    • 1.31. Any preceding Dosage Form wherein the lumateperone is present in (a) a mean particle size of 1 to 200 μm, e.g., 1 to 150 μm, 1 to 100 μm, 1 to 50 μm, 1 to 25 μm, 1 to 15 μm, 1 to 10 μm, 5 to 10 μm, or 1 to 5 μm; and/or (b) a D90 of 100 μm or less, 50 μm or less, 25 μm or less, 15 μm or less, or 10 μm or less; and/or (c) a D10 of 50 μm or less, 25 μm or less, 15 μm or less, or 10 μm or less, or 5 μm or less;
    • 1.32. Dosage Form 1 or any of 1.1-1.31, wherein the Dosage Form is formulated for oral (gastrointestinal) administration;
    • 1.33. Dosage Form 1 or any of 1.1-1.31, wherein the Dosage Form is formulated for oral transmucosal administration, e.g., for sublingual or buccal oral disintegration;
    • 1.34. Any foregoing Dosage Form wherein the lumateperone is in combination (e.g. a fixed combination) with an effective amount of an additional therapeutic agent;
    • 1.35. Dosage Form 1.34, wherein the additional therapeutic agent is an anxiolytic or antidepressant agent;
    • 1.36. Dosage Form 1.35, wherein the anxiolytic or antidepressant agent is selected from one or more compounds in free or pharmaceutically acceptable salt form, selected from selective serotonin reuptake inhibitors (SSRIs), serotonin-norepinephrine reuptake inhibitors (SNRIs), tricyclic antidepressants (TCAs), and atypical antipsychotics, e.g. one or more compounds in free or pharmaceutically acceptable salt form, selected from:
      • (a) Selective serotonin reuptake inhibitors (SSRIs), e.g., Citalopram (Celexa), Escitalopram (Lexapro, Cipralex), Paroxetine (Paxil, Seroxat), Fluoxetine (Prozac), Fluvoxamine (Luvox) Sertraline (Zoloft, Lustral);
      • (b) Serotonin-norepinephrine reuptake inhibitors (SNRIs), e.g., Desvenlafaxine (Pristiq), Duloxetine (Cymbalta), Levomilnacipran (Fetzima), Milnacipran (Ixel, Savella), Tofenacin (Elamol, Tofacine), Venlafaxine (Effexor);
      • (c) Tricyclic antidepressants (TCAs), e.g., Amitriptyline (Elavil, Endep), Amitriptylinoxide (Amioxid, Ambivalon, Equilibrin), Clomipramine (Anafranil), Desipramine (Norpramin, Pertofrane), Dibenzepin (Noveril, Victoril), Dimetacrine (Istonil), Dosulepin (Prothiaden), Doxepin (Adapin, Sinequan), Imipramine (Tofranil), Lofepramine (Lomont, Gamanil), Melitracen (Dixeran, Melixeran, Trausabun), Nitroxazepine (Sintamil), Nortriptyline (Pamelor, Aventyl), Noxiptiline (Agedal, Elronon, Nogedal), Pipofezine (Azafen/Azaphen), Protriptyline (Vivactil), Trimipramine (Surmontil);
      • (d) Benzodiazepines, e.g., selected from 2-keto compounds (e.g., clorazepate, diazepam, flurazepam, halazepam, prazepam); 3-hydroxy compounds (lorazepam, lormetazepam, oxazepam, temazepam); 7-nitro compounds (e.g., clonazepam, flunitrazepam, nimetazepam, nitrazepam); triazolo compounds (e.g., adinazolam, alprazolam, estazolam, triazolam); and imidazo compounds (climazolam, loprazolam, midazolam);
    • 1.37. Dosage Form 1.35, wherein the additional antidepressant agent is selected from a selective serotonin reuptake inhibitor (SSRI), a serotonin reuptake inhibitor (SRI), a tricyclic antidepressant, a monoamine oxidase inhibitor, a norepinephrine reuptake inhibitor (NRI), a dopamine reuptake inhibitor (DRI), an SRI/NRI, an SRI/DRI, an NRI/DRI, an SRI/NRI/DRI (triple reuptake inhibitor), a serotonin receptor antagonist, or any combination thereof);
    • 1.38. Dosage Form 1.36, wherein the additional therapeutic agent is a NMDA receptor antagonist, for example, selected from ketamine (e.g., S-ketamine and/or R-ketamine), hydroxynorketamine, memantine, dextromethorphan, dextroallorphan, dextrorphan, amantadine, and agmatine, or any combination thereof;
    • 1.39. Any preceding Dosage Form, wherein the Dosage Form is manufactured by a dry-blending or dry-granulating process;
    • 1.40. Any preceding Dosage Form, wherein the Dosage Form is intended to be administered once daily, or twice daily, or three times daily, or every other day, or every third day;
    • 1.41. Any preceding Dosage Form, wherein the Dosage Form is packaged in a blister pack (e.g., push-through pack), e.g., a blister pack made of any suitable material (e.g., aluminum foil, polyvinyl chloride, polyvinylidene chloride, polychlorotrifluoroethylene, cyclic olefin copolymers, polyethylene, polypropylene, polyethylene terephthalate, or a combination thereof);
    • 1.42. Any preceding dosage form, wherein the Dosage Form is packaged in a bottle (e.g., plastic or glass, optionally with a screw cap lid or a child-proof lid), optionally wherein the bottle has a compartment to hold a desiccant (e.g., silica or calcium chloride);
    • 1.43. Any preceding dosage form, wherein the Dosage Form is formulated for immediate-release;
    • 1.44. Any preceding dosage form, wherein the Dosage Form is formulated for delayed or sustained release.
    • 1.45. Any preceding dosage form, wherein the dosage form does not comprise an antioxidant;
    • 1.46. Any preceding dosage form, wherein the dosage form comprises an antioxidant selected from ascorbic acid and citric acid;
    • 1.47. Any preceding dosage form, wherein the an assay (e.g., by RP HPLC) of the dosage form at or shortly after the time of manufacture demonstrates that the dosage form comprises from 90-110% of the label amount of lumateperone (in free or pharmaceutically acceptable salt form), and/or that the dosage form comprises not more than 0.5% (e.g., as measured by RP-HPLC) of any single related substance impurity and not more than 3.0% (e.g., as measured by RP-HPLC) of all related substance impurities combined;
    • 1.48. Any preceding dosage form, wherein the an assay (e.g., by RP HPLC) of the dosage form at up to three months after manufacture (e.g., 1, 2 or 3 months) after the time of manufacture demonstrates that the dosage form comprises from 90-110% of the label amount of lumateperone (in free or pharmaceutically acceptable salt form), and/or that the dosage form comprises not more than 0.5% (e.g., as measured by RP-HPLC) of any single related substance impurity and not more than 3.0% (e.g., as measured by RP-HPLC) of all related substance impurities combined, for example, when the dosage form is stored for the up the three months period at ambient temperature and humidity or at elevated temperature (e.g., 40-50° C.) and/or at elevated humidity (e.g., 60-75% relative humidity);
    • 1.49. Any preceding dosage form, wherein the dosage form dissolves in 500 mL of 0.1N aqueous hydrochloric acid to the extent of at least 75% after 15 minutes (e.g., 80-90%), and/or to the extent of at least 90% after 30 minutes (e.g., 92-98%), and/or at least 92% after 45 minutes (e.g., 95-99%).


In some embodiments, binders may include one or more of hydroxypropyl cellulose, hydroxypropyl methylcellulose, ethyl cellulose, methylcellulose, polyvinyl pyrrolidone, povidone, polyvinyl alcohol, gum arabic powder, gelatin, pullulan and the like. Each solid dosage form may comprise from 0.5-10% by weight, e.g., 1-5%, or 1-3% by weight each binder.


Carmellose calcium, croscarmellose sodium, sodium starch glycolate, crospovidone, low substituted hydroxypropyl cellulose, powdered agar and the like are used as the disintegrant. The disintegrants such as sodium starch glycolate, croscarmellose sodium and low substituted hydroxypropyl cellulose are preferable. Each tablet can contain 0.1-15% by weight, preferably 1-5% by weight of the disintegrant.


In some embodiments, the solid dosage form of the present disclosure further comprises an appropriate amount of a flavor, a lubricant, a coloring agent and the like, or various additives which are commonly used for preparing a galenic formulation. Lubricants may include magnesium stearate, calcium stearate, sucrose fatty acid ester, polyethylene glycol, talc, stearic acid, sodium stearyl fumarate and the like. Coloring agents may include the food colors such as food yellow no. 5, food red no. 2, food blue no. 2, food lake colors, iron sesquioxide and the like.


In some embodiments, a coating mixture may be applied to the solid dosage form by using a well-known method with the purpose of, for example, further masking of a taste and an odor, and preparation of an enteric formulation or a sustained-release formulation after coating a particle core with the active ingredient, one or more additives and the like. Coating mixtures may comprise any suitable water-soluble or water-swellable polymers, such as polyvinyl alcohol, polyvinyl pyrrolidone, polyethylene glycol, and polyacrylic acid, for example.


The solid dosage forms of the present disclosure include, for example, tablets, caplets, and pills. They do not include capsules or the granules used in capsules. A tablet may have a variety of shapes, including but not limited to, round, oval, square, rectangular, and oblong. Tablets and caplets may optionally be scored for easier cutting. Tablets and caplets may be coated with one, two, three or more layers designed for different purposes (e.g., taste-masking, enteric protection, delayed or sustained release, improve swallowing).


