US20200093808A1 - Methods for the Administration of Certain VMAT2 Inhibitors - Google Patents

Methods for the Administration of Certain VMAT2 Inhibitors Download PDF

Info

Publication number
US20200093808A1
US20200093808A1 US16/481,037 US201716481037A US2020093808A1 US 20200093808 A1 US20200093808 A1 US 20200093808A1 US 201716481037 A US201716481037 A US 201716481037A US 2020093808 A1 US2020093808 A1 US 2020093808A1
Authority
US
United States
Prior art keywords
inhibitor
administered
patient
composition
vmat2
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/481,037
Inventor
Christopher F. O`Brien
Haig P. Bozigian
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Neurocrine Biosciences Inc
Original Assignee
Neurocrine Biosciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=62978647&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20200093808(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Neurocrine Biosciences Inc filed Critical Neurocrine Biosciences Inc
Priority to US16/481,037 priority Critical patent/US20200093808A1/en
Assigned to NEUROCRINE BIOSCIENCES, INC. reassignment NEUROCRINE BIOSCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: O'Brien, Christopher F., BOZIGIAN, HAIG P.
Assigned to NEUROCRINE BIOSCIENCES, INC. reassignment NEUROCRINE BIOSCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: O'Brien, Christopher F., BOZIGIAN, HAIG P.
Publication of US20200093808A1 publication Critical patent/US20200093808A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/473Quinolines; Isoquinolines ortho- or peri-condensed with carbocyclic ring systems, e.g. acridines, phenanthridines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4375Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4525Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with oxygen as a ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D455/00Heterocyclic compounds containing quinolizine ring systems, e.g. emetine alkaloids, protoberberine; Alkylenedioxy derivatives of dibenzo [a, g] quinolizines, e.g. berberine
    • C07D455/03Heterocyclic compounds containing quinolizine ring systems, e.g. emetine alkaloids, protoberberine; Alkylenedioxy derivatives of dibenzo [a, g] quinolizines, e.g. berberine containing quinolizine ring systems directly condensed with at least one six-membered carbocyclic ring, e.g. protoberberine; Alkylenedioxy derivatives of dibenzo [a, g] quinolizines, e.g. berberine
    • C07D455/04Heterocyclic compounds containing quinolizine ring systems, e.g. emetine alkaloids, protoberberine; Alkylenedioxy derivatives of dibenzo [a, g] quinolizines, e.g. berberine containing quinolizine ring systems directly condensed with at least one six-membered carbocyclic ring, e.g. protoberberine; Alkylenedioxy derivatives of dibenzo [a, g] quinolizines, e.g. berberine containing a quinolizine ring system condensed with only one six-membered carbocyclic ring, e.g. julolidine
    • C07D455/06Heterocyclic compounds containing quinolizine ring systems, e.g. emetine alkaloids, protoberberine; Alkylenedioxy derivatives of dibenzo [a, g] quinolizines, e.g. berberine containing quinolizine ring systems directly condensed with at least one six-membered carbocyclic ring, e.g. protoberberine; Alkylenedioxy derivatives of dibenzo [a, g] quinolizines, e.g. berberine containing a quinolizine ring system condensed with only one six-membered carbocyclic ring, e.g. julolidine containing benzo [a] quinolizine ring systems

Definitions

  • Dysregulation of dopaminergic systems is integral to several central nervous system (CNS) disorders, including neurological and psychiatric diseases and disorders. These neurological and psychiatric diseases and disorders include hyperkinetic movement disorders, and conditions such as schizophrenia and mood disorders.
  • the transporter protein vesicular monoamine transporter-2 (VMAT2) plays an important role in presynaptic dopamine release and regulates monoamine uptake from the cytoplasm to the synaptic vesicle for storage and release.
  • valbenazine which has the following chemical structure:
  • valbenazine ditosylate A formulation of valbenazine:4-toluenesulfonate (1:2) (referred to herein as “valbenazine ditosylate”) has been previously reported in the FDA approved drug label Ingrezza®.
  • cytochrome P450 enzyme system (CYP450) is responsible for the biotransformation of drugs from active substances to inactive metabolites that can be excreted from the body.
  • CYP450 cytochrome P450 enzyme system
  • the metabolism of certain drugs by CYP450 can alter their PK profile and result in sub-therapeutic plasma levels of those drugs over time.
  • the cytochrome P450 gene superfamily is composed of at least 207 genes that have been named based on the evolutionary relationships of the cytochromes P450. For this nomenclature system, the sequences of all of the cytochrome P450 genes are compared, and those cytochromes P450 that share at least 40% identity are defined as a family (designated by CYP followed by a Roman or Arabic numeral, e.g., CYP3), and further divided into subfamilies (designated by a capital letter, e.g., CYP3A), which are comprised of those forms that are at least 55% related by their deduced amino acid sequences. Finally, the gene for each individual form of cytochrome P450 is assigned an Arabic number (e.g., CYP3A4).
  • CYP3A isoenzyme is a member of the cytochrome P450 superfamily which constitutes up to 60% of the total human liver microsomal cytochrome P450 and has been found in alimentary passage of stomach and intestines and livers. CYP3A has also been found in kidney epithelial cells, jejunal mucosa, and the lungs. CYP3A is one of the most abundant subfamilies in cytochrome P450 superfamily.
  • CYP3A At least five (5) forms of CYPs are found in human CYP3A subfamily, and these forms are responsible for the metabolism of a large number of structurally diverse drugs. In non-induced individuals, CYP3A may constitute 15% of the P450 enzymes in the liver; in enterocytes, members of the CYP3A subfamily constitute greater than 70% of the CYP-containing enzymes.
  • CYP3A is responsible for metabolism of a large number of drugs including nifedipine, macrolide antibiotics including erythromycin and troleandomycin, cyclosporin, FK506, teffenadine, tamoxifen, lidocaine, midazolam, triazolam, dapsone, diltiazem, lovastatin, quinidine, ethylestradiol, testosterone, and alfentanil.
  • CYP3A is involved in erythromycin N-demethylation, cyclosporine oxidation, nifedipine oxidation, midazolam hydroxylation, testosterone 6- ⁇ -hydroxylation, and cortisol 6- ⁇ -hydroxylation.
  • CYP3A has also been shown to be involved in both bioactivation and detoxication pathways for several carcinogens in vitro.
  • VMAT2 inhibitor such as valbenazine or (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof, wherein the patient is also being treated with another substance which may interact with the VMAT2 inhibitor, such as a CYP3A4 inhibitor.
  • a VMAT2 inhibitor such as valbenazine or (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 inhibitor chosen from valbenazine and (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinol
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 vesicular monoamine transport 2
  • CYP3A4 cytochrome P450 3A4
  • valbenazine may be referred to as (S)-2-amino-3-methyl-butyric acid (2R, 3R,11bR)-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl ester; or as L-Valine, (2R,3R,11bR)-1,3,4,6,7,11b-hexahydro-9,10-dimethoxy-3-(2-methylpropyl)-2H-benzo[a]quinolizin-2-yl ester or as NBI-98854.
  • (+)- ⁇ -HTBZ means the compound which is an active metabolite of valbenazine having the structure:
  • (+)- ⁇ -HTBZ may be referred to as (2R, 3R, 11bR) or as (+)- ⁇ -DHTBZ or as (+)- ⁇ -HTBZ or as R,R,R-DHTBZ or as (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol; or as (2R, 3R,11bR)-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol or as NBI-98782.
  • NBI-136110 means the compound which is a metabolite of valbenazine having the structure:
  • isotopic variant means a compound that contains an unnatural proportion of an isotope at one or more of the atoms that constitute such a compound.
  • an “isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, hydrogen ( 1 H), deuterium ( 2 H), tritium ( 3 H), carbon-11 ( 11 C), carbon-12 ( 12 C), carbon-13 ( 13 C), carbon-14 ( 14 C), nitrogen-13 ( 13 N), nitrogen-14 ( 14 N), nitrogen-15 ( 15 N), oxygen-14 ( 14 O), oxygen-15 ( 15 0), oxygen-16 ( 16 O), oxygen-17 ( 17 O), oxygen-18 ( 18 O), fluorine-17 ( 17 F), fluorine-18 ( 18 F), phosphorus-31 ( 31 P), phosphorus-32 ( 32 P), phosphorus-33 ( 33 P), sulfur-32 ( 32 S), sulfur-33 ( 33 S), sulfur-34 ( 34 S), sulfur-35 ( 35 S), sulfur-36 ( 36 S), chlorine-35 (
  • an “isotopic variant” of a compound is in a stable form, that is, non-radioactive.
  • an “isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, hydrogen ( 1 H), deuterium ( 2 H), carbon-12 ( 12 C), carbon-13 ( 13 C), nitrogen-14 ( 14 N), nitrogen-15 ( 15 N), oxygen-16 ( 16 O), oxygen-17 ( 17 O), and oxygen-18 ( 18 O).
  • an “isotopic variant” of a compound is in an unstable form, that is, radioactive.
  • an “isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, tritium ( 3 H), carbon-11 ( 11 C), carbon-14 ( 14 C), nitrogen-13 ( 13 N), oxygen-14 ( 14 O), and oxygen-15 ( 15 O).
  • any hydrogen can be 2 H, as example, or any carbon can be 13 C, as example, or any nitrogen can be 15 N, as example, and any oxygen can be 18 O, as example, where feasible according to the judgment of one of skill in the art.
  • an “isotopic variant” of a compound contains an unnatural proportion of deuterium.
  • a position designated as having deuterium typically has a minimum isotopic enrichment factor of, in certain embodiments, at least 1000 (15% deuterium incorporation), at least 2000 (30% deuterium incorporation), at least 3000 (45% deuterium incorporation), at least 3500 (52.5% deuterium incorporation), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation)
  • a substance is a “substrate” of enzyme activity when it can be chemically transformed by action of the enzyme on the substance.
  • Substrates can be either activated or deactivated by the enzyme.
  • Enzyme activity refers broadly to the specific activity of the enzyme (i.e., the rate at which the enzyme transforms a substrate per mg or mole of enzyme) as well as the metabolic effect of such transformations.
  • a substance is an “inhibitor” of enzyme activity when the specific activity or the metabolic effect of the specific activity of the enzyme can be decreased by the presence of the substance, without reference to the precise mechanism of such decrease.
  • a substance can be an inhibitor of enzyme activity by competitive, non-competitive, allosteric or other type of enzyme inhibition, by decreasing expression of the enzyme, or other direct or indirect mechanisms. Co-administration of a given drug with an inhibitor may decrease the rate of metabolism of that drug through the metabolic pathway listed.
  • a substance is an “inducer” of enzyme activity when the specific activity or the metabolic effect of the specific activity of the enzyme can be increased by the presence of the substance, without reference to the precise mechanism of such increase.
  • a substance can be an inducer of enzyme activity by increasing reaction rate, by increasing expression of the enzyme, by allosteric activation or other direct or indirect mechanisms. Co-administration of a given drug with an enzyme inducer may increase the rate of excretion of the drug metabolized through the pathway indicated.
  • a substance can be a substrate, inhibitor, or inducer of an enzyme activity.
  • the substance can be an inhibitor of enzyme activity by one mechanism and an inducer of enzyme activity by another mechanism.
  • the function (substrate, inhibitor, or inducer) of the substance with respect to activity of an enzyme can depend on environmental conditions.
  • a “strong CYP3A4 inhibitor” is a compound that increases the area under the concentration time curve (AUC) of a sensitive index substrate of the CYP3A4 pathway by ⁇ 5-fold.
  • Index substrates predictably exhibit exposure increase due to inhibition or induction of a given metabolic pathway and are commonly used in prospective clinical drug-drug interaction studies.
  • Sensitive index substrates are index substrates that demonstrate an increase in AUC of ⁇ 5-fold with strong index inhibitors of a given metabolic pathway in clinical drug-drug interaction studies studies. Examples of sensitive index substrates for the CYP2D6 pathway are midazolam and triazolam.
  • hypokinetic disorder or “hyperkinetic movement disorder” or “hyperkinesias” refers to disorders or diseases characterized by excessive, abnormal, involuntary movements. These neurological disorders include tremor, dystonia, myoclonus, athetosis, Huntington's disease, tardive dyskinesia, Tourette syndrome, dystonia, hemiballismus, chorea, senile chorea, or tics.
  • Tardive dyskinesia is characterized by rapid, repetitive, stereotypic, involuntary movements of the face, limbs, or trunk.
  • AUC refers to the area under the curve, or the integral, of the plasma concentration of an active pharmaceutical ingredient or metabolite over time following a dosing event.
  • AUC 0-4 is the integral under the plasma concentration curve from time 0 (dosing) to time “t”.
  • AUC 0- ⁇ is the AUC from time 0 (dosing) to time infinity. Unless otherwise stated, AUC refers to AUC 0- ⁇ .
  • a drug is packaged in a salt form, for example valbenazine ditosylate, and the dosage form strength refers to the mass of this salt form or the equivalent mass of the corresponding free base, valbenazine.
  • C max is a pharmacokinetic parameter denoting the maximum observed blood plasma concentration following delivery of an active pharmaceutical ingredient. C max occurs at the time of maximum plasma concentration, t max .
  • co-administer and “co-administration” and variants thereof mean the administration of at least two drugs to a patient either subsequently, simultaneously, or consequently proximate in time to one another (e.g., within the same day, or week or period of 30 days, or sufficiently proximate that each of the at least two drugs can be simultaneously detected in the blood plasma).
  • two or more active agents can be co-formulated as part of the same composition or administered as separate formulations. This also may be referred to herein as “concomitant” administration or variants thereof.
  • adjusting administration As used herein, “adjusting administration”, “altering administration”, “adjusting dosing”, or “altering dosing ” are all equivalent and mean tapering off, reducing or increasing the dose of the substance, ceasing to administer the substance to the patient, or substituting a different active agent for the substance.
  • administering to a patient refers to the process of introducing a composition or dosage form into the patient via an art-recognized means of introduction.
  • disorder is intended to be generally synonymous, and is used interchangeably with, the terms “disease,” “syndrome,” and “condition” (as in medical condition), in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms.
  • a “dose” means the measured quantity of an active agent to be taken at one time by a patient.
  • the quantity is the molar equivalent to the corresponding amount of valbenazine free base.
  • a drug is packaged in a pharmaceutically acceptable salt form, for example valbenazine ditosylate, and the dosage for strength refers to the mass of the molar equivalent of the corresponding free base, valbenazine.
  • 73 mg of valbenazine tosylate is the molar equivalent of 40 mg of valbenazine free base.
  • dose regimen means the dose of an active agent taken at a first time by a patient and the interval (time or symptomatic) at which any subsequent doses of the active agent are taken by the patient such as from about 20 to about 160 mg once daily, e.g., about 20, about 40, about 60, about 80, about 100, about 120, or about 160 mg once daily.
  • the additional doses of the active agent can be different from the dose taken at the first time.
  • an agent, compound, drug, composition or combination is an amount which is nontoxic and effective for producing some desired therapeutic effect upon administration to a subject or patient (e.g., a human subject or patient).
  • the precise therapeutically effective amount for a subject may depend upon, e.g., the subject's size and health, the nature and extent of the condition, the therapeutics or combination of therapeutics selected for administration, and other variables known to those of skill in the art. The effective amount for a given situation is determined by routine experimentation and is within the judgment of the clinician.
  • informing means referring to or providing published material, for example, providing an active agent with published material to a user; or presenting information orally, for example, by presentation at a seminar, conference, or other educational presentation, by conversation between a pharmaceutical sales representative and a medical care worker, or by conversation between a medical care worker and a patient; or demonstrating the intended information to a user for the purpose of comprehension.
  • labeling means all labels or other means of written, printed, graphic, electronic, verbal, or demonstrative communication that is upon a pharmaceutical product or a dosage form or accompanying such pharmaceutical product or dosage form.
  • a “medical care worker” means a worker in the health care field who may need or utilize information regarding an active agent, including a dosage form thereof, including information on safety, efficacy, dosing, administration, or pharmacokinetics.
  • Examples of medical care workers include physicians, pharmacists, physician's assistants, nurses, aides, caretakers (which can include family members or guardians), emergency medical workers, and veterinarians.
  • “Medication Guide” means an FDA-approved patient labeling for a pharmaceutical product conforming to the specifications set forth in 21 CFR 208 and other applicable regulations which contains information for patients on how to safely use a pharmaceutical product.
  • a medication guide is scientifically accurate and is based on, and does not conflict with, the approved professional labeling for the pharmaceutical product under 21 CFR 201.57, but the language need not be identical to the sections of approved labeling to which it corresponds.
  • a medication guide is typically available for a pharmaceutical product with special risk management information.
  • patient or “individual” or “ subject” means a mammal, including a human, for whom or which therapy is desired, and generally refers to the recipient of the therapy.