The solid dosage forms of the present disclosure may further include any one or more of pharmaceutically acceptable solvents, surface tension modifiers (e.g., surfactants), preservatives, antioxidants, colorants, taste masking agents, flavors and sweeteners. Examples of solvents include water and other solvents, which are miscible with water or solubilizing agents and suitable for oral purposes. Examples of suitable solvents are ethanol, propylene glycol, glycerol, polyethylene glycols, poloxamers, sorbitol and benzyl alcohol. In some embodiments, the aqueous solubility of the lumateperone may further be enhanced by the addition to the solution of a pharmaceutically acceptable co-solvent, a cyclodextrin or a derivative thereof (e.g., dextrans).


Preservative agents may be added to prevent the growth of microorganisms such as bacteria, yeasts and fungi in liquid formulations, which are likely to be used repeatedly. Suitable preservatives should be physicochemical stable and effective in the desired pH range. Examples of preservative agents include ethanol, methylparaben, propylparaben and benzyl alcohol.


In some embodiments, the solid dosage forms of the present disclosure include one or more anti-oxidants to guard against degradation of the active. Examples of antioxidants include propyl gallate, ascorbyl palmitate, ascorbic acid, t-butylhydroquinone (TBHQ), butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), tocopherols, tocotrienols, sodium sulfite, sodium metabisulfite, beta-carotene, citric acid and EDTA.


In some embodiments, coloring agents may be used to introduce a uniformity of appearance to the product and/or to protect any light-sensitive ingredients. Suitable coloring agents include all dyes and lakes approved by the U.S. Food and Drug Administration (e.g., FD&C colorants).


In some embodiments, sweetening agents may be used to mask unpleasant taste or to achieve a desired taste. Examples of sweetening agents are glucose, sorbitol, glycerol, acesulfame potassium and neohesperidin dihydrochalcon. The taste may be optimized further by the addition of one or more flavoring substances. Suitable flavoring substances are fruit flavors such as cherry, raspberry, black currant, lemon or strawberry flavor or other flavors such as liquorice, anise, peppermint, and caramel.


The solid dosage forms of the present disclosure may be prepared by, for example, wet granulating lumateperone, in free or pharmaceutically acceptable salt form, and one or more pharmaceutically acceptable carriers or diluents (i.e., excipients), for example, a binder and/or a disintegrant with water or a binder solution, using a machine such as a high speed mixer granulator, a fluidized-bed granulator dryer, a centrifugal tumbling fluidized-bed granulator coating machine or a kneading machine; blending or spraying a lubricant to the granules; and then subjecting to compression molding. Alternatively, the solid dosage forms of the present disclosure can be prepared by dry granulating lumateperone, in free or pharmaceutically acceptable salt form, and one or more pharmaceutically acceptable carriers or diluents (i.e., excipients), for example, a binder (a disintegrant may be further contained), using a machine such as a roller compactor; blending or spraying a disintegrant (a lubricant may be further contained) to the granules; and then subjecting to compression molding.


Suitable forms of lumateperone include the free base form, including amorphous solid dispersions thereof, pharmaceutically acceptable salt forms, including amorphous solid dispersions and crystal forms thereof, and pharmaceutically acceptable co-crystal forms. Amorphous solid dispersion forms of lumateperone free base are disclosed in patent publication WO 2018/71233, and related applications thereto, the contents of which are hereby incorporated by reference in their entireties.


Unless otherwise indicated, the term “pharmaceutically acceptable salt” includes acid addition salts between lumateperone and any pharmaceutically acceptable acid (e.g., Bronsted acid) in any molar ratio permitted by the structure of the acid. For example, “pharmaceutically acceptable salt form” of lumateperone includes the mono-hydrochloride, the di-hydrochloride, the tri-hydrochloride, the mono-tosylate, the di-tosylate and the tri-tosylate, or any mixtures thereof. In some embodiments, the lumateperone salt is a crystalline solid (e.g., a salt crystal). In some embodiments, the lumateperone may exist as a co-crystal, i.e., lumateperone free base co-crystallized with a second species. Pharmaceutically acceptable salt and co-crystal forms of lumateperone include all those forms disclosed in U.S. Pat. Nos. 8,648,077, 9,199,995, and 9,586,960, and patent publications WO 2017/1172811 and WO 2017/172784, and U.S. provisional applications 62/563,341 and 62/681,534, the contents of each of which are hereby incorporated by reference in their entireties.


In a second aspect, the present disclosure provides a process (Process 1) for the manufacture of Dosage Form 1, or any of 1.1-1.49, wherein the process comprises the steps of:

    • (a) combining lumateperone, in free or pharmaceutically acceptable salt form (e.g., tosylate salt form), with at least one diluent or carrier (e.g., with a filler, such as mannitol);
    • (b) blending and/or milling and/or granulating (e.g., dry granulating) the resulting the mixture;
    • (c) optionally filtering (e.g., screening) the resulting mixture, e.g., to achieve a uniform particle size;
    • (d) adding at least one other diluent or carrier (e.g., a disintegrant (e.g., croscarmellose sodium), or a glidant (e.g., talc), or a lubricant (e.g., magnesium stearate), or a combination thereof);
    • (e) blending and/or milling and/or granulating (e.g., dry granulating) the resulting mixture;
    • (f) optionally filtering (e.g. screening) the resulting mixture, e.g., to achieve a uniform particle size;
    • (g) pressing the mixture to form the Dosage Form;
    • (h) optionally applying one or more coatings to the Dosage Form.


It is understood that in embodiments of the present disclosure wherein lumateperone is provided in the form of an amorphous solid dispersion (either of lumateperone free base or lumateperone tosylate), that in step (a) of Process 1 it is the dispersion that is combined with at least one further diluent or carrier. As such, the amorphous solid dispersion would be prepared in a step antecedent to step (a) by combining the lumateperone and any excipients necessary to form the solid dispersion thereof.


In some embodiments, steps (d), (e), and/or (f) may be repeated for additional diluents or carriers. For example, the process steps may comprise steps (a), (b), (c), (d1), (e1), (f1), (d2), (e2), (f2), (g), and (h). The steps (d), (e), and (f) may be repeated any number of times to provide for the additional addition, blending/milling and/or filtering of any individual ingredients or portions of ingredients in order to optimize process flow. Thus, for example, in some embodiments, the binder components may be added in two or three portions, such as in steps (a), (d1) and (d2), or the lubricant may be added in a final addition step (e.g., step (d2)). In some embodiments, the process optionally further includes one or more dry granulation steps (e.g., roller compaction or slugging) which serve to increase the size of solid particles from powder-scale to granule-scale. In some embodiments, one or more blending steps may further include running the blend through a roller compactor, and optionally then milling the roller compacter ribbons. In some embodiments, any dry granulation step may be followed by a blending step to blend the resulting granules with one or more other excipients (e.g., lubricant).


In some embodiments, the final step of coating the Dosage Form is performed by suspending the un-coated Dosage Form in an aqueous suspension of coating polymer followed by drying to remove the water and any co-solvents. Optionally, the coating is applied at high temperature and/or the coated tablets are dried at high temperature (e.g., 40 to 60° C.). In some embodiments, the coating is applied by spraying an aqueous suspension of the coating polymer onto uncoated Dosage Form, followed by drying.


In a third aspect, the present disclosure provides a method (Method 1) for the treatment or prophylaxis of a disease or disorder involving or mediated by the 5-HT2A receptor, serotonin transporter (SERT), and/or dopamine D1/D2 receptor signaling pathways, comprising administering to a patient in need thereof the solid dosage form according to Dosage Form 1 or any of 1.1-1.49. In some embodiments, said disease or disorder is selected from obesity, anorexia, bulimia, depression (including major depressive disorder (MDD), acute depression, post-traumatic depression), anxiety (including acute anxiety, panic disorders, phobias, social anxiety disorder, or social withdrawal), psychosis (including acute psychosis), schizophrenia (including residual symptoms of schizophrenia, such as positive and/or negative symptoms of schizophrenia), obsessive-compulsive disorder, sexual disorders, migraine, attention deficit disorder, attention deficit hyperactivity disorder, sleep disorders, conditions associated with cephalic pain, anger disorders, agitation (including acute agitation), dementia (including Alzheimer's Disease and Parkinson's dementia), gastrointestinal disorders such as dysfunction of gastrointestinal tract motility, and bipolar disorder (e.g., bipolar depression).


The words “treatment” and “treating” are to be understood accordingly as embracing prophylaxis and treatment or amelioration of symptoms of disease and/or treatment of the cause of the disease. In particular embodiments, the words “treatment” and “treating” refer to prophylaxis or amelioration of symptoms of the disease.


The term “patient” may include a human or non-human patient.


Methods of synthesizing lumateperone and related compounds are known in art, and include the methods disclosed in WO PCT/US08/03340 (WO 2008/112280); U.S. application Ser. No. 10/786,935; U.S. Pat. Nos. 6,548,493; 7,238,690; 6,552,017; 6,713,471; 7,183,282; 8,309,722; 8,779,139; 9,315,504; U.S. RE39680, and U.S. RE39679, and WO 2015/154025, the contents of each of which are incorporated by reference in their entirety. Salts of the Compounds of the Invention may also be prepared as similarly described in U.S. Pat. Nos. 6,548,493; 7,238,690; 6,552,017; 6,713,471; 7,183,282, 8,648,077; 9,199,995; 9,588,960; U.S. RE39680; U.S. RE39679; and WO 2009/114181, the contents of each of which are incorporated by reference in their entirety.