  • patient package insert means information for patients on how to safely use a pharmaceutical product that is part of the FDA-approved labeling. It is an extension of the professional labeling for a pharmaceutical product that may be distributed to a patient when the product is dispensed which provides consumer-oriented information about the product in lay language, for example it may describe benefits, risks, how to recognize risks, dosage, or administration.
  • “pharmaceutically acceptable” refers to a material that is not biologically or otherwise undesirable, i.e., the material may be incorporated into a pharmaceutical composition administered to a patient without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the composition in which it is contained.
  • pharmaceutically acceptable refers to a pharmaceutical carrier or excipient, it is implied that the carrier or excipient has met the required standards of toxicological and manufacturing testing or that it is included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug administration.
  • “Pharmacologically active” as in a “pharmacologically active” (or “active”) derivative or analog, refers to a derivative or analog having the same type of pharmacological activity as the parent compound and approximately equivalent in degree.
  • pharmaceutically acceptable salts include acid addition salts which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like.
  • Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • a “product” or “pharmaceutical product” means a dosage form of an active agent plus published material, and optionally packaging.
  • product insert means the professional labeling (prescribing information) for a pharmaceutical product, a patient package insert for the pharmaceutical product, or a medication guide for the pharmaceutical product.
  • “professional labeling” or “prescribing information” means the official description of a pharmaceutical product approved by a regulatory agency (e.g., FDA or EMEA) regulating marketing of the pharmaceutical product, which includes a summary of the essential scientific information needed for the safe and effective use of the drug, such as, for example indication and usage; dosage and administration; who should take it; adverse events (side effects); instructions for use in special populations (pregnant women, children, geriatric, etc.); safety information for the patient, and the like.
  • FDA regulatory agency
  • published material means a medium providing information, including printed, audio, visual, or electronic medium, for example a flyer, an advertisement, a product insert, printed labeling, an internet web site, an internet web page, an internet pop-up window, a radio or television broadcast, a compact disk, a DVD, an audio recording, or other recording or electronic medium.
  • risk means the probability or chance of adverse reaction, injury, or other undesirable outcome arising from a medical treatment.
  • An “acceptable risk” means a measure of the risk of harm, injury, or disease arising from a medical treatment that will be tolerated by an individual or group. Whether a risk is “acceptable” will depend upon the advantages that the individual or group perceives to be obtainable in return for taking the risk, whether they accept whatever scientific and other advice is offered about the magnitude of the risk, and numerous other factors, both political and social.
  • An “acceptable risk” of an adverse reaction means that an individual or a group in society is willing to take or be subjected to the risk that the adverse reaction might occur since the adverse reaction is one whose probability of occurrence is small, or whose consequences are so slight, or the benefits (perceived or real) of the active agent are so great.
  • An “unacceptable risk” of an adverse reaction means that an individual or a group in society is unwilling to take or be subjected to the risk that the adverse reaction might occur upon weighing the probability of occurrence of the adverse reaction, the consequences of the adverse reaction, and the benefits (perceived or real) of the active agent.
  • “At risk” means in a state or condition marked by a high level of risk or susceptibility. Risk assessment consists of identifying and characterizing the nature, frequency, and severity of the risks associated with the use of a product.
  • safety means the incidence or severity of adverse events associated with administration of an active agent, including adverse effects associated with patient-related factors (e.g., age, gender, ethnicity, race, target illness, abnormalities of renal or hepatic function, co-morbid illnesses, genetic characteristics such as metabolic status, or environment) and active agent-related factors (e.g., dose, plasma level, duration of exposure, or concomitant medication).
  • patient-related factors e.g., age, gender, ethnicity, race, target illness, abnormalities of renal or hepatic function, co-morbid illnesses, genetic characteristics such as metabolic status, or environment
  • active agent-related factors e.g., dose, plasma level, duration of exposure, or concomitant medication
  • t max is a pharmacokinetic parameter denoting the time to maximum blood plasma concentration following delivery of an active pharmaceutical ingredient
  • t 1/2 or “plasma half-life” or “elimination half-life” or the like is a pharmacokinetic parameter denoting the apparent plasma terminal phase half-life, i.e., the time, after absorption and distribution of a drug is complete, for the plasma concentration to fall by half
  • treating refers to therapeutic applications to slow or stop progression of a disorder, prophylactic application to prevent development of a disorder, and/or reversal of a disorder.
  • Reversal of a disorder differs from a therapeutic application which slows or stops a disorder in that with a method of reversing, not only is progression of a disorder completely stopped, cellular behavior is moved to some degree, toward a normal state that would be observed in the absence of the disorder.
  • VMAT2 refers to human vesicular monoamine transporter isoform 2, an integral membrane protein that acts to transport monoamines, particularly neurotransmitters such as dopamine, norepinephrine, serotonin, and histamine, from cellular cytosol into synaptic vesicles.
  • VMAT2 inhibitor refers to the ability of a compound disclosed herein to alter the function of VMAT2.
  • a VMAT2 inhibitor may block or reduce the activity of VMAT2 by forming a reversible or irreversible covalent bond between the inhibitor and VMAT2 or through formation of a noncovalently bound complex. Such inhibition may be manifest only in particular cell types or may be contingent on a particular biological event.
  • VMAT2 inhibitor also refers to altering the function of VMAT2 by decreasing the probability that a complex forms between a VMAT2 and a natural substrate.
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 inhibitor chosen from valbenazine and (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinol
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 vesicular monoamine transport 2
  • the method further comprises determining whether the patient is being administered a strong CYP3A4 inhibitor.
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 vesicular monoamine transport 2
  • CYP3A4 cytochrome P450 3A4
  • the method further comprises informing the patient or a medical care worker that administration of the VMAT2 inhibitor to patients who is also being administered a strong CYP3A4 inhibitor in higher exposure of valbenazine and/or (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol than administration of the VMAT2 inhibitor to a patient who is not being administered a strong CYP3A4 inhibitor.
  • the method further comprises informing the patient or a medical care worker that administration of the VMAT2 inhibitor to a patient who is also being administered a strong CYP3A4 inhibitor may result in increased risk of one or more exposure-related adverse reactions than administration of the VMAT2 inhibitor to a patient who is not being administered a strong CYP3A4 inhibitor.
  • the one or more exposure-related adverse reactions is chosen from somnolence, anticholinergic effects, balance disorders or falls, headache, akathisia, vomiting, nausea, arthralgia, QT prolongation, increase in blood glucose, increase in weight, respiratory infections, drooling, dyskinesia, extrapyramidal symptoms (non-akathisia), anxiety, insomnia, increase in prolactin, increase in alkaline phosphatase, and increase in bilirubin.
  • the one or more exposure-related adverse reactions is chosen from somnolence, anticholinergic effects, balance disorders or falls, headache, akathisia, vomiting, nausea, arthralgia, and QT prolongation.
  • the one or more exposure-related adverse reactions is chosen from somnolence and QT prolongation.
  • the method further comprises informing the patient or a medical care worker that co-administration of the VMAT2 inhibitor and the CYP3A4 inhibitor may prolong the patient's QT interval.
  • the method further comprises informing the patient or a medical care worker that administration of the VMAT2 inhibitor to a patient who is also being administered a strong CYP3A4 inhibitor may prolong the patient's QT interval than administration of the VMAT2 inhibitor to a patient who is also being administered a strong CYP3A4 inhibitor.
  • the strong CYP3A4 inhibitor is chosen from clarithromycin, chloramphenicol, cobicistat, indinavir, itraconazole, ketoconazole, nefazodone, nelfinavir, ritonavir, saquinavir, and telithromycin. In certain embodiments, the strong CYP3A4 inhibitor is chosen from clarithromycin, itraconazole, and ketoconazole. In certain embodiments, the strong CYP3A4 inhibitor is ketoconazole.
  • the VMAT2 inhibitor is administered to the patient to treat a neurological or psychiatric disease or disorder.
  • the neurological or psychiatric disease or disorder is a hyperkinetic movement disorder, mood disorder, bipolar disorder, schizophrenia, schizoaffective disorder, mania in mood disorder, depression in mood disorder, treatment-refractory obsessive compulsive disorder, neurological dysfunction associated with Lesch-Nyhan syndrome, agitation associated with Alzheimer's disease, Fragile X syndrome or Fragile X-associated tremor-ataxia syndrome, autism spectrum disorder, Rett syndrome, or chorea-acanthocytosis.
  • the neurological or psychiatric disease or disorder is a hyperkinetic movement disorder.
  • the hyperkinetic movement disorder is tardive dyskinesia.
  • the hyperkinetic movement disorder is Tourette's syndrome.
  • the hyperkinetic movement disorder is Huntington's disease.
  • the hyperkinetic movement disorder is tics.
  • the hyperkinetic movement disorder is chorea associated with Huntington's disease.
  • the hyperkinetic movement disorder is ataxia, chorea, dystonia, Huntington's disease, myoclonus, restless leg syndrome, or tremors.
  • the VMAT2 inhibitor is administered orally.
  • the VMAT2 inhibitor is administered in the form of a tablet or capsule.
  • the VMAT2 inhibitor is administered with or without food.
  • the VMAT2 inhibitor is valbenazine or a pharmaceutically acceptable salt and/or isotopic variant thereof. In certain embodiments, the VMAT2 inhibitor is valbenazine or a pharmaceutically acceptable salt thereof. In certain embodiments, the VMAT2 inhibitor is a valbenazine tosylate salt. In certain embodiments, the VMAT2 inhibitor is a ditosylate salt of valbenazine.
  • the VMAT2 inhibitor is an isotopic variant that is L-Valine, (2R, 3R,11bR)-1,3,4,6,7,11b-hexahydro-9,10-di(methoxy-d 3 )-3-(2-methylpropyl)-2H-benzo[ ⁇ ]quinolizin-2-yl ester or a pharmaceutically acceptable salt thereof.
  • the VMAT2 inhibitor is administered in an amount equivalent to between about 20 mg and about 160 mg of valbenazine free base. In certain embodiments, the VMAT2 inhibitor is administered in an amount equivalent to about 20 mg of valbenazine free base. In certain embodiments, the VMAT2 inhibitor is administered in an amount equivalent to about 40 mg of valbenazine free base. In certain embodiments, the VMAT2 inhibitor is administered in an amount equivalent to about 60 mg of valbenazine free base. In certain embodiments, the VMAT2 inhibitor is administered in an amount equivalent to about 80 mg of valbenazine free base. In certain embodiments, the VMAT2 inhibitor is administered in an amount equivalent to about 120 mg of valbenazine free base. In certain embodiments, the VMAT2 inhibitor is administered in an amount equivalent to about 160 mg of valbenazine free base.
  • the VMAT2 inhibitor is administered for a first period of time in a first amount and then the amount is increased to a second amount.
  • the first period of time is a week.
  • the first amount is equivalent to about 40 mg of valbenazine free base.
  • the second amount is equivalent to about 80 mg of valbenazine free base.
  • the VMAT2 inhibitor is (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof.
  • the VMAT2 inhibitor is (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt thereof.
  • the VMAT2 inhibitor is an isotopic variant that is (+)- ⁇ -3-isobutyl-9,10-di(methoxy-d 3 )-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol or a pharmaceutically acceptable salt thereof.
  • the VMAT2 inhibitor is administered in an amount sufficient to achieve a maximal blood plasma concentration (C max ) of (+)- ⁇ -DHTBZ of between about 15 ng to about 60 ng per mL plasma and a minimal blood plasma concentration (C max ) of (+)- ⁇ -DHTBZ of at least 15 ng per mL plasma over an 8 hour period.
  • the VMAT2 inhibitor is administered in an amount sufficient to achieve a maximal blood plasma concentration (C max ) of (+)- ⁇ -DHTBZ of between about 15 ng to about 60 ng per mL plasma and a minimal blood plasma concentration (C min ) of approximately between about at least 33% -50% of the C max over a 12 hour period.
  • C max maximal blood plasma concentration
  • C min minimal blood plasma concentration
  • the VMAT2 inhibitor is administered in an amount sufficient to achieve: (i) a therapeutic concentration range of about 15 ng to about 60 ng of (+)- ⁇ -DHTBZ per mL plasma; and (ii) a threshold concentration of at least 15 ng (+)- ⁇ -DHTBZ per mL plasma over a period of about 8 hours to about 24 hours.
  • a method for treating neurological or psychiatric diseases or disorders comprises administering to a subject a pharmaceutical composition comprising the VMAT2 inhibitor, in an amount sufficient to achieve a maximal blood plasma concentration (C max ) of R,R,R-DHTBZ of between about 15 ng to about 60 ng per mL plasma and a minimal blood plasma concentration (C min ) of R,R,R-DHTBZ of at least 15 ng per mL plasma over an 8 hour period.
  • C max maximal blood plasma concentration
  • C min minimal blood plasma concentration
  • the C max of R,R,R-DHTBZ is about 15 ng/mL, about 20 ng/mL, about 25 ng/mL, about 30 ng/mL, about 35 ng/mL, about 40 ng/mL, about 45 ng/mL, about 50 ng/mL, about 55 ng/mL or about 60 ng/mL plasma.
  • the C min of R,R,R-DHTBZ is at least 15 ng/mL, at least 20 ng/mL, at least 25 ng/mL, at least 30 ng/mL, or at least 35 ng/mL plasma, over a period of 8 hrs, 12 hrs, 16 hrs, 20 hrs, 24 hrs, 28 hrs, or 32 hrs. In certain embodiments, the C min of R,R,R-DHTBZ is between about 15 ng/mL to about 35 ng/mL.
  • the pharmaceutical composition is administered in an amount sufficient to provide a C max of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a C min of approximately at least 33% of the C max over a 24 hour period. In certain embodiments, the pharmaceutical composition is administered in an amount sufficient to provide a C max of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a C min of approximately at least 50% of the C max over a 24 hour period.
  • the pharmaceutical composition is administered in an amount sufficient to provide a C max of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a C min of approximately between about at least 33% -50% of the C max over a 24 hour period.
  • the pharmaceutical composition is administered in an amount sufficient to provide a C max of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a C min of approximately at least 33% of the C max over a 12 hour period. In certain embodiments, the pharmaceutical composition is administered in an amount sufficient to provide a C max of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a C min of approximately at least 50% of the C max over a 12 hour period.
  • the pharmaceutical composition is administered in an amount sufficient to provide a C max of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a C min of approximately between about at least 33% -50% of the C max over a 12 hour period.
  • the pharmaceutical composition is administered to a subject in an amount that provides a C max of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a C min of between about 5 ng/mL to about 30 ng/mL plasma over a 24 hour period. In certain embodiments, the pharmaceutical composition is administered to a subject in an amount that provides a C max of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a C min of between about 7.5 ng/mL to about 30 ng/mL plasma over a 24 hour period.
  • a method for treating neurological or psychiatric diseases or disorders comprises administering to a subject a pharmaceutical composition comprising the VMAT2 inhibitor, as an active pharmaceutical ingredient, in an amount sufficient to provide: (i) a therapeutic concentration range of about 15 ng to about 60 ng of R,R,R-DHTBZ per mL plasma; and (ii) a threshold concentration of at least 15 ng R,R,R-DHTBZ per mL plasma over a period of about 8 hours to about 24 hours.
  • the therapeutic concentration range is about 15 ng to about 35 ng, to about 40 ng, to about 45 ng, to about 50 ng, or to about 55 ng R,R,R-DHTBZ per mL plasma.
  • the threshold concentration of R,R,R-DHTBZ is about 15 ng/mL, about 20 ng/mL, about 25 ng/mL, about 30 ng/mL, about 35 ng/mL, about 40 ng/mL, about 45 ng/mL, about 50 ng/mL, about 55 ng/mL or about 60 ng/mL plasma, over a period of about 8 hrs, about 12 hrs, about 16 hrs, about 20 hrs, about 24 hrs, about 28 hrs, or about 32 hrs. In certain embodiments, the threshold concentration of R,R,R-DHTBZ is between about 15 ng/mL to about 35 ng/mL over a period of about 8 hours to about 24 hours.
  • Plasma concentrations may be measured by methods known in the art and generally by tandem mass spectroscopy.
  • the therapeutically effective amount of the VMAT2 inhibitor is 10-90% less than the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor.
  • the therapeutically effective amount of the VMAT2 inhibitor is 20-80% less than the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor.
  • the therapeutically effective amount of the VMAT2 inhibitor is 30-70% less than the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor.
  • the therapeutically effective amount of the VMAT2 inhibitor is 40-60% less than the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor.
  • the therapeutically effective amount of the VMAT2 inhibitor is about 50% less than the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor.