Isolation or purification of the diastereomers of the Compounds of the Invention may be achieved by conventional methods known in the art, e.g., column purification, preparative thin layer chromatography, preparative HPLC, crystallization, trituration, simulated moving beds and the like.


The pharmaceutically acceptable salts of lumateperone can be synthesized from the parent compound, which contains basic moieties, by reaction with a suitable acid, by conventional chemical methods. Generally, such salts can be prepared by reacting the free base forms of these compounds with a stoichiometric amount of the appropriate acid in water or in an organic solvent, or in a mixture of the two; generally, non-aqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred.


Dosages employed in practicing the present disclosure will of course vary depending, e.g. on the particular disease or condition to be treated, the particular active compounds used, the mode of administration, and the therapy desired. Unless otherwise indicated, an amount of an active compound for administration (whether administered as a free base or as a salt form) refers to or is based on the amount of the compound in free form (i.e., the calculation of the amount is based on the amount of active moiety in free form, not taking into account the weight of the counter ion in the case of a salt).


For the avoidance of doubt, any disclosure of a numerical range, e.g., “up to X” amount is intended to include the upper numerical limit X. Therefore, a disclosure of “up to 60 mg” is intended to include 60 mg.


Example 1: Excipient Compatibility Study

The chemical compatibility of lumateperone monotosylate with selected excipients is studied. Excipients evaluated are (1) Fillers (silicified microcrystalline cellulose, and lactose monohydrate); (2) Disintegrants (sodium starch glycolate); (3) Binders (pregelatinized starch, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, and copovidone); and (4) Coating Polymers (polyvinyl alcohol-based film coating comprising PVA, titanium dioxide and talc). Lumateperone tosylate is mixed in a 1:1 weight ratio with each excipient and the mixture is evaluated (1) immediately after mixing, (2) after 4, 8 and 12 weeks of aging at 25° C. and 60% relative humidity, and (3) after 4, 8 and 12 weeks of accelerated aging at 40° C. and 75% relative humidity. Comparisons are made to lumateperone tosylate under the same conditions without excipient. Potency, appearance, moisture content and related substances levels are evaluated. It is found that there are no chemical incompatibilities with the selected excipients. All potency measurements of the binary mixtures show lumateperone tosylate levels comparable to the control. Under the accelerated aging conditions, slight decreases in potency are observed for both the control (90.9-93.5% potency over 4-12 weeks) and the binary mixtures, and this is believed due to air oxidation of the lumateperone tosylate. Slight increase in moisture content are observed for samples in the accelerated aging arm, with larger increases for the more hydroscopic excipients (e.g., pregelatinized starch). Related substance levels are acceptable for all binary mixtures analyzed.


Example 2: Tablet Development Process

14 mg, 28 mg, and 42 mg immediate-release film-coated tablets of lumateperone monotosylate are prepared according to the formulae shown in the tables below. Batches are prepared on a multi-kilogram scale, and each batch is prepared in three different runs:
















Ingredient
14 mg Batch









Lumateperone tosylate
7.6 wt %



Silicified microcrystalline cellulose (SMCC)
 60 wt %



Mannitol
 19 wt %



Hydroxypropyl cellulose
  3 wt %



Croscarmellose sodium
  3 wt %



BHT
0.8 wt %



Colloidal silicon dioxide
0.4 wt %



Magnesium stearate
  1 wt %



PVA Coating
4.8 wt %



Common Blend
 95 wt %



PVA Coating
  5 wt %


























Ingredient
28/42 mg Batch Common Blend







Lumateperone tosylate
 16 wt %



SMCC
 55 wt %



Mannitol
 20 wt %



Hydroxypropyl cellulose
3.2 wt %



Croscarmellose sodium
3.2 wt %



BHT
0.8 wt %



Colloidal silicon dioxide
0.4 wt %



Magnesium stearate
  1 wt %
















28 mg
42 mg




Batch
Batch







Common Blend
95 wt %
95 wt %



PVA Coating
 5 wt %
 5 wt %










For 14 mg tablets, mannitol, silicon dioxide, BHT, and lumateperone tosylate are combined in a 3-cubic foot V-blender and mixed for 5 minutes at 25 rpm. A first portion of the microcrystalline cellulose is then added, and the mixture is blended for 5 additional minutes. The blended mixture is passed through a Comil brand conical mill with a round impeller using a 1.6 mm aperture screen and milled/screened. A second portion of cellulose is milled and then combined with the milled blend and a third portion of cellulose, and the mixture is blended for 10 minutes at 25 rpm. The HPMC and croscarmellose sodium portions are pre-screened through a 20-mesh screen, then added to the blended mixture and the further blended for 5 minutes at 25 rpm. Finally, the magnesium stearate portion is pre-screened through a 30-mesh screen, added to the blended mixture, and the mixture is further blended for 3 minutes at 25 rpm.


The common blend for 28 mg and 42 mg tablets is prepared analogously using a 10 cubic foot V-blender, operated at a reduced speed of 20.5 rpm, with similar blending times.


For both the 14 mg and 28/42 mg blends, blend uniformity and physical properties are evaluated by taking samples from throughout the blender at the time final blending is stopped (10 locations are each sampled for each V-blender). Mean blend uniformity is found to be about 97% for the 14 mg batch and about 96% for the common blend batch over all three runs of each batch. Physical properties, including particle size distribution, bulk density, tapped density, and flow, are found to be highly consistent between the three runs of each batch.


From each batch, tablets are prepared using a commercial tablet press using 0.2000 inch by 0.4758-inch modified capsule embossed B tooling and tapered dies. Target weight for the 14 mg and 28 mg tablets is 250 mg, and for the 42 mg tablets 375 mg. All batches are compressed using force feed frame. Compression parameters and compression yields are evaluated for each batch run, including average tablet weight, hardness, thickness, friability, and disintegration time. All parameters are found to be compliant and consistent between the batch runs.


Tablets are then coated using a commercial multi-pan laboratory coating system using a 30-inch pan. Each batch is coated with a target weight of 5 wt % coating, using a commercial, aqueous polyvinyl alcohol coating suspension comprising 20 wt % solids, using two anti-bearding guns equipped with 1.2 mm nozzles. The coating suspension is mixed for 45 minutes in a stainless-steel tank and then allowed to de-aerate for at least 60 minutes prior to use. Target coating parameters are based on the manufacturer's guidelines. The coating process is found to be acceptable.


The coated tablets from each batch run are then tested in standard dissolution and other analytical assays. Each batch is tested in a standard dissolution study using 500 mL of 0.1N aqueous hydrochloric acid as the dissolution media. The results (batch means) are shown in the table below for 14 mg and 42 mg tablets. Results for 28 mg tablets are comparable.














Batch










14 mg tablets
42 mg tablets














1
2
3
1
2
3
















Dissolution (%) at 15 min
86
90
93
86
81
83


Dissolution (%) at 30 min
95
96
99
96
92
93


Dissolution (%) at 45 min
98
97
100
100
98
98


Dissolution (%) at 60 min
98
98
100
101
100
99


Uniformity (%) [USP 905]
99.8
100.4
99.7
100.7
100.2
98.9


Assay (%)
98
99
98
100
99
99









Example 3: Alternative Tablet Formulation and Process

Alternative, anti-oxidant free, tablet formulations are prepared according to the formulas shown below for 14-mg, 28-mg and 42-mg tablets:
















14 mg* Tablets
28 mg* Tablets
42 mg* Tablets














Weight
Weight
Weight
Weight
Weight
Weight


Ingredient
%
(mg)
%
(mg)
%
(mg)
















Lumateperone tosylate
8.0
20.0
16.0
40.0
16.0
60.0


SMCC (e.g., Prosolv HD 90)
64.2
160.5
56.2
140.5
56.2
210.75


Mannitol
20.0
50.0
20.0
50.0
20.0
75.0


Hydroxypropyl cellulose
3.2
8.0
3.2
8.0
3.2
12.0


Croscarmellose sodium
3.2
8.0
3.2
8.0
3.2
12.0


Colloidal Silicon Dioxide
0.4
1.0
0.4
1.0
0.4
1.5


Magnesium Stearate
1.0
2.5
1.0
2.5
1.0
3.75


TOTAL
100%
250
100%
250
100%
375





*equivalent weight of lumateperone free base in each tablet






14 mg and 28 mg tablets are each coated with 12.5 mg of PVA coating (5 wt % of core weight), while 42 mg tablets are coated with 18.75 mg of PVA coating (5 wt % of core weight).


Tablets are prepared, as shown in the above table, in 14 mg, 28 mg and 42 mg sizes (free base equivalent; corresponding to 20 mg, 40 mg, or 60 mg, respectively, of lumateperone tosylate). The procedure for preparing the tablets is as follows (amounts in parentheses are with reference to the total amount of indicated ingredient in the composition):

    • a. Mannitol (e.g., 50%) is added to a V-blender and is blended;
    • b. Lumateperone tosylate (100%) and additional mannitol (e.g., 50%) are added to the V-blender and blended;
    • c. SMCC (e.g., 40%) is added to the V-blender, and blended;
    • d. The preblend from step (c) is milled in a Comil conical mill; additional SMCC (e.g., 40%) is also milled in the Comil;
    • e. The milled materials from step (d) are returned to the V-blender and blended;
    • f. Croscarmellose sodium (e.g., 50%), HPC (e.g., 50%), and silicon dioxide (e.g., 50%) are added to the V-blender and blended;
    • g. Magnesium stearate (e.g., 50%) is added to the v-blender and blended;
    • h. The blend from step (g) is run through a roller compactor and milled to create granules;
    • i. The granules from step (h) are returned to the V-blender, additional croscarmellose sodium (e.g., 50%) and HPC (e.g., 50%) are added, and the mixture is blended;
    • j. Additional SMCC (e.g., 20%) is added to the v-blender and blended;
    • k. Additional magnesium stearate (e.g., 50%) is added to the v-blender and blended;
    • l. The blend is compressed to form tablets on a rotary tablet press;
    • m. The tablets are coated in a perforated coating pan.