  • the dosage administered to a patient who is not also being administered a strong CYP3A4 inhibitor is 40 mg per day
  • an individual may receive a reduced dosage of 4-36 mg per day, e.g., 8-32 mg per day, such as 12-28 mg per day, for example, 16-24 mg per day, or in certain embodiments, about 20 mg per day.
  • the dosage administered to a patient who is not also being administered a strong CYP3A4 inhibitor is 80 mg per day
  • an individual may receive a reduced dosage of 8-72 mg per day, e.g., 16-64 mg per day, such as 24-56 mg per day, for example, 32-48 mg per day, or in certain embodiments, about 24 mg per day.
  • the dose of VMAT2 inhibitor administered to a patient is reduced to, for example, 75% or less, 50% or less, or 25% or less of the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor.
  • the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor is 40 mg per day
  • an individual may receive a reduced dosage of 30, 20, or 10 mg per day.
  • the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor is 80 mg per day, an individual may receive a reduced dosage of 60, 40, or 20 mg per day.
  • VMAT2 vesicular monoamine transport 2
  • a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof wherein the patient has previously been determined to have been administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering the VMAT2 inhibitor in an amount equivalent to about 40 mg of valbenazine free base once daily to the patient.
  • CYP3A4 cytochrome P450 3A4
  • VMAT2 vesicular monoamine transport 2
  • a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof, said method comprising: determining whether the patient has been administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor; selecting the patient for treatment where the patient has been administered a strong CYP3A4 inhibitor; and administering the VMAT2 inhibitor to the selected patient in an amount equivalent to about 40 mg of valbenazine free base once daily.
  • CYP3A4 cytochrome P450 3A4
  • VMAT2 vesicular monoamine transport 2
  • a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof, comprising: administering to the patient a therapeutically effective amount of the VMAT2 inhibitor, subsequently determining that the patient is to begin treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor, selecting the patient for treatment where the patient is to begin treatment with a strong CYP3A4 inhibitor, and administering the VMAT2 inhibitor to the selected patient in an amount equivalent to about 40 mg of valbenazine free base once daily.
  • VMAT2 vesicular monoamine transport 2
  • CYP3A4 cytochrome P450 3A4
  • VMAT2 vesicular monoamine transport 2
  • a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof, said method comprising: determining whether the patient has been administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor; selecting the patient for treatment where the patient has been administered a strong CYP3A4 inhibitor; and administering the VMAT2 inhibitor to the selected patient in an amount that is less than the amount that would be administered to a patient who is not being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • VMAT2 vesicular monoamine transport 2
  • CYP3A4 cytochrome P450 3A4
  • VMAT2 vesicular monoamine transport 2
  • a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof, wherein the patient has previously been determined to have been administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering to the patient a therapeutically effective amount of the VMAT2 inhibitor, subsequently selecting the patient that is not able to tolerate one or more exposure-related adverse reactions, and administering a reduced amount of VMAT2 inhibitor, such as 40 mg once daily, to the patient.
  • CYP3A4 cytochrome P450 3
  • VMAT2 vesicular monoamine transport 2
  • a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof, wherein the patient has previously been determined to have been administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering to the patient a therapeutically effective amount of the VMAT2 inhibitor, subsequently selecting the patient that is able to tolerate one or more exposure-related adverse reactions, and continuing administering the therapeutically effective amount of the VMAT2 inhibitor to the patient.
  • CYP3A4 cytochrome P450 3A4
  • VMAT2 vesicular monoamine transport 2
  • a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof wherein the patient has previously been determined to have been administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering the VMAT2 inhibitor in an amount sufficient to produce a mean valbenazine C max that is about 1 to about 2 fold higher than the mean valbenazine C max for a patient who is not being administered a strong CYP3A4 inhibitor and/or a mean valbenazine A
  • VMAT2 vesicular monoamine transport 2
  • a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof wherein the patient has previously been determined to have been administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering the VMAT2 inhibitor in an amount sufficient to produce a mean (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol C max that is about 1 to
  • Valbenazine can be prepared according to U.S. Pat. Nos. 8,039,627 and 8,357,697, the disclosure of each of which is incorporated herein by reference in its entirety. Tetrabenazine may be administered by a variety of methods including the formulations disclosed in PCT Publications WO 2010/018408, WO 2011/019956, and WO 2014/047167, the disclosure of each of which is incorporated herein by reference in its entirety. In certain embodiments, the valbenazine for use in the compositions and methods provided herein is in polymorphic Form I as disclosed in U.S. Ser. No. 15/338,214, the disclosure of which is incorporated herein by reference in its entirety.
  • VMAT2 vesicular monoamine transport 2
  • VMAT2 vesicular monoamine transport 2
  • compositions for treating a patient in need of a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, comprising the VMAT2 inhibitor, characterized in that the composition comprising the VMAT2 inhibitor in an amount equivalent to about 40 mg of valbenazine free base is administered once daily to the patient subsequently determined to begin treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor following administration of the composition comprising a therapeutically effective amount of the VMAT2 inhibitor.
  • VMAT2 vesicular monoamine transport 2
  • compositions for treating a patient in need of a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, comprising the VMAT2 inhibitor, characterized in that the composition comprising the VMAT2 inhibitor in an amount that would be less than that administered to a patient who has not begun treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor is administered to the patient subsequently determined to have begun treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor following administration of the composition comprising a therapeutically effective amount of the VMAT2 inhibitor.
  • VMAT2 vesicular monoamine transport 2
  • the patient or a medical care worker is informed that administration of the VMAT2 inhibitor to patients who is also being administered a strong CYP3A4 inhibitor in higher exposure of valbenazine and/or (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol than administration of the composition to a patient who is not being administered a strong CYP3A4 inhibitor.
  • the patient or a medical care worker is informed that administration of the VMAT2 inhibitor to a patient who is also being administered a strong CYP3A4 inhibitor may result in increased risk of one or more exposure-related adverse reactions than administration of the composition to a patient who is not being administered a strong CYP3A4 inhibitor.
  • the one or more exposure-related adverse reactions is chosen from somnolence, anticholinergic effects, balance disorders or falls, headache, akathisia, vomiting, nausea, arthralgia, QT prolongation, increase in blood glucose, increase in weight, respiratory infections, drooling, dyskinesia, extrapyramidal symptoms (non-akathisia), anxiety, insomnia, increase in prolactin, increase in alkaline phosphatase, and increase in bilirubin.
  • the one or more exposure-related adverse reactions is chosen from somnolence, anticholinergic effects, balance disorders or falls, headache, akathisia, vomiting, nausea, arthralgia, and QT prolongation.
  • the one or more exposure-related adverse reactions is chosen from somnolence and QT prolongation.
  • the patient or a medical care worker is informed that co-administration of the composition and the CYP3A4 inhibitor may prolong the patient's QT interval.
  • the patient or a medical care worker is informed that administration of the composition to a patient who is also being administered a strong CYP3A4 inhibitor may prolong the patient's QT interval than administration of the composition to a patient who is also being administered a strong CYP3A4 inhibitor.
  • the strong CYP3A4 inhibitor is chosen from clarithromycin, chloramphenicol, cobicistat, indinavir, itraconazole, ketoconazole, nefazodone, nelfinavir, ritonavir, saquinavir, and telithromycin. In certain embodiments, the strong CYP3A4 inhibitor is chosen from clarithromycin, itraconazole, and ketoconazole. In certain embodiments, the strong CYP3A4 inhibitor is ketoconazole.
  • the composition is for treating a neurological or psychiatric disease or disorder.
  • the composition is administered orally.
  • the composition is administered in the form of a tablet or capsule.
  • the composition is administered with or without food.
  • the VMAT2 inhibitor is valbenazine or a pharmaceutically acceptable salt and/or isotopic variant thereof.
  • the VMAT2 inhibitor is valbenazine or a pharmaceutically acceptable salt thereof.
  • the VMAT2 inhibitor is a valbenazine tosylate salt.
  • the VMAT2 inhibitor is a ditosylate salt of valbenazine.
  • the VMAT2 inhibitor is an isotopic variant that is L-Valine, (2R, 3R,11 bR)-1,3,4,6,7,11b-hexahydro-9,10-di(methoxy-d 3 )-3-(2-methylpropyl)-2H-benzo[ ⁇ ]quinolizin-2-yl ester or a pharmaceutically acceptable salt thereof.
  • the VMAT2 inhibitor is (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof.
  • the VMAT2 inhibitor is (+)- ⁇ -3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt thereof.
  • the VMAT2 inhibitor is an isotopic variant that is (+)- ⁇ -3-isobutyl-9,10-di(methoxy-d 3 )-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol or a pharmaceutically acceptable salt thereof.
  • the composition is administered in an amount equivalent to between about 20 mg and about 160 mg of valbenazine free base. In certain embodiments, the composition is administered in an amount equivalent to about 20 mg of valbenazine free base. In certain embodiments, the composition is administered in an amount equivalent to about 40 mg of valbenazine free base. In certain embodiments, the composition is administered in an amount equivalent to about 60 mg of valbenazine free base. In certain embodiments, the composition is administered in an amount equivalent to about 80 mg of valbenazine free base. In certain embodiments, the composition is administered in an amount equivalent to about 120 mg of valbenazine free base.
  • the composition is administered for a first period of time in a first amount and then the amount is increased to a second amount.
  • the first period of time is a week.
  • the first amount is equivalent to about 40 mg of valbenazine free base.
  • the second amount is equivalent to about 80 mg of valbenazine free base.
  • the composition is administered in an amount sufficient to achieve a maximal blood plasma concentration (C max ) of (+)- ⁇ -DHTBZ of between about 15 ng to about 60 ng per mL plasma and a minimal blood plasma concentration (C min ) of (+)- ⁇ -DHTBZ of at least 15 ng per mL plasma over an 8 hour period.
  • C max maximal blood plasma concentration
  • C min minimal blood plasma concentration
  • the composition is administered in an amount sufficient to achieve a maximal blood plasma concentration (C max ) of (+)- ⁇ -DHTBZ of between about 15 ng to about 60 ng per mL plasma and a minimal blood plasma concentration (C min ) of approximately between about at least 33% -50% of the C max over a 12 hour period.
  • C max maximal blood plasma concentration
  • C min minimal blood plasma concentration
  • the composition is administered in an amount sufficient to achieve: (i) a therapeutic concentration range of about 15 ng to about 60 ng of (+)- ⁇ -DHTBZ per mL plasma; and (ii) a threshold concentration of at least 15 ng (+)- ⁇ -DHTBZ per mL plasma over a period of about 8 hours to about 24 hours.
  • the therapeutically effective amount of the VMAT2 inhibitor is 10-90% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • the therapeutically effective amount of the VMAT2 inhibitor is 20-80% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • the therapeutically effective amount of the VMAT2 inhibitor is 30-70% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • the therapeutically effective amount of the VMAT2 inhibitor is 40-60% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • the therapeutically effective amount of the VMAT2 inhibitor is about 50% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • compositions for use in treating neurological or psychiatric diseases or disorders comprising the VMAT2 inhibitor as an active pharmaceutical ingredient, in combination with one or more pharmaceutically acceptable carriers or excipients.
  • excipient to a large extent, depends on factors, such as the particular mode of administration, the effect of the excipient on the solubility and stability of the active ingredient, and the nature of the dosage form.
  • the pharmaceutical compositions provided herein may be provided in unit dosage forms or multiple-dosage forms.
  • Unit-dosage forms refer to physically discrete units suitable for administration to human and animal subjects and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of the active ingredient(s) sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carriers or excipients. Examples of unit-dosage forms include ampoules, syringes, and individually packaged tablets and capsules. Unit dosage forms may be administered in fractions or multiples thereof.
  • a multiple-dosage form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dosage form. Examples of multiple-dosage forms include vials, bottles of tablets or capsules, or bottles of pints or gallons.
  • the pharmaceutical compositions provided herein may be administered alone, or in combination with one or more other compounds provided herein, one or more other active ingredients.
  • the pharmaceutical compositions provided herein may be formulated in various dosage forms for oral, parenteral, and topical administration.
  • the pharmaceutical compositions may also be formulated as a modified release dosage form, including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms. These dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art).
  • the pharmaceutical compositions provided herein may be administered at once, or multiple times at intervals of time.
  • dosage and duration of treatment may vary with the age, weight, and condition of the patient being treated, and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test or diagnostic data. It is further understood that for any particular individual, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the formulations.
  • oral administration also includes buccal, lingual, and sublingual administration.
  • Suitable oral dosage forms include, but are not limited to, tablets, capsules, pills, troches, lozenges, pastilles, cachets, pellets, medicated chewing gum, granules, bulk powders, effervescent or non-effervescent powders or granules, solutions, emulsions, suspensions, solutions, wafers, sprinkles, elixirs, and syrups.
  • the pharmaceutical compositions may contain one or more pharmaceutically acceptable carriers or excipients, including, but not limited to, binders, fillers, diluents, disintegrants, wetting agents, lubricants, glidants, coloring agents, dye-migration inhibitors, sweetening agents, and flavoring agents.
  • pharmaceutically acceptable carriers or excipients including, but not limited to, binders, fillers, diluents, disintegrants, wetting agents, lubricants, glidants, coloring agents, dye-migration inhibitors, sweetening agents, and flavoring agents.
  • Binders or granulators impart cohesiveness to a tablet to ensure the tablet remaining intact after compression.
  • Suitable binders or granulators include, but are not limited to, starches, such as corn starch, potato starch, and pre-gelatinized starch (e.g., STARCH 1500); gelatin; sugars, such as sucrose, glucose, dextrose, molasses, and lactose; natural and synthetic gums, such as acacia, alginic acid, alginates, extract of Irish moss, Panwar gum, ghatti gum, mucilage of isabgol husks, carboxymethylcellulose, methylcellulose, polyvinylpyrrolidone (PVP), Veegum, larch arabogalactan, powdered tragacanth, and guar gum; celluloses, such as ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose, methyl cellulose, hydroxyeth
  • Suitable fillers include, but are not limited to, talc, calcium carbonate, microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pregelatinized starch, and mixtures thereof.
  • the binder or filler may be present from about 50 to about 99% by weight in the pharmaceutical compositions provided herein.
  • Suitable diluents include, but are not limited to, dicalcium phosphate, calcium sulfate, lactose, sorbitol, sucrose, inositol, cellulose, kaolin, mannitol, sodium chloride, dry starch, and powdered sugar.
  • Certain diluents, such as mannitol, lactose, sorbitol, sucrose, and inositol when present in sufficient quantity, can impart properties to some compressed tablets that permit disintegration in the mouth by chewing. Such compressed tablets can be used as chewable tablets.
  • Suitable disintegrants include, but are not limited to, agar; bentonite; celluloses, such as methylcellulose and carboxymethylcellulose; wood products; natural sponge; cation-exchange resins; alginic acid; gums, such as guar gum and Vee gum HV; citrus pulp; cross-linked celluloses, such as croscarmellose; cross-linked polymers, such as crospovidone; cross-linked starches; calcium carbonate; microcrystalline cellulose, such as sodium starch glycolate; polacrilin potassium; starches, such as com starch, potato starch, tapioca starch, and pre-gelatinized starch; clays; aligns; and mixtures thereof.
  • the amount of disintegrant in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art.
  • the pharmaceutical compositions provided herein may contain from about 0.5 to about 15% or from about 1 to about 5% by weight of a disintegrant.
  • Suitable lubricants include, but are not limited to, calcium stearate; magnesium stearate; mineral oil; light mineral oil; glycerin; sorbitol; mannitol; glycols, such as glycerol behenate and polyethylene glycol (PEG); stearic acid; sodium lauryl sulfate; talc; hydrogenated vegetable oil, including peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, com oil, and soybean oil; zinc stearate; ethyl oleate; ethyl laureate; agar; starch; lycopodium; silica or silica gels, such as AEROSIL®200 (W.R.
  • compositions provided herein may contain about 0.1 to about 5% by weight of a lubricant.
  • Suitable glidants include colloidal silicon dioxide, CAB-0-SIL® (Cabot Co. of Boston, Mass.), and asbestos-free talc.
  • Coloring agents include any of the approved, certified, water soluble FD&C dyes, and water insoluble FD&C dyes suspended on alumina hydrate, and color lakes and mixtures thereof.
  • a color lake is the combination by adsorption of a water-soluble dye to a hydrous oxide of a heavy metal, resulting in an insoluble form of the dye.
  • Flavoring agents include natural flavors extracted from plants, such as fruits, and synthetic blends of compounds which produce a pleasant taste sensation, such as peppermint and methyl salicylate.
  • Sweetening agents include sucrose, lactose, mannitol, syrups, glycerin, and artificial sweeteners, such as saccharin and aspartame.
  • Suitable emulsifying agents include gelatin, acacia, tragacanth, bentonite, and surfactants, such as polyoxyethylene sorbitan monooleate (TWEEN® 20), polyoxyethylene sorbitan monooleate 80 (TWEEN® 80), and triethanolamine oleate.