Tablets prepared according to the above formulas are packaged in blister packs comprising PVC/PE/PCTFE film and 20-micron aluminum foil (peel-push). The packaged tablets are tested for stability using standard procedures. Tested conditions are (1) initial, (2) 50° C./ambient humidity for 1-3 months, (3) 40° C./75% relative humidity for 1-3 months, and (4) 25° C./60% relative humidity for 1 month. Tablets are assayed using reverse-phase HPLC for lumateperone tosylate content, as well as for known impurities. Tablets are also subject to a standard dissolution test (dissolution in 500 mL 0.1N aqueous hydrochloric acid). The results (batch means) are shown in the table below:




















3 mo.
3 mo.
1 mo.




50° C./
40° C./
25° C./



Initial
amb RH
75% RH
60% RH










28 mg Tablets











Assay (% of label claimed
100.0%
98.8%
97.9%
99.8%


amount)






Net Related Substance
 0.28%
0.97%
 1.1%
0.34%


Impurities (max single)
(0.15%)
(0.22%)
(0.24%)
(0.17%)


R.S. Impurities Detectable
4 (2)
9 (6)
14 (6)
4 (2)


(Quantifiable)






Dissolution (%) at 15 min
  76%
  83%
  78%
  79%


Dissolution (%) at 30 min
  90%
  94%
  90%
  91%


Dissolution (%) at 45 min
  99%
  99%
  97%
  97%


Dissolution (%) at 60 min
  104%
 100%
  99%
 101%


Moisture (%) [USP 921]
  2.6%
 1.2%
 3.0%
 2.5%







14 mg Tablets











Assay (% of label claimed
100.0%
97.9%
95.6%
100.0%


amount)






Net Related Substance
 0.44%
 1.5%
 2.3%
 0.49%


Impurities (max single)
(0.17%)
(0.29%)
(0.31%)
(0.21%)


R.S. Impurities Detectable
3 (3)
12 (7)
24 (12)
5 (3)


(Quantifiable)






Dissolution (%) at 15 min
  81%
  95%
  82%
  90%


Dissolution (%) at 30 min
  90%
 101%
  95%
  99%


Dissolution (%) at 45 min
  92%
 103%
  99%
  101%


Dissolution (%) at 60 min
  92%
 103%
 100%
  102%


Moisture (%) [USP 921]
 2.8%
 1.3%
 3.4%
  2.8%







42 mg Tablets











Assay (% of label claimed
100.8%
98.9%
97.5%
100.4%


amount)






Net Related Substance
 0.25%
 1.1%
 1.3%
 0.40%


Impurities (max single)
(0.15%)
(0.27%)
(0.28%)
(0.16%)


R.S. Impurities Detectable
4 (2)
12 (6)
19 (7)
6 (3)


(Quantifiable)






Dissolution (%) at 15 min
   82%
  85%
  73%
  79%


Dissolution (%) at 30 min
   92%
  94%
  86%
  95%


Dissolution (%) at 45 min
   96%
  98%
  93%
  100%


Dissolution (%) at 60 min
   97%
  99%
  96%
  101%


Moisture (%) [USP 921]
  2.6%
 1.3%
 3.1%
  2.7%









The lumateperone assay FIGURE is a reflection of the accuracy of the label claim, as it is presented as a percentage of the label amount (e.g., 14-mg, 28-mg or 42-mg) rather than as a percentage of the tablet composition. Acceptance of a batch requires that the batch of tablets is measured to have a mean of 90.0-110.0% of the claimed label amount of active drug.


The FIGURE for Net Related Substance Impurities indicates the percentage of all related substance impurities in the composition (as judged by HPLC peak area). Parenthetically provided is the highest percentage of any single impurity detected. Acceptance of a batch requires that total related substance impurities amount to not more than 3.0%, with no single related substance impurity amounting to more than 0.5%.


The Quantifiable R.S. (Related Substances) Impurities FIGURE is the number of distinct detectable HPLC peaks associated with related substance impurities, while the parenthetical FIGURE is for the number of such peaks above the lower limit of quantifiability. For all conditions reported in the table above, no single impurity exceeded the 0.5% acceptance limit.


The presence of increasing amounts of impurities during the test conditions reflects instability of the active ingredient, as does a drop in the Assay FIGURE. The data demonstrates that the tablets formulated according to the invention have acceptable physical and chemical stability based on all measured tested.


Example 4: Alternative Anti-Oxidants

A study is conducted to evaluate the effectiveness of the anti-oxidants propyl gallate, ascorbic acid, citric acid (anhydrous) and sodium metabisulfite. Each antioxidant is combined with either lumateperone tosylate (pure API) or with the lumateperone tosylate tablet formulation final blend of Example 3 (not pressed into tablets) in various weight ratios in amber scintillation vials. In addition, as controls, one vial holds lumateperone tosylate API and another holds the lumateperone tosylate tablet formulation final blend (42 mg strength). All vials are then stored at 60° C. for 2 weeks, 4 weeks, or 8 weeks, after which the vial contents are tested for physical appearance, HPLC potency and HPLC impurities (related substances/degradation products). The samples can be summarized as follows:














Sample
Active
Anti-oxidant



















1
Lumateperone blend
2250 mg

0


2
Lumateperone API
 362 mg

0


3

 0
Propyl Gallate
 8.16 mg


4

 0
Ascorbic Acid
170.6 mg


5

 0
Citric Acid (anhyd.)
122.4 mg


6

 0
Sodium metabisulfite
   48 mg


7
Lumateperone API
362
Propyl Gallate
 8.16 mg


8
Lumateperone API
362
Ascorbic Acid
170.6 mg


9
Lumateperone API
362
Citric Acid (anhyd.)
122.4 mg


10
Lumateperone API
362
Sodium metabisulfite
   48 mg


11
Lumateperone blend
2250 mg
Propyl Gallate
 8.16 mg


12
Lumateperone blend
2250 mg
Ascorbic Acid
170.6 mg


13
Lumateperone blend
2250 mg
Citric Acid (anhyd.)
122.4 mg


14
Lumateperone blend
2250 mg
Ascorbic Acid
 85.3 mg


15
Lumateperone blend
2250 mg
Citric Acid (anhyd.)
 61.2 mg


16
Lumateperone blend
2250 mg
Sodium metabisulfite
   24 mg









The following table provides the result of the study at 8-weeks:

















Assay
RS Impurities (%)












Sample
Appearance
(%)
Known
Unknown
Mass Bal.















1
Original (off-white
95.7
0.61
0.93
97.2



powder)






2
Granules present
99.7
0.36
None
100.0


7
Black residue and
95.5
0.65
1.1
97.3



granules






8
Original
99.8
0.42
none
100.2


9
Dark lumps present
95.5
0.62
0.59
96.7


10
Granules present
98.6
0.44
0.14
99.2


11
Black particles present
94.6
0.57
1.1
96.3


12
Original
96.4
0.60
0.36
97.4


13
Black granules present
96.8
0.59
0.60
98.0


14
Original
96.8
0.60
1.0
98.4


15
Dark lumps present
95.8
0.56
0.73
97.1


16
Grey powder
86.8
0.64
1.6
80.7









The results of the study are summarized as follows:

    • a. Both controls remain off-white powders through 8 weeks, although pure API formed some granules by 8 weeks. Pure API retains full potency at 8 weeks (99.7%), while the blend (without antioxidant) drops from 100.0% to 95.7% potency at 8 weeks.
    • b. Ascorbic acid is the only anti-oxidant which maintained full physical stability (appearance unchanged) at 8 weeks, and it was effective both for lumateperone tosylate API and for the blend formula. Sodium metabisulfite was substantially unchanged at 8 weeks when mixed with API (some granules formed), but mixed with the blend the powder changed to a grey color.
    • c. Ascorbic acid mixed with API retained full chemical potency, but the other anti-oxidants resulted in a drop in pure API potency (from 99.7% to 95.5-98.6%).
    • d. At both weight ratios, ascorbic acid mixed with the blend retained >95.7% potency, as did citric acid, but the other anti-oxidants mixed with the blend resulted in <95.7% potency at 8 weeks (and thus less than blend without antioxidants). In addition, for both ascorbic acid and citric acid mixed with blend, after 8 weeks, known and known related substances impurities were comparable to or better than the blend alone.


Overall, the study suggests that ascorbic acid is a preferred anti-oxidant for maintaining physical and chemical stability of lumateperone tosylate in a tablet formulation blend.