  • Suspending and dispersing agents include sodium carboxymethylcellulose, pectin, tragacanth, Veegum, acacia, sodium carbomethylcellulose, hydroxypropyl methylcellulose, and polyvinylpyrolidone.
  • Preservatives include glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol.
  • Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate, and polyoxyethylene lauryl ether.
  • Solvents include glycerin, sorbitol, ethyl alcohol, and syrup. Examples of non-aqueous liquids utilized in emulsions include mineral oil and cottonseed oil.
  • Organic acids include citric and tartaric acid.
  • Sources of carbon dioxide include sodium bicarbonate and sodium carbonate.
  • compositions provided herein may be provided as compressed tablets, tablet triturates, chewable lozenges, rapidly dissolving tablets, multiple compressed tablets, or enteric-coating tablets, sugar-coated, or film-coated tablets.
  • Enteric coated tablets are compressed tablets coated with substances that resist the action of stomach acid but dissolve or disintegrate in the intestine, thus protecting the active ingredients from the acidic environment of the stomach.
  • Enteric-coatings include, but are not limited to, fatty acids, fats, phenylsalicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalates.
  • Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which may be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation.
  • Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material.
  • Film coatings include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coating imparts the same general characteristics as sugar coating.
  • Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets.
  • the tablet dosage forms may be prepared from the active ingredient in powdered, crystalline, or granular forms, alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, controlled-release polymers, lubricants, diluents, and/or colorants. Flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges.
  • the pharmaceutical compositions provided herein may be provided as soft or hard capsules, which can be made from gelatin, methylcellulose, starch, or calcium alginate.
  • the hard gelatin capsule also known as the dry-filled capsule (DFC)
  • DFC dry-filled capsule
  • the soft elastic capsule (SEC) is a soft, globular shell, such as a gelatin shell, which is plasticized by the addition of glycerin, sorbitol, or a similar polyol.
  • the soft gelatin shells may contain a preservative to prevent the growth of microorganisms. Suitable preservatives are those as described herein, including methyl- and propyl-parabens, and sorbic acid.
  • liquid, semisolid, and solid dosage forms may be encapsulated in a capsule.
  • suitable liquid and semisolid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils, or triglycerides.
  • the capsules may also be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient.
  • compositions provided herein may be provided in liquid and semisolid dosage forms, including emulsions, solutions, suspensions, elixirs, and syrups.
  • An emulsion is a two-phase system, in which one liquid is dispersed in the form of small globules throughout another liquid, which can be oil-in-water or water-in-oil.
  • Emulsions may include a pharmaceutically acceptable non-aqueous liquids or solvent, emulsifying agent, and preservative.
  • Suspensions may include a pharmaceutically acceptable suspending agent and preservative.
  • Aqueous alcoholic solutions may include a pharmaceutically acceptable acetal, such as a di(lower alkyl) acetal of a lower alkyl aldehyde (the term “lower” means an alkyl having between 1 and 6 carbon atoms), e.g., acetaldehyde diethyl acetal; and a water-miscible solvent having one or more hydroxyl groups, such as propylene glycol and ethanol.
  • Elixirs are clear, sweetened, and hydroalcoholic solutions.
  • Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may also contain a preservative.
  • a solution in a polyethylene glycol may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be measured conveniently for administration.
  • liquid and semisolid dosage forms include, but are not limited to, those containing the active ingredient(s) provided herein, and a dialkylated mono- or polyalkylene glycol, including, 1,2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether, wherein 350, 550, and 750 refer to the approximate average molecular weight of the polyethylene glycol.
  • a dialkylated mono- or polyalkylene glycol including, 1,2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether, wherein 350, 550, and 750 refer to the approximate average molecular weight of the polyethylene glycol.
  • formulations may further comprise one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.
  • antioxidants such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.
  • antioxidants such as but
  • compositions provided herein for oral administration may be also provided in the forms of liposomes, micelles, microspheres, or nanosystems.
  • compositions provided herein may be provided as noneffervescent or effervescent, granules and powders, to be reconstituted into a liquid dosage form.
  • Pharmaceutically acceptable carriers and excipients used in the non-effervescent granules or powders may include diluents, sweeteners, and wetting agents.
  • Pharmaceutically acceptable carriers and excipients used in the effervescent granules or powders may include organic acids and a source of carbon dioxide. Coloring and flavoring agents can be used in all of the above dosage forms.
  • the pharmaceutical compositions provided herein may be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms.
  • compositions provided herein may be co-formulated with other active ingredients which do not impair the desired therapeutic action, or with substances that supplement the desired action, such as antacids, proton pump inhibitors, and Hz-receptor antagonists.
  • compositions provided herein may be administered parenterally by injection, infusion, or implantation, for local or systemic administration.
  • Parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial, and subcutaneous administration.
  • compositions provided herein may be formulated in any dosage forms that are suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems, and solid forms suitable for solutions or suspensions in liquid prior to injection.
  • dosage forms can be prepared according to conventional methods known to those skilled in the art of pharmaceutical science.
  • compositions intended for parenteral administration may include one or more pharmaceutically acceptable carriers and excipients, including, but not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, cryoprotectants, lyoprotectants, thickening agents, pH adjusting agents, and inert gases.
  • aqueous vehicles water-miscible vehicles
  • non-aqueous vehicles non-aqueous vehicles
  • antimicrobial agents or preservatives against the growth of microorganisms stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emuls
  • Suitable aqueous vehicles include, but are not limited to, water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringers injection, isotonic dextrose injection, sterile water injection, dextrose and lactated Ringers injection.
  • Non-aqueous vehicles include, but are not limited to, fixed oils of vegetable origin, castor oil, com oil, cottonseed oil, olive oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oils, hydrogenated soybean oil, and medium-chain triglycerides of coconut oil, and palm seed oil.
  • Water-miscible vehicles include, but are not limited to, ethanol, 1,3-butanediol, liquid polyethylene glycol (e.g., polyethylene glycol 300 and polyethylene glycol 400), propylene glycol, glycerin, N-methyl-2-pyrrolidone, dimethylacetamide, and dimethylsulfoxide.
  • Suitable antimicrobial agents or preservatives include, but are not limited to, phenols, cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl phydroxybenzates, thimerosal, benzalkonium chloride, benzethonium chloride, methyl- and propylparabens, and sorbic acid.
  • Suitable isotonic agents include, but are not limited to, sodium chloride, glycerin, and dextrose.
  • Suitable buffering agents include, but are not limited to, phosphate and citrate.
  • Suitable antioxidants are those as described herein, including bisulfite and sodium metabisulfite.
  • Suitable local anesthetics include, but are not limited to, procaine hydrochloride.
  • Suitable suspending and dispersing agents are those as described herein, including sodium carboxymethylcelluose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone.
  • Suitable emulsifying agents include those described herein, including polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate 80, and triethanolamine oleate.
  • Suitable sequestering or chelating agents include, but are not limited to EDTA.
  • Suitable pH adjusting agents include, but are not limited to, sodium hydroxide, hydrochloric acid, citric acid, and lactic acid.
  • Suitable complexing agents include, but are not limited to, cyclodextrins, including alpha-cyclodextrin, beta-cyclodextrin, hydroxypropyl-beta-cyclodextrin, sulfobutylether-beta-cyclodextrin, and sulfobutylether 7-beta-cyclodextrin (CAPTISOL®, CyDex, Lenexa, Kans.).
  • cyclodextrins including alpha-cyclodextrin, beta-cyclodextrin, hydroxypropyl-beta-cyclodextrin, sulfobutylether-beta-cyclodextrin, and sulfobutylether 7-beta-cyclodextrin (CAPTISOL®, CyDex, Lenexa, Kans.).
  • compositions provided herein may be formulated for single or multiple dosage administration.
  • the single dosage formulations are packaged in an ampule, a vial, or a syringe.
  • the multiple dosage parenteral formulations must contain an antimicrobial agent at bacteriostatic or fungistatic concentrations. All parenteral formulations must be sterile, as known and practiced in the art.
  • the pharmaceutical compositions are provided as ready-to-use sterile solutions.
  • the pharmaceutical compositions are provided as sterile dry soluble products, including lyophilized powders and hypodermic tablets, to be reconstituted with a vehicle prior to use.
  • the pharmaceutical compositions are provided as ready-to-use sterile suspensions.
  • the pharmaceutical compositions are provided as sterile dry insoluble products to be reconstituted with a vehicle prior to use.
  • the pharmaceutical compositions are provided as ready-to-use sterile emulsions.
  • compositions provided herein may be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms.
  • the pharmaceutical compositions may be formulated as a suspension, solid, semi-solid, or thixotropic liquid, for administration as an implanted depot.
  • the pharmaceutical compositions provided herein are dispersed in a solid inner matrix, which is surrounded by an outer polymeric membrane that is insoluble in body fluids but allows the active ingredient in the pharmaceutical compositions diffuse through.
  • Suitable inner matrixes include polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinylacetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers, such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol, and cross-linked partially hydrolyzed polyvinyl acetate.
  • Suitable outer polymeric membranes include polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinylacetate copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinylchloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer.
  • compositions provided herein may be administered topically to the skin, orifices, or mucosa.
  • topical administration include (intra)dermal, conjuctival, intracorneal, intraocular, ophthalmic, auricular, transdermal, nasal, vaginal, uretheral, respiratory, and rectal administration.
  • compositions provided herein may be formulated in any dosage forms that are suitable for topical administration for local or systemic effect, including emulsions, solutions, suspensions, creams, gels, hydrogels, ointments, dusting powders, dressings, elixirs, lotions, suspensions, tinctures, pastes, foams, films, aerosols, irrigations, sprays, suppositories, bandages, dermal patches.
  • the topical formulation of the pharmaceutical compositions provided herein may also comprise liposomes, micelles, microspheres, nanosystems, and mixtures thereof.
  • Pharmaceutically acceptable carriers and excipients suitable for use in the topical formulations provided herein include, but are not limited to, aqueous vehicles, water miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, penetration enhancers, cryopretectants, lyoprotectants, thickening agents, and inert gases.
  • compositions may also be administered topically by electroporation, iontophoresis, phonophoresis, sonophoresis and microneedle or needle-free injection, such as POWDERJECTTM (Chiron Corp., Emeryville, Calif.), and BIOJECTTM (Bioject Medical Technologies Inc., Tualatin, Oreg.).
  • electroporation iontophoresis, phonophoresis, sonophoresis and microneedle or needle-free injection
  • BIOJECTTM Bioject Medical Technologies Inc., Tualatin, Oreg.
  • Suitable ointment vehicles include oleaginous or hydrocarbon bases, including such as lard, benzoinated lard, olive oil, cottonseed oil, and other oils, white petrolatum; emulsifiable or absorption bases, such as hydrophilic petrolatum, hydroxystearin sulfate, and anhydrous lanolin; water-removable bases, such as hydrophilic ointment; water-soluble ointment bases, including polyethylene glycols of varying molecular weight; emulsion bases, either water-in-oil (W/O) emulsions or oil-in-water (O/W) emulsions, including cetyl alcohol, glyceryl monostearate, lanolin, and stearic acid. These vehicles are emollient but generally require addition of antioxidants and preservatives.
  • Suitable cream base can be oil-in-water or water-in-oil.
  • Cream vehicles may be water-washable, and contain an oil phase, an emulsifier, and an aqueous phase.
  • the oil phase is also called the “internal” phase, which is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol.
  • the aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant.
  • the emulsifier in a cream formulation may be a nonionic, anionic, cationic, or amphoteric surfactant.
  • Gels are semisolid, suspension-type systems. Single-phase gels contain organic macromolecules distributed substantially uniformly throughout the liquid carrier. Suitable gelling agents include crosslinked acrylic acid polymers, such as carbomers, carboxypolyalkylenes, Carbopol®; hydrophilic polymers, such as polyethylene oxides, polyoxyethylene-polyoxypropylene copolymers, and polyvinylalcohol; cellulosic polymers, such as hydroxypropyl cellulose, hydroxyethyl cellulose, hydroxypropyl methylcellulose, hydroxypropyl methylcellulose phthalate, and methylcellulose; gums, such as tragacanth and xanthan gum; sodium alginate; and gelatin.
  • dispersing agents such as alcohol or glycerin can be added, or the gelling agent can be dispersed by trituration, mechanical mixing, and/or stirring.
  • compositions provided herein may be administered rectally, urethrally, vaginally, or perivaginally in the forms of suppositories, pessaries, bougies, poultices or cataplasm, pastes, powders, dressings, creams, plasters, contraceptives, ointments, solutions, emulsions, suspensions, tampons, gels, foams, sprays, or enemas.
  • These dosage forms can be manufactured using conventional processes.
  • Rectal, urethral, and vaginal suppositories are solid bodies for insertion into body orifices, which are solid at ordinary temperatures but melt or soften at body temperature to release the active ingredient(s) inside the orifices.
  • Pharmaceutically acceptable carriers utilized in rectal and vaginal suppositories include vehicles, such as stiffening agents, which produce a melting point in the proximity of body temperature, when formulated with the pharmaceutical compositions provided herein; and antioxidants as described herein, including bisulfite and sodium metabisulfite.
  • Suitable vehicles include, but are not limited to, cocoa butter (theobroma oil), glycerin-gelatin, carbowax (polyoxyethylene glycol), spermaceti, paraffin, white and yellow wax, and appropriate mixtures of mono-, di- and triglycerides of fatty acids, hydrogels, such as polyvinyl alcohol, hydroxyethyl methacrylate, polyacrylic acid; glycerinated gelatin. Combinations of the various vehicles may be used. Rectal and vaginal suppositories may be prepared by the compressed method or molding. The typical weight of a rectal and vaginal suppository is about 2 to 3 g.
  • compositions provided herein may be administered ophthalmically in the forms of solutions, suspensions, ointments, emulsions, gel-forming solutions, powders for solutions, gels, ocular inserts, and implants.
  • the pharmaceutical compositions provided herein may be administered intranasally or by inhalation to the respiratory tract.
  • the pharmaceutical compositions may be provided in the form of an aerosol or solution for delivery using a pressurized container, pump, spray, atomizer, such as an atomizer using electrohydrodynamics to produce a fine mist, or nebulizer, alone or in combination with a suitable propellant, such as 1,1,1,2-tetrafluoroethane or 1,1,1,2,3,3,3-heptafluoropropane.
  • atomizer such as an atomizer using electrohydrodynamics to produce a fine mist, or nebulizer, alone or in combination with a suitable propellant, such as 1,1,1,2-tetrafluoroethane or 1,1,1,2,3,3,3-heptafluoropropane.
  • a suitable propellant such as 1,1,1,2-tetrafluoroethane or 1,1,1,2,3,3,3-heptafluoropropane.
  • Solutions or suspensions for use in a pressurized container, pump, spray, atomizer, or nebulizer may be formulated to contain ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilizing, or extending release of the active ingredient provided herein, a propellant as solvent; and/or a surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
  • compositions provided herein may be micronized to a size suitable for delivery by inhalation, such as 50 micrometers or less, or 10 micrometers or less.
  • Particles of such sizes may be prepared using a comminuting method known to those skilled in the art, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenization, or spray drying.
  • Capsules, blisters and cartridges for use in an inhaler or insufflator may be formulated to contain a powder mix of the pharmaceutical compositions provided herein; a suitable powder base, such as lactose or starch; and a performance modifier, such as /-leucine, mannitol, or magnesium stearate.
  • the lactose may be anhydrous or in the form of the monohydrate.
  • Other suitable excipients include dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose, and trehalose.
  • the pharmaceutical compositions provided herein for inhaled/intranasal administration may further comprise a suitable flavor, such as menthol and levomenthol, or sweeteners, such as saccharin or saccharin sodium.
  • compositions provided herein for topical administration may be formulated to be immediate release or modified release, including delayed-, sustained-, pulsed-, controlled-, targeted, and programmed release.
  • modified release dosage forms may be formulated as a modified release dosage form.
  • modified release refers to a dosage form in which the rate or place of release of the active ingredient(s) is different from that of an immediate dosage form when administered by the same route.
  • Modified release dosage forms include delayed-, extended-, prolonged-, sustained-, pulsatile- or pulsed-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms.
  • compositions in modified release dosage forms can be prepared using a variety of modified release devices and methods known to those skilled in the art, including, but not limited to, matrix controlled release devices, osmotic controlled release devices, multiparticulate controlled release devices, ion-exchange resins, enteric coatings, multilayered coatings, microspheres, liposomes, and combinations thereof.
  • the release rate of the active ingredient(s) can also be modified by varying the particle sizes and polymorphorism of the active ingredient(s).
  • compositions provided herein in a modified release dosage form may be fabricated using a matrix controlled release device known to those skilled in the art.
  • the pharmaceutical compositions provided herein in a modified release dosage form is formulated using an erodible matrix device, which is water swellable, erodible, or soluble polymers, including synthetic polymers, and naturally occurring polymers and derivatives, such as polysaccharides and proteins.
  • an erodible matrix device which is water swellable, erodible, or soluble polymers, including synthetic polymers, and naturally occurring polymers and derivatives, such as polysaccharides and proteins.
  • Materials useful in forming an erodible matrix include, but are not limited to, chitin, chitosan, dextran, and pullulan; gum agar, gum arabic, gum karaya, locust bean gum, gum tragacanth, carrageenans, gum ghatti, guar gum, xanthan gum, and scleroglucan; starches, such as dextrin and maltodextrin; hydrophilic colloids, such as pectin; phosphatides, such as lecithin; alginates; propylene glycol alginate; gelatin; collagen; and cellulosics, such as ethyl cellulose (EC), methylethyl cellulose (MEC), carboxymethyl cellulose (CMC), CMEC, hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), cellulose acetate (CA), cellulose propionate (CP), cellulose butyrate (CB), cellulose a
  • the pharmaceutical compositions are formulated with a non-erodible matrix device.
  • the active ingredient(s) is dissolved or dispersed in an inert matrix and is released primarily by diffusion through the inert matrix once administered.
  • Materials suitable for use as a non-erodible matrix device included, but are not limited to, insoluble plastics, such as polyethylene, polypropylene, polyisoprene, polyisobutylene, polybutadiene, polymethylmethacrylate, polybutylmethacrylate, chlorinated polyethylene, polyvinylchloride, methyl acrylate-methyl methacrylate copolymers, ethylene-vinylacetate copolymers, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, vinylchloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene
  • the desired release kinetics can be controlled, for example, via the polymer type employed, the polymer viscosity, the particle sizes of the polymer and/or the active ingredient(s), the ratio of the active ingredient(s) versus the polymer, and other excipients in the compositions.
  • compositions provided herein in a modified release dosage form may be prepared by methods known to those skilled in the art, including direct compression, dry or wet granulation followed by compression, melt-granulation followed by compression.
  • compositions provided herein in a modified release dosage form may be fabricated using an osmotic controlled release device, including one-chamber system, two-chamber system, asymmetric membrane technology (AMT), and extruding core system (ECS).
  • an osmotic controlled release device including one-chamber system, two-chamber system, asymmetric membrane technology (AMT), and extruding core system (ECS).
  • such devices have at least two components: (a) the core which contains the active ingredient(s); and (b) a semipermeable membrane with at least one delivery port, which encapsulates the core.
  • the semipermeable membrane controls the influx of water to the core from an aqueous environment of use so as to cause drug release by extrusion through the delivery port(s).
  • the core of the osmotic device optionally includes an osmotic agent, which creates a driving force for transport of water from the environment of use into the core of the device.
  • osmotic agents waterswellable hydrophilic polymers, which are also referred to as “osmopolymers” and “hydrogels,” including, but not limited to, hydrophilic vinyl and acrylic polymers, polysaccharides such as calcium alginate, polyethylene oxide (PEO), polyethylene glycol (PEG), polypropylene glycol (PPG), poly(2-hydroxyethyl methacrylate), poly(acrylic) acid, poly(methacrylic) acid, polyvinylpyrrolidone (PVP), crosslinked PVP, polyvinyl alcohol (PVA), PVA/PVP copolymers, PVA/PVP copolymers with hydrophobic monomers such as methyl methacrylate and vinyl acetate, hydrophilic polyurethanes containing large PEO
  • osmogens which are capable of imbibing water to affect an osmotic pressure gradient across the barrier of the surrounding coating.
  • Suitable osmogens include, but are not limited to, inorganic salts, such as magnesium sulfate, magnesium chloride, calcium chloride, sodium chloride, lithium chloride, potassium sulfate, potassium phosphates, sodium carbonate, sodium sulfite, lithium sulfate, potassium chloride, and sodium sulfate; sugars, such as dextrose, fructose, glucose, inositol, lactose, maltose, mannitol, raffinose, sorbitol, sucrose, trehalose, and xylitol,; organic acids, such as ascorbic acid, benzoic acid, fumaric acid, citric acid, maleic acid, sebacic acid, sorbic acid, adipic acid, edetic acid, glut
  • Osmotic agents of different dissolution rates may be employed to influence how rapidly the active ingredient(s) is initially delivered from the dosage form.
  • amorphous sugars such as Mannogeme EZ (SPI Pharma, Lewes, Del.) can be used to provide faster delivery during the first couple of hours to promptly produce the desired therapeutic effect, and gradually and continually release of the remaining amount to maintain the desired level of therapeutic or prophylactic effect over an extended period of time.
  • the active ingredient(s) is released at such a rate to replace the amount of the active ingredient metabolized and excreted.
  • the core may also include a wide variety of other excipients and carriers as described herein to enhance the performance of the dosage form or to promote stability or processing.
  • Materials useful in forming the semipermeable membrane include various grades of acrylics, vinyls, ethers, polyamides, polyesters, and cellulosic derivatives that are water-permeable and water-insoluble at physiologically relevant pHs, or are susceptible to being rendered water-insoluble by chemical alteration, such as crosslinking.
  • Suitable polymers useful in forming the coating include plasticized, unplasticized, and reinforced cellulose acetate (CA), cellulose diacetate, cellulose triacetate, CA propionate, cellulose nitrate, cellulose acetate butyrate (CAB), CA ethyl carbamate, CAP, CA methyl carbamate, CA succinate, cellulose acetate trimellitate (CAT), CA dimethylaminoacetate, CAethyl carbonate, CA chloroacetate, CA ethyl oxalate, CA methyl sulfonate, CA butyl sulfonate, CA p-toluene sulfonate, agar acetate, amylose triacetate, beta glucan acetate, beta glucan triacetate, acetaldehyde dimethyl acetate, triacetate of locust bean gum, hydroxlated ethylene-vinylacetate, EC, PEG, PPG, PEG/PPG copoly
  • the delivery port(s) on the semipermeable membrane may be formed postcoating by mechanical or laser drilling. Delivery port(s) may also be formed in situ by erosion of a plug of water-soluble material or by rupture of a thinner portion of the membrane over an indentation in the core. In addition, delivery ports may be formed during coating process.
  • the total amount of the active ingredient(s) released and the release rate can substantially by modulated via the thickness and porosity of the semipermeable membrane, the composition of the core, and the number, size, and position of the delivery ports.
  • compositions in an osmotic controlled-release dosage form may further comprise additional conventional excipients as described herein to promote performance or processing of the formulation.
  • the osmotic controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art.
  • the pharmaceutical compositions provided herein are formulated as AMT controlled-release dosage form, which comprises an asymmetric osmotic membrane that coats a core comprising the active ingredient(s) and other pharmaceutically acceptable excipients.
  • AMT controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art, including direct compression, dry granulation, wet granulation, and a dip-coating method.
  • the pharmaceutical compositions provided herein are formulated as ESC controlled-release dosage form, which comprises an osmotic membrane that coats a core comprising the active ingredient(s), hydroxylethyl cellulose, and other pharmaceutically acceptable excipients.
  • compositions provided herein in a modified release dosage form may be fabricated a multiparticulate controlled release device, which comprises a multiplicity of particles, granules, or pellets, ranging from about 10 pm to about 3 mm, about 50 pm to about 2.5 mm, or from about 100 pm to 1 mm in diameter.
  • multiparticulates may be made by the processes know to those skilled in the art, including wet-and dry-granulation, extrusion/spheronization, roller-compaction, melt-congealing, and by spray-coating seed cores.
  • excipients as described herein may be blended with the pharmaceutical compositions to aid in processing and forming the multiparticulates.
  • the resulting particles may themselves constitute the multiparticulate device or may be coated by various film forming materials, such as enteric polymers, water-swellable, and water-soluble polymers.
  • the multiparticulates can be further processed as a capsule or a tablet.
  • compositions provided herein may also be formulated to be targeted to a particular tissue, receptor, or other area of the body of the subject to be treated, including liposome-, resealed erythrocyte-, and antibody-based delivery systems.
  • an appropriate dosage level generally is about 0.001 to 100 mg per kg patient body weight per day (mg/kg per day), about 0.01 to about 80 mg/kg per day, about 0.1 to about 50 mg/kg per day, about 0.5 to about 25 mg/kg per day, or about 1 to about 20 mg/kg per day, which may be administered in single or multiple doses. Within this range the dosage may be 0.005 to 0.05, 0.05 to 0.5, or 0.5 to 5.0, 1 to 15, 1 to 20, or 1 to 50 mg/kg per day. In certain embodiments, the dosage level is about 0.001 to 100 mg/kg per day.
  • the dosage level is about from 25 to 100 mg/kg per day. In certain embodiments, the dosage level is about 0.01 to about 40 mg/kg per day. In certain embodiments, the dosage level is about 0.1 to about 80 mg/kg per day. In certain embodiments, the dosage level is about 0.1 to about 50 mg/kg per day. In certain embodiments, the dosage level is about 0.1 to about 40 mg/kg per day. In certain embodiments, the dosage level is about 0.5 to about 80 mg/kg per day. In certain embodiments, the dosage level is about 0.5 to about 40 mg/kg per day. In certain embodiments, the dosage level is about 0.5 to about 25 mg/kg per day. In certain embodiments, the dosage level is about 1 to about 80 mg/kg per day.
  • the dosage level is about 1 to about 75 mg/kg per day. In certain embodiments, the dosage level is about 1 to about 50 mg/kg per day. In certain embodiments, the dosage level is about 1 to about 40 mg/kg per day. In certain embodiments, the dosage level is about 1 to about 25 mg/kg per day.
  • the dosage level is about from 5.0 to 150 mg per day, and in certain embodiments from 10 to 100 mg per day. In certain embodiments, the dosage level is about 80 mg per day. In certain embodiments, the dosage level is about 40 mg per day.
  • the pharmaceutical compositions can be provided in the form of tablets containing 1.0 to 1,000 mg of the active ingredient, particularly about 1, about 5, about 10, about 15, about 20, about 25, about 30, about 40, about 45, about 50, about 75, about 80, about 100, about 150, about 200, about 250, about 300, about 400, about 500, about 600, about 750, about 800, about 900, and about 1,000 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated.
  • the pharmaceutical compositions can be provided in the form of tablets containing about 100 mg of the active ingredient.
  • the pharmaceutical compositions can be provided in the form of tablets containing about 80 mg of the active ingredient.
  • the pharmaceutical compositions can be provided in the form of tablets containing about 75 mg of the active ingredient. In certain embodiments, the pharmaceutical compositions can be provided in the form of tablets containing about 50 mg of the active ingredient. In certain embodiments, the pharmaceutical compositions can be provided in the form of tablets containing about 40 mg of the active ingredient. In certain embodiments, the pharmaceutical compositions can be provided in the form of tablets containing about 25 mg of the active ingredient.
  • the compositions may be administered on a regimen of 1 to 4 times per day, including once, twice, three times, and four times per day.
  • the compounds provided herein may also be combined or used in combination with other agents useful in the treatment, prevention, or amelioration of one or more symptoms of the diseases or conditions for which the compounds provided herein are useful, including tic disorders and other conditions commonly treated with antipsychotic medication.
  • the compounds provided herein may also be combined or used in combination with a typical antipsychotic drug.
  • the typical antipsychotic drug is fluphenazine, haloperidol, loxapine, molindone, perphenazine, pimozide, sulpiride, thioridazine, or trifluoperazine.
  • the antipsychotic drug is an atypical antipsychotic drug.
  • the atypical antipsychotic drug is aripiprazole, asenapine, clozapine, iloperidone, olanzapine, paliperidone, quetiapine, risperidone, or ziprasidone.
  • the atypical antipsychotic drug is clozapine.
  • Such other agents, or drugs may be administered, by a route and in an amount commonly used thereof, simultaneously or sequentially with the compounds provided herein.
  • a pharmaceutical composition containing such other drugs in addition to the compounds provided herein may be utilized, but is not required.
  • the pharmaceutical compositions provided herein include those that also contain one or more other active ingredients or therapeutic agents, in addition to the compounds provided herein.
  • the weight ratio of the compounds provided herein to the second active ingredient may be varied, and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used. Thus, for example, when the compounds provided herein are used in combination with the second drug, or a pharmaceutical composition containing such other drug, the weight ratio of the particulates to the second drug may range from about 1,000:1 to about 1: 1,000, or about 200:1 to about 1:200.
  • Combinations of the particulates provided herein and other active ingredients will generally also be within the aforementioned range, but in each case, an effective dose of each active ingredient should be used.
  • cytochrome P450 2D6 CYP2D6
  • NBI-98854 was administered as a 50 mg capsule orally as a single dose, twice during the treatment period on Days 1 and 6.
  • Ketoconazole was administered as a 200 mg tablet orally as a single dose, twice daily on Days 5 through 9.
  • Blood samples for PK analyses of NBI-98854 and its metabolites were collected before and after the Days 1 and 6 dosing: at 45 minutes prior to NBI-98854 dosing, and at 15, 30, 45 minutes, and 1, 1.5, 2, 3, 4, 8, 12, 18, 24, 48, 72 and 96 hours after NBI-98854 dosing or at early termination.
  • ketoconazole plasma concentrations were collected at 45 minutes pre- and 1 hour post-morning ketoconazole dosing on Day 5, at 1 and 8 hours post-morning ketoconazole dosing on Day 6, and at 1 hour post-morning ketoconazole dosing on Days 7-9 or at early termination.
  • Plasma concentrations of NBI-98854, its metabolites, and ketoconazole were summarized with descriptive statistics and in figures.
  • An analysis of variance (ANOVA) model was used to compare AUC 0- ⁇ and C max for NBI-98854 administered with ketoconazole (“test”) versus AUC 0- ⁇ and C max for NBI-98854 alone (“reference”). The t max was analyzed using non-parametric methods. The PK parameters for metabolites of NBI-98854 were also evaluated. Safety data were summarized using descriptive statistics.
  • the PK data for t max , T lag , t 1/2 , MRT, and Vz/F were rounded to 2 significant figures and all other parameters (AUC 0-24 , AUC 0-tlast , AUC 0- ⁇ , C max , and CL/F) were rounded to 3 significant figures. The last significant figure was rounded up if the digit to the right of it was ⁇ 5, and was rounded down if the digit to the right of it was ⁇ 4.
  • NBI-98854 was readily absorbed after oral administration alone or in combination with ketoconazole. Mean plasma concentrations were higher after combination treatment compared to NBI-98854 alone.
  • PK parameters for NBI-98854 after treatment with NBI-98854 alone or in combination with ketoconazole are summarized below.
  • Mean AUC 0- ⁇ for NBI-98854 after treatment with ketoconazole was approximately 2.1-fold greater than that after treatment with NBI-98854 alone.
  • Mean C max for NBI-98854 after treatment with NBI-98854 plus ketoconazole was also approximately 1.5-fold greater than that after treatment with NBI-98854 alone.
  • Median t max values were the same after treatment with NBI-98854 plus ketoconazole and NBI-98854 alone (0.75 hours) and mean t 1/2 values were higher after treatment with NBI-98854 plus ketoconazole than after treatment with NBI-98854 alone (20 and 16 hours, respectively).
  • the geometric CV % for NBI-98854 after treatment with NBI-98854 plus ketoconazole and NBI-98854 alone was generally similar for AUC 0- ⁇ , C max , t 1/2 , and MRT.
  • the geometric mean ratios and associated 90% confidence intervals (CIs) for AUC 0- ⁇ and C max for NBI-98854 after treatment with NBI-98854 alone or in combination with ketoconazole are provided below.
  • Geometric mean ratios for AUC 0- ⁇ and C max for NBI-98854 after treatment with NBI-98854 alone or in combination with ketoconazole were 213.7% and 151.1%, respectively.
  • the corresponding upper and lower 90% CI bounds for AUC 0- ⁇ (2.04 to 2.24) and C max (1.41 to 1.62) were outside the ‘no effect’ range of 80% to 125%, indicating an effect of treatment with ketoconazole on NBI-98854 AUC 0- ⁇ and C max .
  • Ratio a (%) (NBI-98854 with 90% Confidence Parameter ketoconazole vs. NBI-98854 alone) Interval b AUC 0- ⁇ 213.7 203.6, 224.3 C max (ng/mL) 151.1 140.6, 162.3 a Ratio of geometric least-squares means was based on a mixed model using log-transformed (base 10) data. b The 90% confidence interval for geometric mean ratio was based on least-squares means using log-transformed (base 10) data
  • Plasma concentrations for the active metabolite NBI-98782 increased after oral administration of NBI-98854 alone or in combination with ketoconazole.