Claims
  • 1. A solid oral dosage form, comprising lumateperone:
  • 2. The dosage form of claim 1, wherein the dosage form further comprises lumateperone in di-tosylate salt form.
  • 3. The dosage form of claim 1, wherein the lumateperone mono-tosylate solid crystal form exhibits an X-ray powder diffraction pattern comprising at least two peaks having 2-theta values selected from the group consisting of 5.68°, 12.11°, 16.04°, 17.03°, 18.16°, 19.00°, 21.67°, 22.55°, 23.48° and 24.30°, each of said peaks ±0.2°, wherein the X-ray powder diffraction data is collected on a diffractometer operating with a copper anode with a nickel filter.
  • 4. The dosage form of claim 1, wherein the dosage form further comprises toluenesulfonic acid.
  • 5. The dosage form of claim 1, wherein the dosage form comprises the lumateperone mono-tosylate in a total unit amount equivalent to 0.01 to 120 mg of lumateperone free base.
  • 6. The dosage form of claim 1, wherein the one or more pharmaceutically acceptable diluents or carriers further comprises one or more of (f) effervescent, (g) polymer, (h) plasticizer, (i) drying agent or desiccant, (j) humectant, (k) wetting agent, (l) anti-oxidant, (m) thickening agent, (n) surfactant, (o) buffer, (p) sweetener or flavor, and (q) dye or colorant.
  • 7. The dosage form of claim 1, wherein the dosage form comprises or consists of (a) lumateperone mono-tosylate, lactose monohydrate, starch, cellulose, hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), copovidone (cross-linked polyvinyl pyrrolidone), sodium starch glycolate, flavors and/or colors and/or antioxidants, or (b) lumateperone mono-tosylate, cellulose, hydroxypropyl cellulose (HPC), croscarmellose sodium (cross-linked carboxymethyl cellulose sodium), silicon dioxide, magnesium stearate, flavor and/or colors and/or antioxidants.
  • 8. The dosage form of claim 1, wherein the dosage form comprises one or more surface coatings.
  • 9. The dosage form of claim 1, wherein the dosage form is a spherical or approximately spherical, oval or oblong tablet, or is a caplet.
  • 10. The dosage form of claim 1, wherein the lumateperone mono-tosylate is in the form of paricles having (a) a mean particle size of 1 to 200 μm, 1 to 150 μm, 1 to 100 μm, 1 to 50 μm, 1 to 25 μm, 1 to 15 μm, 1 to 10 μm, 5 to 10 μm, or 1 to 5 μm; and/or (b) a D90 of 100 μm or less, 50 μm or less, 25 μm or less, 15 μm or less, or 10 μm or less; and/or (c) a D10 of 50 μm or less, 25 μm or less, 15 μm or less, or 10 μm or less, or 5 μm or less.
  • 11. The dosage form of claim 1, wherein the lumateperone is in combination with an effective amount of an additional therapeutic agent.
  • 12. A process for the manufacture of the dosage form according to claim 1, wherein the process comprises the steps of: (a) combining lumateperone mono-tosylate, with at least one diluent or carrier;(b) blending and/or milling and/or granulating the resulting the mixture;(c) optionally filtering the resulting mixture to achieve a uniform particle size;(d) adding at least one other diluent or carrier, or a glidant, or a lubricant, or a combination thereof;(e) blending and/or milling and/or granulating the resulting mixture;(f) optionally filtering the resulting mixture to achieve a uniform particle size;(g) pressing the mixture to form the dosage form; and(h) optionally applying one or more coating to the dosage form.
  • 13. A method for the treatment or prophylaxis of a disease or disorder involving or mediated by the 5-HT2A receptor, serotonin transporter (SERT), and/or dopamine D1/D2 receptor signaling pathways, comprising administering to a patient in need thereof the solid dosage form according to claim 1.
  • 14. The dosage form of claim 1, wherein the dosage form comprises silicified microcrystalline cellulose, mannitol, hydroxypropyl cellulose, croscarmellose sodium, magnesium stearate, and silicon dioxide.
  • 15. The dosage form of claim 14, wherein the dosage form comprises 60-90% by weight of microcrystalline cellulose and mannitol, 1-10% by weight of hydroxypropyl cellulose, 1-10% by weight of croscarmellose sodium, 0.1-5% by weight of magnesium stearate, and 0.1-5% by weight of silicon dioxide.
  • 16. The dosage form of claim 15, wherein the dosage form comprises the lumateperone mono-tosylate in a total unit amount equivalent to 1 to 60 mg of lumateperone free base.
  • 17. The dosage form of claim 1, wherein the dosage form comprises the lumateperone mono-tosylate in a total unit amount equivalent to 1 to 60 mg of lumateperone free base.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a U.S. National Stage application under 35 U.S.C. § 371 of International Application No. PCT/US2019/049061, filed on Aug. 30, 2019, which claims priority to and, the benefit of, U.S. Provisional Application No. 62/725,944, filed on Aug. 31, 2018, and U.S. Provisional Application No. 62/779,920, filed on Dec. 14, 2018, the contents of each of which are hereby incorporated by reference in their entireties.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2019/049061 8/30/2019 WO
Publishing Document Publishing Date Country Kind
WO2020/047407 3/5/2020 WO A
US Referenced Citations (221)
Number Name Date Kind
1960534 Gibney May 1934 A
2490813 Hughes et al. Dec 1949 A
3299078 Pachter Jan 1967 A
3813392 Sellstedt et al. May 1974 A
3914421 Rajagopalan Oct 1975 A
4001263 Plattner et al. Jan 1977 A
4115577 Rajagopalan Sep 1978 A
4136145 Fuchs et al. Jan 1979 A
4183936 Rajagopalan Jan 1980 A
4219550 Rajagopalan Aug 1980 A
4238607 Rajagopalan Dec 1980 A
4389330 Tice et al. Jun 1983 A
4522944 Doria et al. Jun 1985 A
4530840 Tice et al. Jul 1985 A
4849246 Schmidt Jul 1989 A
4971971 Tokunaga et al. Nov 1990 A
4985432 Tokunaga et al. Jan 1991 A
5114976 Norden May 1992 A
5151419 Perenyi et al. Sep 1992 A
5538739 Bodmer et al. Jul 1996 A
5576460 Buchwald et al. Nov 1996 A
5629003 Horstmann et al. May 1997 A
5648539 Goodbrand Jul 1997 A
5648542 Goodbrand et al. Jul 1997 A
5654482 Goodbrand Aug 1997 A
5705697 Goodbrand et al. Jan 1998 A
5723669 Goodbrand et al. Mar 1998 A
5723671 Goodbrand et al. Mar 1998 A
5763476 Delbressine et al. Jun 1998 A
5834493 Gil Quintero et al. Nov 1998 A
5847166 Buchwald et al. Dec 1998 A
5902901 Goodbrand et al. May 1999 A
5948430 Zerbe et al. Sep 1999 A
6043370 Kubo et al. Mar 2000 A
6166226 Buchwald et al. Dec 2000 A
6221335 Foster Apr 2001 B1
6235936 Buchwald et al. May 2001 B1
6307087 Buchwald et al. Oct 2001 B1
6323366 Wolfe et al. Nov 2001 B1
6395916 Buchwald et al. May 2002 B1
6407092 Hester et al. Jun 2002 B1
6440710 Keinan et al. Aug 2002 B1
6448243 Kitazawa et al. Sep 2002 B1
6465693 Buchwald et al. Oct 2002 B2
6541639 Zhou et al. Apr 2003 B2
6544559 Mesens et al. Apr 2003 B2
6548493 Robichaud et al. Apr 2003 B1
6552017 Robichaud et al. Apr 2003 B1
6552024 Chen et al. Apr 2003 B1
6603008 Ando et al. Aug 2003 B1
6699852 Robichaud et al. Mar 2004 B2
6713471 Robichaud et al. Mar 2004 B1
6759554 Buchwald et al. Jul 2004 B2
6762329 Marcoux et al. Jul 2004 B2
6828314 Frank et al. Dec 2004 B2
6849619 Robichaud et al. Feb 2005 B2
6849640 Ennis et al. Feb 2005 B2
6867298 Buchwald et al. Mar 2005 B2
6884429 Koziak et al. Apr 2005 B2
6888032 Buchwald et al. May 2005 B2
6946560 Buchwald et al. Sep 2005 B2
7026498 Buchwald et al. Apr 2006 B2
7071186 Robichaud et al. Jul 2006 B2
7081455 Robichaud et al. Jul 2006 B2
7109339 Lee et al. Sep 2006 B2
7115784 Buchwald et al. Oct 2006 B2
7183282 Robichaud et al. Feb 2007 B2
7223879 Buchwald et al. May 2007 B2
RE39679 Robichaud et al. Jun 2007 E
RE39680 Robichaud et al. Jun 2007 E
7238690 Robichaud et al. Jul 2007 B2
7247731 Buchwald et al. Jul 2007 B2