  • PK parameters for NBI-98782 after treatment with NBI-98854 alone or in combination with ketoconazole are below.
  • Mean AUC 0- ⁇ for NBI-98782 after treatment with NBI-98854 in combination with ketoconazole was approximately 2.1-fold greater than mean AUC after treatment with NBI-98854 alone.
  • Mean C max for NBI-98782 after treatment with NBI-98854 plus ketoconazole was approximately 1.6-fold greater than that after treatment with NBI-98854 alone.
  • Median t max values were the same after treatment with NBI-98854 plus ketoconazole and NBI-98854 alone (4.0 hours) and mean t 1/2 was higher after treatment with NBI-98854 plus ketoconazole than after treatment with NBI-98854 alone (21 and 18 hours, respectively).
  • the variability in PK (ie, geometric CV %) for NBI-98782 after treatment with NBI-98854 alone and in combination with ketoconazole was generally similar for AUC, C max , t 1/2 , and MRT.
  • NBI-98782 geometric mean ratios and associated 90% CIs for AUC 0- ⁇ and C max after treatment with NBI-98854 alone versus in combination with ketoconazole are provided below.
  • Geometric mean ratios for AUC 0- ⁇ and C max for NBI-98782 after treatment with NBI-98854 alone versus in combination with ketoconazole were 206.7% and 162.9%, respectively, which were similar to the observed ratios for NBI-98854.
  • the corresponding upper and lower 90% CI bounds for both parameters were outside the ‘no effect’ range of 80% to 125%, indicating an effect of treatment with ketoconazole on NBI-98782 AUC 0- ⁇ and C max .
  • Ratio a (%) (NBI-98854 with 90% Confidence Parameter ketoconazole vs. NBI-98854 alone) Interval b AUC 0- ⁇ 206.7 198.4, 215.4 (ngxhr/mL) C max (ng/mL) 162.9 153.8, 172.5 a Ratio of geometric least-squares means was based on a mixed model using log-transformed (base 10) data. b The 90% confidence interval for geometric mean ratio was based on least-squares means using log-transformed (base 10) data
  • Plasma concentrations for the metabolite NBI-136110 decreased after oral administration of NBI-98854 in combination with ketoconazole compared with administration of NBI-98854 alone.
  • PK parameters for NBI-136110 after treatment with NBI-98854 alone or in combination with ketoconazole are summarized below.
  • mean C max for NBI-136110 after treatment with NBI-98854 plus ketoconazole was 22% of the observed value with NBI-98854 alone.
  • Median t max values were approximately 4-fold higher after treatment with NBI-98854 plus ketoconazole versus NBI-98854 alone.
  • Mean t1/2 for NBI-136110 after treatment with NBI-98854 with ketoconazole was higher than for NBI-98854 alone; however, because the calculated t 1/2 of NBI-136110 after treatment with NBI-98854 plus ketoconazole was longer than the blood collection period of 96 hours (120 hours), this value is of uncertain accuracy. Consequently, the AUC 0- ⁇ and MRT of NBI-136110 after treatment of NBI-98854 plus ketoconazole are also of uncertain accuracy.
  • the NBI-136110 geometric mean ratios and associated 90% CIs for AUC 0- ⁇ and C max after treatment with NBI-98854 alone or in combination with ketoconazole are provided below. Geometric mean ratios for AUC 0-tlast and C max for NBI-136110 after treatment with NBI-98854 alone or in combination with ketoconazole were 45.6% and 21.6%, respectively. The 90% CI for AUC 0-tlast and C max were outside the ‘no effect’ range of 0.80 to 1.25.
  • Ratio a (%) (NBI-98854 with 90% Confidence Parameter ketoconazole vs. NBI-98854 alone) Interval b AUC 0- ⁇ 45.6 41.8, 49.7 C max (ng/mL) 21.6 18.8, 24.8 a Ratio of geometric least-squares means was based on a mixed model using log-transformed (base 10) data. b The 90% confidence interval for geometric mean ratio was based on least-squares means using log-transformed (base 10) data
  • Ketoconazole plasma concentrations were measured on Day 5 at 45 minutes before and 1 hour after the morning ketoconazole dose, on Day 6 at 1 and 8 hours after the morning ketoconazole dose, and on Days 7 to 9 at 1 hour after the morning ketoconazole dose, or upon early termination._Measurable concentrations of ketoconazole were present in all postdose plasma samples analyzed. Mean concentrations of ketoconazole ranged from 1,643 to 5,177 ng/mL on Days 5 to 9 at 1 hour postdose.
  • NBI-98854 plus ketoconazole led to a 1.5-fold increase in peak plasma concentration (C max ) and a 2.1-fold increase in total exposure (AUC 0- ⁇ ) of NBI-98854 compared with administration of NBI-98854 alone.
  • the 90% CI for the geometric mean ratios (1.41 to 1.62 for C max and 2.04 to 2.24 for AUC 0- ⁇ ) were outside the ‘no effect’ range of 0.80 to 1.25.
  • NBI-98854 plus ketoconazole also led to a 1.6-fold increase in C max and a 2.1-fold increase in AUC 0- ⁇ of the metabolite NBI-98782 compared with administration of NBI-98854 alone.
  • the 90% CI for the geometric mean ratios were outside the ‘no effect’ range of 0.80 to 1.25.
  • NBI-98854 plus ketoconazole led to a 1.5-fold increase in peak plasma concentration (C max ) and a 2.1-fold increase in total exposure (AUC 0- ⁇ ) of NBI-98854 compared with administration of NBI-98854 alone.
  • the 90% confidence intervals (CI) for the geometric mean ratios (1.41 to 1.62 for C max and 2.04 to 2.24 for AUC 0- ⁇ ) were outside the ‘no effect’ range of 0.80 to 1.25.
  • NBI-98854 plus ketoconazole also led to a 1.6-fold increase in C max and a 2.1-fold increase in AUC 0- ⁇ of the metabolite NBI-98782 compared with administration of NBI-98854 alone.
  • the 90% CI for the geometric mean ratios were outside the ‘no effect’ range of 0.80 to 1.25.
  • AEs adverse events
  • clinical laboratory tests hematology, serum chemistry, and urinalysis
  • vital signs including orthostatic blood pressure
  • physical examinations including orthostatic blood pressure
  • ECGs 12-lead electrocardiograms
  • AEs were assigned to a treatment based on the time of the most recent treatment administered prior to the AE onset. TEAEs are summarized below. There were no pretreatment AEs. There were no deaths, SAEs, or discontinuations due to an AE reported in this study. Five subjects (20.8%) experienced 6 AEs during the study; all AEs occurred after the initial treatment with NBI-98854 alone. All AEs had an onset on Day 1 or 2. The only AE that occurred in >1 subject was headache (2 subjects, 8.3%). All AEs were mild in intensity. Two events were considered possibly related to study drug, 1 event of headache and the event of dizziness postural. The other event of headache was considered unlikely related to study drug.
  • NBI-98854 50 mg
  • NBI-98854 50 mg
  • Ketoconazole 200 mg
  • Term N 24
  • N 24
  • Overall 5 20.8) 0 Gastrointestinal Disorders 2 (8.3) 0 Constipation 1 (4.2) 0 Nausea 1 (4.2) 0 Injury, Poisoning and 1 (4.2) 0 Procedural Complications 1 (4.2) 0 Procedural dizziness Nervous System Disorders 3 (12.5) 0 Dizziness postural 1 (4.2) 0 Headache 2 (8.3) 0
  • NBI-98854 plus ketoconazole led to a 1.5-fold increase in C max and a 2.1-fold increase in AUC 0- ⁇ of NBI-98854 compared with administration of NBI-98854 alone.
  • NBI-98854 plus ketoconazole also led to a 1.6-fold increase in C max and a 2.1-fold increase in AUC 0- ⁇ of the metabolite NBI-98782 compared with administration of NBI-98854 alone.
  • the 90% CI for all the above parameters were outside the 90% CI range of 0.80 to 1.25.
  • NBI-98854 was well tolerated when administered alone and concomitantly with ketoconazole in healthy volunteers.
  • TBZ Upon oral administration, TBZ is reduced to form four discrete isomeric secondary alcohol metabolites, collectively referred to as dihydrotetrabenazine (DHTBZ), which contains three asymmetric carbon centers (C-2, C-3, and C-11 ⁇ ), which could hypothetically result in eight stereoisomers.
  • DHTBZ dihydrotetrabenazine
  • the affinity of each compound was measured by inhibition of [ 3 H]-DHTBZ binding to rat forebrain membranes.
  • the affinities relative to R,R,R-DHTBZ were also calculated and are presented. Data are reported as both the negative logarithm of the Ki (pKi) for statistical calculation with the normally distributed binding parameter used to determine the mean and SEM.
  • the Ki value was determined from the mean pKi as 10(-pKi).
  • VMAT2 Affinity pK i mean Relative to Compound K i ,, nm (SEM) N R,R,R-DHTBZ a R,R,R-DHTBZ 4.2 8.38 (0.42) 27 1.0 S,R,R-DHTBZ 9.7 8.01 (0.32) 6 2.3 S,S,S-DHTBZ 250 6.60 (0.22) 4 60 R,S,S-DHTBZ 690 6.16 (0.05) 5 160 a Affinity relative to R,R,R-DHTBZ was calculated using the K i value determined in the same study
  • valbenazine The primary metabolic clearance pathways of valbenazine (VBZ, NBI-98854) are hydrolysis (to form R,R,R-DHTBZ) and mono-oxidation (to form the metabolite NBI-136110).
  • R,R,R-DHTBZ and NBI-136110 the two most abundant circulating metabolites of VBZ, are formed gradually and their plasma concentrations decline with half-lives similar to VBZ.
  • the affinity of each compound was measured by inhibition of [ 3 H]-DHTBZ binding to either human platelets or rat striatal membranes.
  • the primary metabolite R,R,R-DHTBZ was the most potent inhibitor of VMAT2 in rat striatum and human platelet homogenates.
  • Rat Striatum Human Platelets Affinity Affinity Relative Relative Relative K i , pK i mean to R,R,R- K i , pK i mean to R,R,R- Compound nm (SEM) DHTBZ nm (SEM) DHTBZ Valbenazine 110 6.95 (0.02) 39 150 6.82 45 (0.02) R,R,R- 1.98 8.70 (0.09) 1.0 3.1 8.52 1.0 DHTBZ (0.03) NBI-136610 160 6.80 (0.02) 57 220 6.65(0.04) 67
  • VBZ and NBI-136110 had similar effects on VMAT2 inhibition, but with Ki values that were approximately 40-65 times the Ki values (lower affinity) of R,R,R-DHTBZ.
  • the multi-target activity screen of more than 80 targets for these compounds demonstrated that VBZ and its metabolites, R,R,R-DHTBZ and NBI-136110, did not inhibit the binding of cognate ligands to any of the targets by more than 50% at concentrations of 1-10 ⁇ M.
  • the other three DHTBZ stereoisomers S,R,R-DHTBZ, S,S,S-DHTBZ, R,S,S-DHTBZ
  • S,S,S-DHTBZ S,S,S-DHTBZ, R,S,S-DHTBZ
  • Results expressed as percent of control specific binding: (tested compound specific binding/control specific binding) ⁇ 100. All compounds were tested at 1 or 10 ⁇ M final concentration and results are an excerpt of a larger 80 target panel performed as an initial screen at Cerep (n 2 for each compound at each target). Bolded results (>50%) indicate activity at target receptor.
  • S,S,S- and R,S,S-netabolites were tested as a 50/50 mixture.
  • radioligand binding assays were performed for dopamine, serotonin and adrenergic receptor subtypes as well as the transporters for dopamine (DAT), serotonin (SERT), and norepinephrine (NET) for the common metabolite of TBZ and VBZ (R,R,R-DHTBZ) and the other relevant metabolites unique to TBZ and VBZ.
  • DAT dopamine
  • SERT serotonin
  • NET norepinephrine
  • R,R,R-DHTBZ common metabolite of TBZ and VBZ
  • This detailed analysis revealed the high specificity of R,R,R-DHTBZ for the VMAT2 transporter and the non-specific activities of the other TBZ metabolites, including relatively high affinity for dopamine and serotonin receptor subtypes.
  • the R,R,R-DHTBZ metabolite showed the greatest non-selectivity with respect to the monoamine receptors. None of the TBZ or VBZ metabolites had any affinity for the monoamine transporters DAT, SERT or NET.
  • the functional activity for the human VMAT1 transporter of these compounds was tested in cells expressing VMAT1. While the non-selective irreversible high-affinity uptake inhibitor of VMAT1, reserpine, substantially inhibited uptake through VMAT1, there was no significant inhibitory activity of TBZ, VBZ, or its metabolites R,R,R-DHTBZ or NBI-136110 at concentrations up to 10 ⁇ M. For both VMAT1 and VMAT2, uptake was measured in the untransfected host cells and was found to be similar to transfected cells in the presence of excess reserpine.
  • Radioligand binding assays and the broad panel screen indicate that in addition to varying potency at the VMAT2 transporter, two of the other DHTBZ metabolites of TBZ (S,S,S-DHTBZ and R,S,S-DHTBZ) interact with D1 and D2 receptors. Since VBZ is not metabolized to either of these DHTBZ stereoisomers, its effects on postsynaptic dopamine receptors either directly or indirectly through the metabolites are non-existent.
  • results from the broad panel screen indicate that VBZ and its major metabolites (R,R,R-DHTBZ and NBI-136110) have little to no affinity for more than 80 binding sites, including receptors, monoamine transporters, and ion channels. This profile suggests a low potential for off-target pharmacological effects.
  • uptake studies using TBZ, VBZ and its metabolites, R,R,R-DHTBZ and NBI-136110 confirmed the selectivity of these compounds for VMAT2 as they had no significant effect on the uptake of monoamines through VMAT1 compared to reserpine, a known VMAT1/VMAT2 inhibitor.
  • VBZ The selectivity and specificity of VBZ was distinctively demonstrated using two in vivo surrogate measures of pharmacological effects.
  • Ptosis known to occur via adrenergic activation and prolactin release from the pituitary, modulated through the D2 dopamine receptor, demonstrated the difference between treatment with TBZ and VBZ.
  • TBZ, VBZ and R,R,R-DHTBZ induced ptosis in an equivalent manner.

Abstract

Provided are methods of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof wherein the patient is also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.

Description

  • This application claims the benefit of U.S. Provisional Application No. 62/451,605, filed Jan. 27, 2017, which is incorporated herein by reference for all purposes.
  • Dysregulation of dopaminergic systems is integral to several central nervous system (CNS) disorders, including neurological and psychiatric diseases and disorders. These neurological and psychiatric diseases and disorders include hyperkinetic movement disorders, and conditions such as schizophrenia and mood disorders. The transporter protein vesicular monoamine transporter-2 (VMAT2) plays an important role in presynaptic dopamine release and regulates monoamine uptake from the cytoplasm to the synaptic vesicle for storage and release.
  • Despite the advances that have been made in this field, there remains a need for new therapeutic products useful to treatment of neurological and psychiatric diseases and disorders and other related diseases or conditions described herein. One such agent is valbenazine, which has the following chemical structure:
  • Figure US20200093808A1-20200326-C00001
  • A formulation of valbenazine:4-toluenesulfonate (1:2) (referred to herein as “valbenazine ditosylate”) has been previously reported in the FDA approved drug label Ingrezza®.
  • The cytochrome P450 enzyme system (CYP450) is responsible for the biotransformation of drugs from active substances to inactive metabolites that can be excreted from the body. In addition, the metabolism of certain drugs by CYP450 can alter their PK profile and result in sub-therapeutic plasma levels of those drugs over time.
  • There are more than 1500 known P450 sequences which are grouped into families and subfamily. The cytochrome P450 gene superfamily is composed of at least 207 genes that have been named based on the evolutionary relationships of the cytochromes P450. For this nomenclature system, the sequences of all of the cytochrome P450 genes are compared, and those cytochromes P450 that share at least 40% identity are defined as a family (designated by CYP followed by a Roman or Arabic numeral, e.g., CYP3), and further divided into subfamilies (designated by a capital letter, e.g., CYP3A), which are comprised of those forms that are at least 55% related by their deduced amino acid sequences. Finally, the gene for each individual form of cytochrome P450 is assigned an Arabic number (e.g., CYP3A4).
  • CYP3A isoenzyme is a member of the cytochrome P450 superfamily which constitutes up to 60% of the total human liver microsomal cytochrome P450 and has been found in alimentary passage of stomach and intestines and livers. CYP3A has also been found in kidney epithelial cells, jejunal mucosa, and the lungs. CYP3A is one of the most abundant subfamilies in cytochrome P450 superfamily.
  • At least five (5) forms of CYPs are found in human CYP3A subfamily, and these forms are responsible for the metabolism of a large number of structurally diverse drugs. In non-induced individuals, CYP3A may constitute 15% of the P450 enzymes in the liver; in enterocytes, members of the CYP3A subfamily constitute greater than 70% of the CYP-containing enzymes.
  • CYP3A is responsible for metabolism of a large number of drugs including nifedipine, macrolide antibiotics including erythromycin and troleandomycin, cyclosporin, FK506, teffenadine, tamoxifen, lidocaine, midazolam, triazolam, dapsone, diltiazem, lovastatin, quinidine, ethylestradiol, testosterone, and alfentanil. CYP3A is involved in erythromycin N-demethylation, cyclosporine oxidation, nifedipine oxidation, midazolam hydroxylation, testosterone 6-β-hydroxylation, and cortisol 6-β-hydroxylation. CYP3A has also been shown to be involved in both bioactivation and detoxication pathways for several carcinogens in vitro.
  • There is a significant, unmet need for methods for administering a VMAT2 inhibitor, such as valbenazine or (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof, wherein the patient is also being treated with another substance which may interact with the VMAT2 inhibitor, such as a CYP3A4 inhibitor. The present disclosure fulfills these and other needs, as evident in reference to the following disclosure.
  • BRIEF SUMMARY
  • Provided is a method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof wherein the patient is also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering the VMAT2 inhibitor in an amount equivalent to about 40 mg of valbenazine free base once daily to the patient.
  • Also provided is a method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof wherein the patient is also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering a therapeutically effective amount of the VMAT2 inhibitor to the patient, wherein the therapeutically effective amount of the VMAT2 inhibitor is less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • Also provided is a method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof, comprising: administering to the patient a therapeutically effective amount of the VMAT2 inhibitor, subsequently determining that the patient is to begin treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor, and administering the VMAT2 inhibitor in an amount equivalent to about 40 mg of valbenazine free base once daily to the patient.
  • Also provided is a method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof, comprising: administering a therapeutically effective amount of the VMAT2 inhibitor to the patient, subsequently determining that the patient is to begin treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor, and administering the VMAT2 inhibitor in an amount that is less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • Also provided is a method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof wherein the patient is also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering a therapeutically effective amount of the VMAT2 inhibitor to the patient, wherein the administration produces a mean valbenazine Cmax that is about 1 to about 2 fold higher than the mean valbenazine Cmax for a patient who is not being administered a strong CYP3A4 inhibitor and/or a mean valbenazine AUC0-∞ that is about 1.5 to about 2.5 fold higher than the mean valbenazine AUC0-∞ for a patient who is not being administered a strong CYP3A4 inhibitor.
  • Also provided is a method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof to a patient in need thereof wherein patient is also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering a therapeutically effective amount of the VMAT2 inhibitor, wherein the administration produces a mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol Cmax that is about 1 to about 2 fold higher than the mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol Cmax for a patient who is not being administered a strong CYP3A4 inhibitor and/or a mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol AUC0-∞ that is about 1.5 to about 2.5 fold higher than the mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol AUC0-∞ for a patient who is not being administered a strong CYP3A4 inhibitor.
  • These and other aspects of the invention will be apparent upon reference to the following detailed description. To this end, various references are set forth herein which describe in more detail certain background information, procedures, compounds, and/or compositions, and are each hereby incorporated by reference in their entirety.
  • DETAILED DESCRIPTION
  • In the following description, certain specific details are set forth in order to provide a thorough understanding of various embodiments. However, one skilled in the art will understand that the invention may be practiced without these details. In other instances, well-known structures have not been shown or described in detail to avoid unnecessarily obscuring descriptions of the embodiments. Unless the context requires otherwise, throughout the specification and claims which follow, the word “comprise” and variations thereof, such as, “comprises” and “comprising” are to be construed in an open, inclusive sense, that is, as “including, but not limited to.” Further, headings provided herein are for convenience only and do not interpret the scope or meaning of the claimed invention.
  • Reference throughout this specification to “one embodiment” or “an embodiment” or “some embodiments” or “a certain embodiment” means that a particular feature, structure or characteristic described in connection with the embodiment is included in at least one embodiment. Thus, the appearances of the phrases “in one embodiment” or “in an embodiment” or “in some embodiments” or “in a certain embodiment” in various places throughout this specification are not necessarily all referring to the same embodiment. Furthermore, the particular features, structures, or characteristics may be combined in any suitable manner in one or more embodiments.
  • Also, as used in this specification and the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the content clearly dictates otherwise.
  • As used herein, “valbenazine” may be referred to as (S)-2-amino-3-methyl-butyric acid (2R, 3R,11bR)-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-yl ester; or as L-Valine, (2R,3R,11bR)-1,3,4,6,7,11b-hexahydro-9,10-dimethoxy-3-(2-methylpropyl)-2H-benzo[a]quinolizin-2-yl ester or as NBI-98854.
  • As used herein, “(+)-α-HTBZ” means the compound which is an active metabolite of valbenazine having the structure:
  • Figure US20200093808A1-20200326-C00002
  • (+)-α-HTBZ may be referred to as (2R, 3R, 11bR) or as (+)-α-DHTBZ or as (+)-α-HTBZ or as R,R,R-DHTBZ or as (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol; or as (2R, 3R,11bR)-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol or as NBI-98782.
  • As used herein, “NBI-136110” means the compound which is a metabolite of valbenazine having the structure:
  • Figure US20200093808A1-20200326-C00003
  • As used herein, “isotopic variant” means a compound that contains an unnatural proportion of an isotope at one or more of the atoms that constitute such a compound. In certain embodiments, an “isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, hydrogen (1H), deuterium (2H), tritium (3H), carbon-11 (11C), carbon-12 (12C), carbon-13 (13C), carbon-14 (14C), nitrogen-13 (13N), nitrogen-14 (14N), nitrogen-15 (15N), oxygen-14 (14O), oxygen-15 (150), oxygen-16 (16O), oxygen-17 (17O), oxygen-18 (18O), fluorine-17 (17F), fluorine-18 (18F), phosphorus-31 (31P), phosphorus-32 (32P), phosphorus-33 (33P), sulfur-32 (32S), sulfur-33 (33S), sulfur-34 (34S), sulfur-35 (35S), sulfur-36 (36S), chlorine-35 (35Cl), chlorine-36 (36Cl), chlorine-37 (37Cl), bromine-79 (79Br), bromine-81 (81Br), iodine-123 (123I), iodine-125 (125I), iodine-127 (127I), iodine-129 (129I), and iodine-131 (131I). In certain embodiments, an “isotopic variant” of a compound is in a stable form, that is, non-radioactive. In certain embodiments, an “isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, hydrogen (1H), deuterium (2H), carbon-12 (12C), carbon-13 (13C), nitrogen-14 (14N), nitrogen-15 (15N), oxygen-16 (16O), oxygen-17 (17O), and oxygen-18 (18O). In certain embodiments, an “isotopic variant” of a compound is in an unstable form, that is, radioactive. In certain embodiments, an “isotopic variant” of a compound contains unnatural proportions of one or more isotopes, including, but not limited to, tritium (3H), carbon-11 (11C), carbon-14 (14C), nitrogen-13 (13N), oxygen-14 (14O), and oxygen-15 (15O). It will be understood that, in a compound as provided herein, any hydrogen can be 2H, as example, or any carbon can be 13C, as example, or any nitrogen can be 15N, as example, and any oxygen can be 18O, as example, where feasible according to the judgment of one of skill in the art. In certain embodiments, an “isotopic variant” of a compound contains an unnatural proportion of deuterium.
  • With regard to the compounds provided herein, when a particular atomic position is designated as having deuterium or “D” or “d”, it is understood that the abundance of deuterium at that position is substantially greater than the natural abundance of deuterium, which is about 0.015%. A position designated as having deuterium typically has a minimum isotopic enrichment factor of, in certain embodiments, at least 1000 (15% deuterium incorporation), at least 2000 (30% deuterium incorporation), at least 3000 (45% deuterium incorporation), at least 3500 (52.5% deuterium incorporation), at least 4000 (60% deuterium incorporation), at least 4500 (67.5% deuterium incorporation), at least 5000 (75% deuterium incorporation), at least 5500 (82.5% deuterium incorporation), at least 6000 (90% deuterium incorporation), at least 6333.3 (95% deuterium incorporation), at least 6466.7 (97% deuterium incorporation), at least 6600 (99% deuterium incorporation), or at least 6633.3 (99.5% deuterium incorporation) at each designated deuterium position. The isotopic enrichment of the compounds provided herein can be determined using conventional analytical methods known to one of ordinary skill in the art, including mass spectrometry, nuclear magnetic resonance spectroscopy, and crystallography.
  • As used herein, a substance is a “substrate” of enzyme activity when it can be chemically transformed by action of the enzyme on the substance. Substrates can be either activated or deactivated by the enzyme.
  • “Enzyme activity” refers broadly to the specific activity of the enzyme (i.e., the rate at which the enzyme transforms a substrate per mg or mole of enzyme) as well as the metabolic effect of such transformations.
  • A substance is an “inhibitor” of enzyme activity when the specific activity or the metabolic effect of the specific activity of the enzyme can be decreased by the presence of the substance, without reference to the precise mechanism of such decrease. For example, a substance can be an inhibitor of enzyme activity by competitive, non-competitive, allosteric or other type of enzyme inhibition, by decreasing expression of the enzyme, or other direct or indirect mechanisms. Co-administration of a given drug with an inhibitor may decrease the rate of metabolism of that drug through the metabolic pathway listed.
  • A substance is an “inducer” of enzyme activity when the specific activity or the metabolic effect of the specific activity of the enzyme can be increased by the presence of the substance, without reference to the precise mechanism of such increase. For example, a substance can be an inducer of enzyme activity by increasing reaction rate, by increasing expression of the enzyme, by allosteric activation or other direct or indirect mechanisms. Co-administration of a given drug with an enzyme inducer may increase the rate of excretion of the drug metabolized through the pathway indicated.
  • Any of these effects on enzyme activity can occur at a given concentration of active agent in a single sample, donor, or patient without regard to clinical significance. It is possible for a substance to be a substrate, inhibitor, or inducer of an enzyme activity. For example, the substance can be an inhibitor of enzyme activity by one mechanism and an inducer of enzyme activity by another mechanism. The function (substrate, inhibitor, or inducer) of the substance with respect to activity of an enzyme can depend on environmental conditions.
  • Lists of inhibitors, inducers and substrates for CYP3A4 can be found, for instance, at http://www.genemedrx.com/Cytochrome_P450_Metabolism_Table.php, and other sites and http://www.ildcare.eu/downloads/artseninfo/ drugs_metabolized_by_cyp450s.pdf
  • As used herein, a “strong CYP3A4 inhibitor” is a compound that increases the area under the concentration time curve (AUC) of a sensitive index substrate of the CYP3A4 pathway by ≥5-fold. Index substrates predictably exhibit exposure increase due to inhibition or induction of a given metabolic pathway and are commonly used in prospective clinical drug-drug interaction studies. Sensitive index substrates are index substrates that demonstrate an increase in AUC of ≥5-fold with strong index inhibitors of a given metabolic pathway in clinical drug-drug interaction studies studies. Examples of sensitive index substrates for the CYP2D6 pathway are midazolam and triazolam. See, e.g., Drug Development and Drug Interactions: Table of Substrates, Inhibitor and Inducers at https://www.fda.gov/drugs/developmentapprovalprocess/developmentresources/druginteractionslabeling/ucm093664.htm and http://www.ildcare.eu/downloads/artseninfo/drugs_metabolized_by_cyp450s.pdf.
  • As used herein, “hyperkinetic disorder” or “hyperkinetic movement disorder” or “hyperkinesias” refers to disorders or diseases characterized by excessive, abnormal, involuntary movements. These neurological disorders include tremor, dystonia, myoclonus, athetosis, Huntington's disease, tardive dyskinesia, Tourette syndrome, dystonia, hemiballismus, chorea, senile chorea, or tics.
  • As used herein, “tardive syndrome” encompasses but is not limited to tardive dyskinesia, tardive dystonia, tardive akathisia, tardive tics, myoclonus, tremor and withdrawal-emergent syndrome. Tardive dyskinesia is characterized by rapid, repetitive, stereotypic, involuntary movements of the face, limbs, or trunk.
  • As used herein, “about” means ±20% of the stated value, and includes more specifically values of ±10%, ±5%, ±2% and ±1% of the stated value.
  • As used herein, “AUC” refers to the area under the curve, or the integral, of the plasma concentration of an active pharmaceutical ingredient or metabolite over time following a dosing event.
  • As used herein “AUC0-4” is the integral under the plasma concentration curve from time 0 (dosing) to time “t”.
  • As used herein, “AUC0-∞” is the AUC from time 0 (dosing) to time infinity. Unless otherwise stated, AUC refers to AUC0-∞. Often a drug is packaged in a salt form, for example valbenazine ditosylate, and the dosage form strength refers to the mass of this salt form or the equivalent mass of the corresponding free base, valbenazine.
  • As used herein, Cmax is a pharmacokinetic parameter denoting the maximum observed blood plasma concentration following delivery of an active pharmaceutical ingredient. Cmax occurs at the time of maximum plasma concentration, tmax.
  • As used herein, “co-administer” and “co-administration” and variants thereof mean the administration of at least two drugs to a patient either subsequently, simultaneously, or consequently proximate in time to one another (e.g., within the same day, or week or period of 30 days, or sufficiently proximate that each of the at least two drugs can be simultaneously detected in the blood plasma). When co-administered, two or more active agents can be co-formulated as part of the same composition or administered as separate formulations. This also may be referred to herein as “concomitant” administration or variants thereof.
  • As used herein, “adjusting administration”, “altering administration”, “adjusting dosing”, or “altering dosing ” are all equivalent and mean tapering off, reducing or increasing the dose of the substance, ceasing to administer the substance to the patient, or substituting a different active agent for the substance.
  • As used herein, “administering to a patient” refers to the process of introducing a composition or dosage form into the patient via an art-recognized means of introduction.
  • As used herein the term “disorder” is intended to be generally synonymous, and is used interchangeably with, the terms “disease,” “syndrome,” and “condition” (as in medical condition), in that all reflect an abnormal condition of the human or animal body or of one of its parts that impairs normal functioning, is typically manifested by distinguishing signs and symptoms.
  • As used herein, a “dose” means the measured quantity of an active agent to be taken at one time by a patient. In certain embodiments, wherein the active agent is not valbenazine free base, the quantity is the molar equivalent to the corresponding amount of valbenazine free base. For example, often a drug is packaged in a pharmaceutically acceptable salt form, for example valbenazine ditosylate, and the dosage for strength refers to the mass of the molar equivalent of the corresponding free base, valbenazine. As an example, 73 mg of valbenazine tosylate is the molar equivalent of 40 mg of valbenazine free base.
  • As used herein, “dosing regimen” means the dose of an active agent taken at a first time by a patient and the interval (time or symptomatic) at which any subsequent doses of the active agent are taken by the patient such as from about 20 to about 160 mg once daily, e.g., about 20, about 40, about 60, about 80, about 100, about 120, or about 160 mg once daily. The additional doses of the active agent can be different from the dose taken at the first time.
  • As used herein, “effective amount” and “therapeutically effective amount” of an agent, compound, drug, composition or combination is an amount which is nontoxic and effective for producing some desired therapeutic effect upon administration to a subject or patient (e.g., a human subject or patient). The precise therapeutically effective amount for a subject may depend upon, e.g., the subject's size and health, the nature and extent of the condition, the therapeutics or combination of therapeutics selected for administration, and other variables known to those of skill in the art. The effective amount for a given situation is determined by routine experimentation and is within the judgment of the clinician.
  • As used herein, “informing” means referring to or providing published material, for example, providing an active agent with published material to a user; or presenting information orally, for example, by presentation at a seminar, conference, or other educational presentation, by conversation between a pharmaceutical sales representative and a medical care worker, or by conversation between a medical care worker and a patient; or demonstrating the intended information to a user for the purpose of comprehension.
  • As used herein, “labeling” means all labels or other means of written, printed, graphic, electronic, verbal, or demonstrative communication that is upon a pharmaceutical product or a dosage form or accompanying such pharmaceutical product or dosage form.
  • As used herein, “a “medical care worker” means a worker in the health care field who may need or utilize information regarding an active agent, including a dosage form thereof, including information on safety, efficacy, dosing, administration, or pharmacokinetics. Examples of medical care workers include physicians, pharmacists, physician's assistants, nurses, aides, caretakers (which can include family members or guardians), emergency medical workers, and veterinarians.