7323608 Buchwald et al. Jan 2008 B2
7375226 Jolidon et al. May 2008 B2
7462641 Igo et al. Dec 2008 B2
7517990 Ito et al. Apr 2009 B2
7592454 Lee et al. Sep 2009 B2
7614727 Hori Nov 2009 B2
7645752 McDevitt et al. Jan 2010 B2
7998971 Barlow et al. Aug 2011 B2
8309722 Tomesch et al. Nov 2012 B2
8414922 Bryson et al. Apr 2013 B2
8461148 Hollander Jun 2013 B2
8598119 Mates et al. Dec 2013 B2
8603514 Yang et al. Dec 2013 B2
8648077 Tomesch et al. Feb 2014 B2
8652378 Yang et al. Feb 2014 B1
8697700 Surman et al. Apr 2014 B2
8779139 Tomesch et al. Jul 2014 B2
8791138 Seeman et al. Jul 2014 B2
8835459 Kottayil et al. Sep 2014 B2
8900497 Yang et al. Dec 2014 B2
8900498 Yang et al. Dec 2014 B2
8906277 Yang et al. Dec 2014 B2
8993572 Mates et al. Mar 2015 B2
9108340 Yang et al. Aug 2015 B2
9168258 Mates et al. Oct 2015 B2
9199995 Tomesch et al. Dec 2015 B2
9216175 Amancha et al. Dec 2015 B2
9315504 Tomesch et al. Apr 2016 B2
9371324 Mates et al. Jun 2016 B2
9393192 Yam et al. Jul 2016 B2
9427412 Bryson et al. Aug 2016 B2
9567327 Xiong et al. Feb 2017 B2
9586960 Tomesch et al. Mar 2017 B2
9616061 Mates et al. Apr 2017 B2
9708322 Li et al. Jul 2017 B2
9745300 Mates et al. Aug 2017 B2
9751883 Tomesch et al. Sep 2017 B2
9956227 Vanover et al. May 2018 B2
10077267 Mates et al. Sep 2018 B2
10117867 Mates et al. Nov 2018 B2
10221176 Tomesch et al. Mar 2019 B2
10322134 Vanover et al. Jun 2019 B2
10464938 Tomesch et al. Nov 2019 B2
10597395 Tomesch et al. Mar 2020 B2
10688097 Yao et al. Jun 2020 B2
10695345 Li Jun 2020 B2
10702522 Mates et al. Jul 2020 B2
10716786 Li et al. Jul 2020 B2
10844061 Li et al. Nov 2020 B2
10960009 Vanover et al. Mar 2021 B2
10960010 Vanover et al. Mar 2021 B2
11026951 Mates et al. Jun 2021 B2
11052084 Li Jul 2021 B2
20010008942 Buchwald et al. Jul 2001 A1
20010036472 Wong et al. Nov 2001 A1
20020155154 Wong et al. Oct 2002 A1
20030065187 Buchwald et al. Apr 2003 A1
20040019216 Buchwald et al. Jan 2004 A1
20040034015 Robichaud et al. Feb 2004 A1
20040085699 Anthony May 2004 A1
20040092534 Yam et al. May 2004 A1
20040122226 Ghosh et al. Jun 2004 A1
20040127482 Robichaud et al. Jul 2004 A1
20040138468 Buchwald et al. Jul 2004 A1
20040142970 Chung et al. Jul 2004 A1
20040180875 Lee et al. Sep 2004 A1
20040186136 Alken et al. Sep 2004 A1
20040209864 Robichaud et al. Oct 2004 A1
20040220178 Robichaud et al. Nov 2004 A1
20050166771 Gygi et al. Aug 2005 A1
20050215794 Buchwald et al. Sep 2005 A1
20050222209 Zeldis et al. Oct 2005 A1
20050222238 Alken Oct 2005 A1
20050248900 Anthony Nov 2005 A1
20050250959 Buchwald et al. Nov 2005 A1
20060148808 Robichaud et al. Jul 2006 A1
20060178362 Robichaud et al. Aug 2006 A1
20060205787 Muller et al. Sep 2006 A1
20060264673 Buchwald et al. Nov 2006 A1
20070049759 Ghosh et al. Mar 2007 A1
20070066677 Igo et al. Mar 2007 A1
20070082929 Gant et al. Apr 2007 A1
20070197695 Potyen et al. Aug 2007 A1
20070203120 McDevitt et al. Aug 2007 A1
20080069885 Mesens et al. Mar 2008 A1
20080132552 Kleinman et al. Jun 2008 A1
20080194592 Mates et al. Aug 2008 A1
20080249082 Hollander Oct 2008 A1
20080280941 Lourtie Nov 2008 A1
20080303137 Ward et al. Dec 2008 A1
20090076159 Czarnik Mar 2009 A1
20090209608 Czarnik Aug 2009 A1
20100113781 Tomesch et al. May 2010 A1
20100159033 Gant et al. Jun 2010 A1
20110020369 De Wall Malefyt et al. Jan 2011 A1
20110071080 Mates et al. Mar 2011 A1
20110112105 Tomesch May 2011 A1
20110269777 Bachurin et al. Nov 2011 A1
20120196814 Gong et al. Aug 2012 A1
20130046097 Tomesch et al. Feb 2013 A1
20130202692 Mates et al. Aug 2013 A1
20140050783 Mates Feb 2014 A1
20140088083 Hollander Mar 2014 A1
20140210117 Friesen et al. Jul 2014 A1
20140323491 Tomesch et al. Oct 2014 A1
20140364609 Tomesch et al. Dec 2014 A1
20150004237 Edgar et al. Jan 2015 A1
20150031804 Shiramizu et al. Jan 2015 A1
20150038519 Mates et al. Feb 2015 A1
20150072964 Mates Mar 2015 A1
20150079172 Mates et al. Mar 2015 A1
20150080404 Mates et al. Mar 2015 A1
20150166540 Mates et al. Jun 2015 A1
20160031885 Li et al. Feb 2016 A1
20160194325 Tomesch et al. Jul 2016 A1
20160194326 Tomesch et al. Jul 2016 A1
20160235720 Foster et al. Aug 2016 A1
20160310502 Vanover et al. Oct 2016 A1
20160354315 Li Dec 2016 A1
20170037048 Mates et al. Feb 2017 A1
20170114037 Davis et al. Apr 2017 A1
20170183350 Mates et al. Jun 2017 A1
20170189398 Mates et al. Jul 2017 A1
20170283417 Li et al. Oct 2017 A1
20180044337 Tomesch et al. Feb 2018 A1
20180200256 Vanover et al. Jul 2018 A1
20190062334 Mates et al. Feb 2019 A1
20190071445 Li et al. Mar 2019 A1
20190112310 Li et al. Apr 2019 A1
20190183888 Mates et al. Jun 2019 A1
20190211015 Mittelman et al. Jul 2019 A1
20190218219 Tomesch et al. Jul 2019 A1
20190231780 Yao et al. Aug 2019 A1
20190290655 Vanover et al. Sep 2019 A1
20190292185 Tomesch et al. Sep 2019 A1
20190298730 Vanover et al. Oct 2019 A1
20190328745 Vanover et al. Oct 2019 A1
20190388418 Li Dec 2019 A1
20200017499 Mates et al. Jan 2020 A1
20200087305 Tomesch et al. Mar 2020 A1
20200102304 Li et al. Apr 2020 A1
20200102310 Li et al. Apr 2020 A1
20200115380 Tomesch et al. Apr 2020 A1
20200157100 Li May 2020 A1
20200392135 Wennogle et al. Dec 2020 A1
20200405713 Mates et al. Dec 2020 A1
20200407362 Mates et al. Dec 2020 A1
20210002280 Mates et al. Jan 2021 A1
20210032247 Li et al. Feb 2021 A1
Foreign Referenced Citations (30)
Number Date Country
0 058 481 Aug 1982 EP
0 856 508 Aug 1998 EP
0 976 732 Feb 2000 EP
1 245 553 Oct 2002 EP
1 254 884 Nov 2002 EP
1 539 115 Jun 2005 EP
1 564 671 Aug 2005 EP
1476087 Jun 1977 GB
2145422 Mar 1985 GB
2465267 Oct 2012 RU
WO 1995013814 May 1995 WO
WO 1995026325 Oct 1995 WO
WO 1999043643 Sep 1999 WO
WO 2000002887 Jan 2000 WO
WO 2000048610 Aug 2000 WO
WO 2000064899 Nov 2000 WO
WO 2000077001 Dec 2000 WO
WO 2000077002 Dec 2000 WO
WO 2000077010 Dec 2000 WO
WO 2002059129 Aug 2002 WO
WO 2003014118 Feb 2003 WO
WO 2004064738 Aug 2004 WO
WO 2005030214 Apr 2005 WO
WO 2006081332 Aug 2006 WO
WO 2014110322 Jul 2014 WO
WO 2017117514 Jul 2017 WO
WO-2018031535 Feb 2018 WO
WO 2018106916 Jun 2018 WO
WO-2018189646 Oct 2018 WO
WO 2019102240 May 2019 WO
Non-Patent Literature Citations (96)
Entry
“Study of a Novel Antipsychotic ITI-007 in Schizophrenia,” Clinical Trials.gov, 6 pages, Dec. 26, 2011.
Alvir, et al., “Clozapine-Induced Agranulocytosis,” The New England Journal of Medicine, vol. 329, No. 3, pp. 162-167, (1993).
Angst et al. “Prevalence and Characteristics of Undiagnosed Bipolar Disorders in Patients With a Major Depressive Episode”, Arch Gen Psychiatry, vol. 68, No. 8, pp. 701-709, (2011).
Baille, T.A., “The Use of Stable Isotopes in Pharmacological Research,” Pharmacol. Reviews, vol. 33, No. 2, pp. 81-132, (1981).
Balbach, et al. “Pharmaceutical evaluation of early development candidates 'the 100 mg-approach”, International Journal of Pharmaceutics, vol. 275, pp. 1-12 (2004).
Bastin, “Salt Selection and Optimized Procedures for Pharmaceutical New Chemical Entities”, Organic Process and Research Development, vol. 4, No. 5, pp. 427-435 (2000).
Bechtold, D.A., et al., “Circadian Dysfunction in Disease,” Trends in Pharmacological Sciences,31(5): 191-198, (2010); Abstract.
Bennett, et al., “Cecil Textbook of Medicine,” 20th Edition, vol. 1, pp. 1004-1010, (1996).
Borghans et al., “Animal Models for Posttraumatic Stress Disorder: An Overview of What is Used in Research,” World J. Psychiatr., vol. 5, No. 4, pp. 387-396, (2015); DOI: 10.5498/wjp.v5.i4.387.
Bremner, et al., “Neuroimaging of Posttraumatic Stress Disorder”, Psychiatric Annals Journal, vol. 28, No. 8, p. 445-450, (1998).
Browne, T.R., “Stable Isotope Techniques in Early Drug Development: An Economic Evaluation,” J. Clin. Pharmacol., vol. 38, pp. 213-220, (1998).
Bryan-Lluka, et al., “Potencies of Haloperidol Metabolites as Inhibitors of the Human Noradrenaline, Dopamine and Serotonin Transporters in Transfected COS-7 Cells,” Naunyn-Shemiedeberg's Arch Pharmacol, vol. 360, pp. 109-115, (1999).
Byrn, “Pharmaceutical Solids: A Strategic Approach to Regulatory Considerations”, vol. 12, No. 7, p. 945-954 (1995).
Caira, M.R., “Crystalline Polymorphism of Organic Compounds,” Topics in Current Chemistry, vol. 198, p. 163-203, (1998).
Cherrah, et al., “Study of Deuterium Isotope Effects on Protein Binding by Gas Chromatography/Mass Spectrometry. Caffeine and Deuterated Isotopomers,” Biomedical and Environmental Mass Spectrometry, vol. 14, pp. 653-657, (1987).
Darmani, et al., “Do Functional Relationships Exist Between 5-HT1A and 5-HT2 Receptors?” Pharmacology and Biochemistry & Behavior, vol. 36, pp. 901-906, (1990).
Davis et al., “ITI-007: A Novel Treatment for Behavioral Disturbances Associated with Dementia and Related Disorders,” Clinical Trials in Alzheimer's Disease (CTAD) Congress 2014 (2014) (poster presentation).
Davis, et al., “ITI-007 demonstrates brain occupancy at serotonin 5-HT2A and dopamine D2 receptors and serotonin transporters using positron emission tomography in healthy volunteers,” Psychopharmacology, vol. 232, pp. 2863-2872, (2015); DOI: 10.1007/s00213-015-3922-1.
Davis, et al., “ITI-007 in the Treatment of Schizophrenia: From Novel Pharmacology to Clinical Outcomes,” Expert Review of Neurotherapeutics, vol. 16, No. 6, pp. 601-614, (2016).
Davis, et al., “Lumateperone (ITI-007), A Novel Drug in Development for the Treatment of Agitation in Patients with Dementia, including Alzheimer's Disease: Rationale and Clinical Design,” The Journal of Prevention of Alzheimer's Disease, 4(4): 372 (2017) (Clinical Trials in Alzheimer's Disease (CTAD) Congress, Symposium Summary P93.
Davis et al., “Rationale for the Development of Low Doses of ITI-007 for the Treatment of Behavioral Disturbances Associated with Dementia,” The Journal of Prevention of Alzheimer's Disease, 2(4):302 (2015) (Clinical Trials in Alzheimer's Disease (CTAD) Congress, Symposium Summary OC51).
Dhawan et al., “Sleep-related Problems of Parkinson's Disease,” Age and Ageing, vol. 35, pp. 220-228, (2006); DOI: 10.1093/ageing/afj087.
Dyck, et al., “Effects of Deuterium Substitution on the Catabolismof β-Phenylethylamine: An In Vivo Study,” Journal of Neurochemistry, vol. 46, Issue 2, pp. 399-404, (1986).
Fawcett, J., “Posttraumatic Stress Disorder, Stress, and Happiness”, Psychiatric Annals Journal, vol. 28, No. 8, pp. 427-428, (1998).
Fletcher et al., “Perceiving is Believing: A Bayesian Approach to Explaining the Positive Symptoms of Schizophrenia,” Nature Reviews/Neuroscience, vol. 10, pp. 48-58, (2009).
Foster, A.B., “Deuterium Isotope Effects in the Metabolism of Drugs and Xenobiotics: Implications for Drug Design,” Advances in Drug Research, vol. 14, pp. 1-40, (1985).
Foster, et al., “Acetylcholinesterase Inhibitors Reduce Spreading Activation in Dementia,” Neuropsychologia, vol. 50, pp. 2093-2099, (2012).
Friedman, M.J.., “Current and Future Drug Treatment for Posttraumatic Stress Disorder Patients”, Psychiatric Annals Journal, vol. 28, No. 8, pp. 464-468, (1998).
Grant, “Polymorphism in Pharmaceutical Solids”, Chapter 1, pp. 1-10 (1999).
Guillory, “Polymorphism in Pharmaceutical Solids”, Chapter 5, pp. 183-226 (1999).
Hackam, et al., “Translation of Research Evidence from Animals to Humans,” JAMA, vol. 296, No. 14, pp. 1731-1732, (2006).
Harvey, et al., “Serotonin and Stress: Protective or Malevolent Actions in the Biobehavioral Response to Repeated Trauma?,” Annals of the New York Academy of Sciences, vol. 1032, pp. 267-272, (2004); DOI: 10.1196/annals.1314.035.
Haskins, N.J., “The Application of Stable Isotopes in Biomedical Research,” Biological Mass Spectrometry, vol. 9, No. 7, pp. 269-277, (1982).
Honma, S., et al., “The Metabolism of Roxatidine Acetate Hydrochloride: Liberation of Deuterium from the Piperidine Ring during Hydroxylation,” Drug Metabolism and Disposition, vol. 15, No. 4, pp. 551, (1987).
International Search Report issued in International Application No. PCT/US2019/049061, dated Nov. 13, 2019, 3 pages.
International Search Report issued in International Application No. PCT/US2019/049062, dated Nov. 15, 2019, 3 pages.
Izrayelit, L., “Schizoaffective Disorder and PTSD Successfully Treated With Olanzapine and Supportive Psychotherapy”, Psychiatric Annals Journal, vol. 28, No. 8, pp. 424-426, (1998).
Jordan, V.C. Nature Reviews: Drug Discovery, 2, 2003, 205.
Kahn et al., “Residual Symptoms of Schizophrenia. What are Realistic Treatment Goals? Lingering Symptoms Require you to Evaluate Pharmacotherapy and Offer Psychosocial Interventions,” Current Psychiatry, vol. 16, No. 3, pp. 35-40, (2017).
Kay, et al., “The Positive and Negative Syndrome Scale (PANSS) for Schizophrenia,” Schizophrenia Bulletin, vol. 13, No. 2, pp. 261-276, (1987).
Kessler, et al., “Lifetime Prevalence and Age-of-Onset Distributions of DSM-IV Disorders in the National Comorbidity Survey Replication,” Arch Gen Psychiatry, vol. 62, pp. 593-602, (2005).
Khorana, et al., “Gamma-Carbolines: Binding at 5-HT5A Serotonin Receptors,” Bioorganic & Medicinal Chemistry, vol. 11, pp. 717-722, p. 718 Table 1, (2003).
Koppel, et al., “Optimal Treatment of Alzheimer's Disease Psychosis: Challenges and Solutions,” Neuropsychiatric Disease and Treatment, vol. 10, pp. 2253-2262, (2014).
Krystal, J.H., et al., “Adjunctive Risperidone Treatment for Antidepressant-Resistant Symptoms of Chronic Military Service-Related PTSD: A Randomized Trial,” JAMA, 306(5):493-502, (2011).
Lammers, L. et al., “Risperidone long-acting injection in Schizophrenia Spectrum Illnesses compared to first generation depot antipsychotics in an outpatient setting in Canada,” BMC Psychiatry, 13:155; pp. 1-9 (2013).
Lebert, et al., “Trazodone in Fronto-Temporal Dementia,” Research and Practice in Alzheimer's Disease, vol. 11, pp. 356-360, (2006).
Lee, et al. “Novel, Highly Potent, Selective 5-HT2A/D2 Receptor Antagonists as Potential Atypical Antipsychotics,” Bioorg. Med. Chem. Lett., vol. 13, pp. 767-770, (2003).
Li et al., “Discovery of a Tetracyclic Quinoxaline Derivative as a Potent and Orally Active Multifunctional Drug Candidate for the Treatment of Neuropsychiatric and Neurological Disorders”, vol. 57, pp. 2670-2682 (2014).
Lieberman, et al., “ITI-007 for the Treatment of Schizophrenia: A 4-Week Randomized, Double-Blind, Controlled Trial,” Biol. Psychiatry, vol. 79, No. 12, pp. 952-961, (2015).
Lin, et al., “Dosage and Duration of Antipsychotic Treatment in Demented Outpatients with Agitation or Psychosis,” Journal of the Formosan Medical Association, vol. 114, pp. 147-153, (2015).
Lipschitz, et al., “Childhood Posttraumatic Stress Disorder: A Review of Neurobiologic Sequelae,” Psychiatric Annals Journal, vol. 28, No. 8, pp. 452-457, (1998).
Liriano et al., “Ketamine as treatment for post-traumatic stress disorder: a review.” Drugs in Context, vol. 8, 7 pages (2019).
Lopez, et al., “Psychiatric Symptoms Vary with the Severity of Dementia in Probably Alzheimer's Disease,” J. Neuropsychiatry Clin. Neurosc., vol. 15, No. 3, pp. 346-353, (2003).
Madhusoodanan, et al., “Pharmacological Management of Behavioral Symptoms Associated with Dementia,” World J. Psychiatr., vol. 4, No. 4, pp. 72-79, (2014).
Makadia et al., “Poly Lactic-co-Glycolic Acid (PLGA) as Biodegradable Controlled Drug Delivery Carrier,” Polymers (Basel), vol. 3, No. 3, pp. 1377-1397, (2011).
Marek et al. Synergistic Action of 5-HT2A Antagonists and Selective Serotonin Reuptake Inhibitors in Neuropsychiatric Disorders. Neuropsychopharmacology, 2003. Vol. 28, pp. 402-412. (Year: 2003).
Medisorb Fact Sheet, Medisorb Microspheres Technology (retrieved from the internet Nov. 13, 2018), 2 pages (2009).
Mohamed, et al., “Pharmacotherapy of PTSD in the U.S. Department of Veterans Affairs: Diagnostic- and Symptom-guided Drug Selection,” J. Clin. Psychiatry, vol. 69, pp. 959-965, (2008).
Morgan, et al., “Acoustic Startle in Individuals With Posttraumatic Stress Disorder,” Psychiatric Annals Journal, vol. 28, Issue 8, pp. 430-434, (1998).
Müller et al., “Detection of Depression in Acute Schizophrenia: Sensitivity and Specificity of 2 Standard Observer Rating Scales,” Can J Psychiatry, vol. 51, No. 6, pp. 387-392, (2006).
Newman et al., “Solid-state analysis of the active pharmaceutical ingredient in drug products”, Drug Discovery Today, vol. 8, No. 9, 898-903 (2003).
O'Gorman, et al., “Lumateperone (ITI-007): A Novel Investigational Agent with Broad Therapeutic Potential Across Multiple Neuropsychiatric Disorders,” Poster P.l.g.038, European College of Neuropsychopharmacology (ECNP) Congress (2017).
Palanisamy, M. et al., “Cellulose-Based Matrix Microspheres of Prednisolone Inclusion Complex; Preparation and Characterization.” American Association of Pharmaceutical Scientists PharmSciTech, vol. 12, No. 1, pp. 388-400, (2011).
Perlis et al., “Clinical Features of Bipolar Depression Versus Major Depressive Disorder in Large Multicenter Trials”, Am J Psychiatry, vol. 163, vol. 2, p. 225-231, (2006).
Pieniaszek, et al., “Moricizine Bioavailability via Simultaneous Dual, Stable Isotope Administration: Bioequivalence Implications,” J. Clin. Pharmacol., vol. 39, pp. 817-825, (1999).
Pine et al., “Dopamine, Time, and Impulsivity in Humans,” The Journal of Neuroscience, vol. 30, No. 26, pp. 8888-8896, (2010).
Press Release, “Intra-Cellular Therapies Announces Additional Results from Phase I/II Clinical Trial for ITI-007 in Healthy Geriatric Subjects and Patients With Dementia,” Intra-Cellular Therapies, Press Release Date: Nov. 21, 2014, (http://ir.intracellulartherapies.com/releasedetail.cfm?ReleaseID=884325), accessed on May 31, 2016.
Pubchem, OPEN Chemistry Database, PubChem SID 103920954, PubChem CID 90655118, (2011), 6 pages.
Rackova, et al., “Free Radical Scavenging and Antioxidant Activities of Substituted Hexahydropyridoindoles. Quantitative Structure-Activity Relationships.” Journal of Medicinal Chemistry, vol. 49, No. 8, pp. 2543-2548, (2006).
Rainer, M.K., “Risperidone Long-acting Injection: A Review of its Long Term Safety and Efficacy,” Neuropsychiatric Disease and Treatment, vol. 4, No. 5, pp. 919-927, (2008).
Ramaswamy et al., “Failed Efficacy of Ziprasidone in the Treatment of Post-Traumatic Stress Disorder,” Contemporary Clinical Trials Communications, vol. 2, pp. 1-5, (2016).
Reynolds et al., “Longitudinal Change in Memory Performance Associated with HTR2A Polymorphism,” Neurobiology of Aging, vol. 27, pp. 150-154, (2006).
Rye (Sleep Disorders and Parkinson's Disease, 2000, accessed online http://www.waparkinsons.org/edu_research/articles/Sleep_Disorders.html), 2 pages.
Satlin, et al., “ITI-007 (Lumateperone) for the Treatment of Agitation in Patients with Dementia, including Alzheimer's Disease,” Alzheimer's & Dementia 14(7) (Suppl.): P678-79 (2018) (Alzheimer's Assoc. International Conference 2018, summary of Poster P2-032).
Satlin, et al., “ITI-007 (Lumateperone) for the Treatment of Agitation in Patients with Dementia, including Alzheimer's Disease,” Poster P2-032, Alzheimer's Assoc. International Conference 2018 (2018).
Savjani et al., “Drug Solubility: Importance and Enhancement Techniques”, International Scholarly Research Network Pharmaceutics (2012), vol. 2012, pp. 1-10.
Schennach et al., “What Are Residual Symptoms in Schizophrenia Spectrum Disorder? Clinical Description and 1-year Persistence Within a Naturalistic Trial,” Eur. Arch. Psychiatry Clin. Neurosci., vol. 265, pp. 107-116, (2015); DOI: 10.1007/s00406-014-0528-2.
Seishinkei Shi, vol. 110, No. 7, pp. 557-584, (2008). Partial English translation only.
Semla et al., “Off-Label Prescribing of Second-Generation Antipsychotics to Elderly Veterans with Posttraumatic Stress Disorder and Dementia,” J. Am. Geriatr. Soc., vol. 65, No. 8, pp. 1789-1795, (2017); DOI: 10.1111/jgs.14897.
Singhal, et al., “Drug Polymorphism and Dosage Form Design: A Practical Perspective,” Advanced Drug Delivery Reviews, vol. 56, pp. 335-347, (2004).
Snyder et al., “Functional profile of a novel modulator of serotonin, dopamine, and glutamate neurotransmission”, Psychopharmacology, 232:605-621 (2015).
Southwick, et al., “Neuroendocrine Alterations in Posttraumatic Stress Disorder,” Psychiatric Annals Journal, vol. 28, No. 8, pp. 436-442, (1998).
Taragano, et al., “A Double-Blind, Randomized, Fixed-Dose Trial of Fluoxetine vs. Amitriptyline in the Treatment of Major Depression Complicating Alzheimer's Disease,” Psychosomatics, vol. 38, No. 3, pp. 246-252, (1997).
Timmins, G.S., “Deuterated drugs: where are we now?” Expert Opinion on Therapeutic Patents, 1-9 (2014).
Tung, R., “The Development of Deuterium-Containing Drugs,” Innovations in Pharmaceutical Technology, vol. 32, pp. 1-4, (2010).
Vanover, et al., “A Novel Approach to Address an Unmet Need in the Treatment of Schizophrenia and Depression: Lumateperone, an Innovative Modulator of Dopamine, Serotonin, and Glutamate,” Abstract presented at the American Society of Clinical Psychopharmacology (ASCP) Annual Meeting; May 29-Jun. 1, 2018; Miami, FL.
Vanover, et al., “Dopamine D2 receptor occupancy of lumateperone (ITI-007): a Positron Emission Tomography Study in patients with schizophrenia,” Neuropsychopharmacology 44:598-605, (2019).
Vanover, K., et al., “ITI-007: A Novel Therapy for the Treatment of Schizophrenia and Related Psychoses,” International Clinical Psychopharamcology, vol. 26, e56, 1 page, (2011).
Vloeberghs et al., “Altered Circadian Locomotor Activity in APP23 Mice: A Model for BPSD Disturbances,” European Journal of Neuroscience, vol. 20, pp. 2757-2766, (2004); DOI: 10.1111/j.1460-9568.2004.03755.x.
Vyas et al., “An Evaluation of Lumateperone Tosylate for the Treatment of Schizophrenia,” Expert Opinion on Pharmacotherapy, vol. 21, No. 2, pp. 139-145, (2020); https://doi.org/10.1080/14656566.2019.1695778.
Warner-Schmidt JL. et al. “Antidepressant effects of selective serotonin reuptake inhibitors (SSRis) are attenuated by anti-inflammatory drugs in mice and humans”. Proc.Natl. Acad.Sci., 108(22):9262-7 (2011).
Wennogle, et al., “Activation of NMDA and AMPA Receptors by Lumateperone (ITI-007): Implications for Antidepressant Activity,” Abstract presented at the 2017 Collegium Internationale Neuro-Psychopharmacologicum (CINP) Thematic Meeting: Treatment Resistant Depression; Jul. 20-22, 2017; Prague.
Weschules, et al., “Acetylcholinesterase Inhibitor and N-Methyl--Aspartic Acid Receptor Antagonist Use among Hospice Enrollees with a Primary Diagnosis of Dementia,” Journal of Palliative Medicine, vol. 11, No. 5, pp. 738-745, (2008).
Wiese, M., “DSC Detection of Polymorphism in Pharmaceutical Anhydrous Dexamethasone Acetate,” TA Instruments, TA 302, pp. 1-4, (2002).
Wolen, R. L., “The Application of Stable Isotopes to Studies of Drug Bioavailability and Bioequivalence,” J. Clin. Pharmacol., vol. 26, pp. 419-424, (1986).
Zhang et al., “The Role of Serotonin 5-HT2A Receptors in Memory and Cognition,” Front. Pharmacol., vol. 6, No. 225, pp. 1-17, (2015); DOI: 10.3389/fphar.2015.00225.
Related Publications (1)
Number Date Country
20210220280 A1 Jul 2021 US
Provisional Applications (2)
Number Date Country
62779920 Dec 2018 US
62725944 Aug 2018 US