  • As used herein, “Medication Guide” means an FDA-approved patient labeling for a pharmaceutical product conforming to the specifications set forth in 21 CFR 208 and other applicable regulations which contains information for patients on how to safely use a pharmaceutical product. A medication guide is scientifically accurate and is based on, and does not conflict with, the approved professional labeling for the pharmaceutical product under 21 CFR 201.57, but the language need not be identical to the sections of approved labeling to which it corresponds. A medication guide is typically available for a pharmaceutical product with special risk management information.
  • As used herein, “ patient ” or “individual” or “ subject” means a mammal, including a human, for whom or which therapy is desired, and generally refers to the recipient of the therapy.
  • As used herein, “patient package insert” means information for patients on how to safely use a pharmaceutical product that is part of the FDA-approved labeling. It is an extension of the professional labeling for a pharmaceutical product that may be distributed to a patient when the product is dispensed which provides consumer-oriented information about the product in lay language, for example it may describe benefits, risks, how to recognize risks, dosage, or administration.
  • As used herein, “pharmaceutically acceptable” refers to a material that is not biologically or otherwise undesirable, i.e., the material may be incorporated into a pharmaceutical composition administered to a patient without causing any undesirable biological effects or interacting in a deleterious manner with any of the other components of the composition in which it is contained. When the term “pharmaceutically acceptable” is used to refer to a pharmaceutical carrier or excipient, it is implied that the carrier or excipient has met the required standards of toxicological and manufacturing testing or that it is included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug administration. “Pharmacologically active” (or simply “active”) as in a “pharmacologically active” (or “active”) derivative or analog, refers to a derivative or analog having the same type of pharmacological activity as the parent compound and approximately equivalent in degree. The term “pharmaceutically acceptable salts” include acid addition salts which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
  • As used herein, a “product” or “pharmaceutical product” means a dosage form of an active agent plus published material, and optionally packaging.
  • As used herein, “product insert” means the professional labeling (prescribing information) for a pharmaceutical product, a patient package insert for the pharmaceutical product, or a medication guide for the pharmaceutical product.
  • As used herein, “professional labeling” or “prescribing information” means the official description of a pharmaceutical product approved by a regulatory agency (e.g., FDA or EMEA) regulating marketing of the pharmaceutical product, which includes a summary of the essential scientific information needed for the safe and effective use of the drug, such as, for example indication and usage; dosage and administration; who should take it; adverse events (side effects); instructions for use in special populations (pregnant women, children, geriatric, etc.); safety information for the patient, and the like.
  • As used herein, “published material” means a medium providing information, including printed, audio, visual, or electronic medium, for example a flyer, an advertisement, a product insert, printed labeling, an internet web site, an internet web page, an internet pop-up window, a radio or television broadcast, a compact disk, a DVD, an audio recording, or other recording or electronic medium.
  • As used herein, “risk” means the probability or chance of adverse reaction, injury, or other undesirable outcome arising from a medical treatment. An “acceptable risk” means a measure of the risk of harm, injury, or disease arising from a medical treatment that will be tolerated by an individual or group. Whether a risk is “acceptable” will depend upon the advantages that the individual or group perceives to be obtainable in return for taking the risk, whether they accept whatever scientific and other advice is offered about the magnitude of the risk, and numerous other factors, both political and social. An “acceptable risk” of an adverse reaction means that an individual or a group in society is willing to take or be subjected to the risk that the adverse reaction might occur since the adverse reaction is one whose probability of occurrence is small, or whose consequences are so slight, or the benefits (perceived or real) of the active agent are so great. An “unacceptable risk” of an adverse reaction means that an individual or a group in society is unwilling to take or be subjected to the risk that the adverse reaction might occur upon weighing the probability of occurrence of the adverse reaction, the consequences of the adverse reaction, and the benefits (perceived or real) of the active agent. “At risk” means in a state or condition marked by a high level of risk or susceptibility. Risk assessment consists of identifying and characterizing the nature, frequency, and severity of the risks associated with the use of a product.
  • As used herein, “safety” means the incidence or severity of adverse events associated with administration of an active agent, including adverse effects associated with patient-related factors (e.g., age, gender, ethnicity, race, target illness, abnormalities of renal or hepatic function, co-morbid illnesses, genetic characteristics such as metabolic status, or environment) and active agent-related factors (e.g., dose, plasma level, duration of exposure, or concomitant medication).
  • As used herein, “tmax” is a pharmacokinetic parameter denoting the time to maximum blood plasma concentration following delivery of an active pharmaceutical ingredient
  • As used herein, “t1/2” or “plasma half-life” or “elimination half-life” or the like is a pharmacokinetic parameter denoting the apparent plasma terminal phase half-life, i.e., the time, after absorption and distribution of a drug is complete, for the plasma concentration to fall by half
  • As used herein, “treating” or “treatment” refers to therapeutic applications to slow or stop progression of a disorder, prophylactic application to prevent development of a disorder, and/or reversal of a disorder. Reversal of a disorder differs from a therapeutic application which slows or stops a disorder in that with a method of reversing, not only is progression of a disorder completely stopped, cellular behavior is moved to some degree, toward a normal state that would be observed in the absence of the disorder.
  • As used herein, “VMAT2” refers to human vesicular monoamine transporter isoform 2, an integral membrane protein that acts to transport monoamines, particularly neurotransmitters such as dopamine, norepinephrine, serotonin, and histamine, from cellular cytosol into synaptic vesicles.
  • As used herein, the term “VMAT2 inhibitor”, “inhibit VMAT2”, or “inhibition of VMAT2” refers to the ability of a compound disclosed herein to alter the function of VMAT2. A VMAT2 inhibitor may block or reduce the activity of VMAT2 by forming a reversible or irreversible covalent bond between the inhibitor and VMAT2 or through formation of a noncovalently bound complex. Such inhibition may be manifest only in particular cell types or may be contingent on a particular biological event. The term “VMAT2 inhibitor”, “inhibit VMAT2”, or “inhibition of VMAT2” also refers to altering the function of VMAT2 by decreasing the probability that a complex forms between a VMAT2 and a natural substrate.
  • Provided is a method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof wherein the patient is also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering the VMAT2 inhibitor in an amount equivalent to about 40 mg of valbenazine free base once daily to the patient.
  • Also provided is a method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof wherein the patient is also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering a therapeutically effective amount of the VMAT2 inhibitor to the patient, wherein the therapeutically effective amount of the VMAT2 inhibitor is less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • In certain embodiments, the method further comprises determining whether the patient is being administered a strong CYP3A4 inhibitor.
  • Also provided is a method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof, comprising: administering to the patient a therapeutically effective amount of the VMAT2 inhibitor, subsequently determining that the patient is to begin treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor, and administering the VMAT2 inhibitor in an amount equivalent to about 40 mg of valbenazine free base once daily to the patient.
  • Also provided is a method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof, comprising: administering a therapeutically effective amount of the VMAT2 inhibitor to the patient, subsequently determining that the patient is to begin treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor, and administering the VMAT2 inhibitor in an amount that is less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • Also provided is a method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof wherein the patient is also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering a therapeutically effective amount of the VMAT2 inhibitor to the patient, wherein the administration produces a mean valbenazine Cmax that is about 1 to about 2 fold higher than the mean valbenazine Cmax for a patient who is not being administered a strong CYP3A4 inhibitor and/or a mean valbenazine AUC0-∞ that is about 1.5 to about 2.5 fold higher than the mean valbenazine AUC0-∞ for a patient who is not being administered a strong CYP3A4 inhibitor.
  • Also provided is a method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof to a patient in need thereof wherein patient is also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering a therapeutically effective amount of the VMAT2 inhibitor, wherein the administration produces a mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol Cmax that is about 1 to about 2 fold higher than the mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol Cmax for a patient who is not being administered a strong CYP3A4 inhibitor and/or a mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol AUC0-∞ that is about 1.5 to about 2.5 fold higher than the mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol AUC0-∞ for a patient who is not being administered a strong CYP3A4 inhibitor.
  • In certain embodiments, the method further comprises informing the patient or a medical care worker that administration of the VMAT2 inhibitor to patients who is also being administered a strong CYP3A4 inhibitor in higher exposure of valbenazine and/or (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol than administration of the VMAT2 inhibitor to a patient who is not being administered a strong CYP3A4 inhibitor.
  • In certain embodiments, the method further comprises informing the patient or a medical care worker that administration of the VMAT2 inhibitor to a patient who is also being administered a strong CYP3A4 inhibitor may result in increased risk of one or more exposure-related adverse reactions than administration of the VMAT2 inhibitor to a patient who is not being administered a strong CYP3A4 inhibitor.
  • In certain embodiments, the one or more exposure-related adverse reactions is chosen from somnolence, anticholinergic effects, balance disorders or falls, headache, akathisia, vomiting, nausea, arthralgia, QT prolongation, increase in blood glucose, increase in weight, respiratory infections, drooling, dyskinesia, extrapyramidal symptoms (non-akathisia), anxiety, insomnia, increase in prolactin, increase in alkaline phosphatase, and increase in bilirubin. In certain embodiments, the one or more exposure-related adverse reactions is chosen from somnolence, anticholinergic effects, balance disorders or falls, headache, akathisia, vomiting, nausea, arthralgia, and QT prolongation. In certain embodiments, the one or more exposure-related adverse reactions is chosen from somnolence and QT prolongation.
  • In certain embodiments, the method further comprises informing the patient or a medical care worker that co-administration of the VMAT2 inhibitor and the CYP3A4 inhibitor may prolong the patient's QT interval.
  • In certain embodiments, the method further comprises informing the patient or a medical care worker that administration of the VMAT2 inhibitor to a patient who is also being administered a strong CYP3A4 inhibitor may prolong the patient's QT interval than administration of the VMAT2 inhibitor to a patient who is also being administered a strong CYP3A4 inhibitor.
  • In certain embodiments, the strong CYP3A4 inhibitor is chosen from clarithromycin, chloramphenicol, cobicistat, indinavir, itraconazole, ketoconazole, nefazodone, nelfinavir, ritonavir, saquinavir, and telithromycin. In certain embodiments, the strong CYP3A4 inhibitor is chosen from clarithromycin, itraconazole, and ketoconazole. In certain embodiments, the strong CYP3A4 inhibitor is ketoconazole.
  • In certain embodiments, the VMAT2 inhibitor is administered to the patient to treat a neurological or psychiatric disease or disorder. In certain embodiments, the neurological or psychiatric disease or disorder is a hyperkinetic movement disorder, mood disorder, bipolar disorder, schizophrenia, schizoaffective disorder, mania in mood disorder, depression in mood disorder, treatment-refractory obsessive compulsive disorder, neurological dysfunction associated with Lesch-Nyhan syndrome, agitation associated with Alzheimer's disease, Fragile X syndrome or Fragile X-associated tremor-ataxia syndrome, autism spectrum disorder, Rett syndrome, or chorea-acanthocytosis.
  • In certain embodiments, the neurological or psychiatric disease or disorder is a hyperkinetic movement disorder. In certain embodiments, the hyperkinetic movement disorder is tardive dyskinesia. In certain embodiments, the hyperkinetic movement disorder is Tourette's syndrome. In certain embodiments, the hyperkinetic movement disorder is Huntington's disease. In certain embodiments, the hyperkinetic movement disorder is tics. In certain embodiments, the hyperkinetic movement disorder is chorea associated with Huntington's disease. In certain embodiments, the hyperkinetic movement disorder is ataxia, chorea, dystonia, Huntington's disease, myoclonus, restless leg syndrome, or tremors.
  • In certain embodiments, the VMAT2 inhibitor is administered orally.
  • In certain embodiments, the VMAT2 inhibitor is administered in the form of a tablet or capsule.
  • In certain embodiments, the VMAT2 inhibitor is administered with or without food.
  • In certain embodiments, the VMAT2 inhibitor is valbenazine or a pharmaceutically acceptable salt and/or isotopic variant thereof. In certain embodiments, the VMAT2 inhibitor is valbenazine or a pharmaceutically acceptable salt thereof. In certain embodiments, the VMAT2 inhibitor is a valbenazine tosylate salt. In certain embodiments, the VMAT2 inhibitor is a ditosylate salt of valbenazine.
  • In certain embodiments, the VMAT2 inhibitor is an isotopic variant that is L-Valine, (2R, 3R,11bR)-1,3,4,6,7,11b-hexahydro-9,10-di(methoxy-d3)-3-(2-methylpropyl)-2H-benzo[α]quinolizin-2-yl ester or a pharmaceutically acceptable salt thereof.
  • In certain embodiments, the VMAT2 inhibitor is administered in an amount equivalent to between about 20 mg and about 160 mg of valbenazine free base. In certain embodiments, the VMAT2 inhibitor is administered in an amount equivalent to about 20 mg of valbenazine free base. In certain embodiments, the VMAT2 inhibitor is administered in an amount equivalent to about 40 mg of valbenazine free base. In certain embodiments, the VMAT2 inhibitor is administered in an amount equivalent to about 60 mg of valbenazine free base. In certain embodiments, the VMAT2 inhibitor is administered in an amount equivalent to about 80 mg of valbenazine free base. In certain embodiments, the VMAT2 inhibitor is administered in an amount equivalent to about 120 mg of valbenazine free base. In certain embodiments, the VMAT2 inhibitor is administered in an amount equivalent to about 160 mg of valbenazine free base.
  • In certain embodiments, the VMAT2 inhibitor is administered for a first period of time in a first amount and then the amount is increased to a second amount. In certain embodiments, the first period of time is a week. In certain embodiments, the first amount is equivalent to about 40 mg of valbenazine free base. In certain embodiments, the second amount is equivalent to about 80 mg of valbenazine free base.
  • In certain embodiments, the VMAT2 inhibitor is (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof.
  • In certain embodiments, the VMAT2 inhibitor is (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt thereof.
  • In certain embodiments, the VMAT2 inhibitor is an isotopic variant that is (+)-α-3-isobutyl-9,10-di(methoxy-d3)-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol or a pharmaceutically acceptable salt thereof.
  • In certain embodiments, the VMAT2 inhibitor is administered in an amount sufficient to achieve a maximal blood plasma concentration (Cmax) of (+)-α-DHTBZ of between about 15 ng to about 60 ng per mL plasma and a minimal blood plasma concentration (Cmax) of (+)-α-DHTBZ of at least 15 ng per mL plasma over an 8 hour period.
  • In certain embodiments, the VMAT2 inhibitor is administered in an amount sufficient to achieve a maximal blood plasma concentration (Cmax) of (+)-α-DHTBZ of between about 15 ng to about 60 ng per mL plasma and a minimal blood plasma concentration (Cmin) of approximately between about at least 33% -50% of the Cmax over a 12 hour period.
  • In certain embodiments, the VMAT2 inhibitor is administered in an amount sufficient to achieve: (i) a therapeutic concentration range of about 15 ng to about 60 ng of (+)-α-DHTBZ per mL plasma; and (ii) a threshold concentration of at least 15 ng (+)-α-DHTBZ per mL plasma over a period of about 8 hours to about 24 hours.
  • In certain embodiments, a method for treating neurological or psychiatric diseases or disorders is provided herein that comprises administering to a subject a pharmaceutical composition comprising the VMAT2 inhibitor, in an amount sufficient to achieve a maximal blood plasma concentration (Cmax) of R,R,R-DHTBZ of between about 15 ng to about 60 ng per mL plasma and a minimal blood plasma concentration (Cmin) of R,R,R-DHTBZ of at least 15 ng per mL plasma over an 8 hour period.
  • In certain embodiments, the Cmax of R,R,R-DHTBZ is about 15 ng/mL, about 20 ng/mL, about 25 ng/mL, about 30 ng/mL, about 35 ng/mL, about 40 ng/mL, about 45 ng/mL, about 50 ng/mL, about 55 ng/mL or about 60 ng/mL plasma. In certain embodiments, the Cmin of R,R,R-DHTBZ is at least 15 ng/mL, at least 20 ng/mL, at least 25 ng/mL, at least 30 ng/mL, or at least 35 ng/mL plasma, over a period of 8 hrs, 12 hrs, 16 hrs, 20 hrs, 24 hrs, 28 hrs, or 32 hrs. In certain embodiments, the Cmin of R,R,R-DHTBZ is between about 15 ng/mL to about 35 ng/mL.
  • In certain embodiments, the pharmaceutical composition is administered in an amount sufficient to provide a Cmax of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a Cmin of approximately at least 33% of the Cmax over a 24 hour period. In certain embodiments, the pharmaceutical composition is administered in an amount sufficient to provide a Cmax of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a Cmin of approximately at least 50% of the Cmax over a 24 hour period. In certain embodiments, the pharmaceutical composition is administered in an amount sufficient to provide a Cmax of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a Cmin of approximately between about at least 33% -50% of the Cmax over a 24 hour period.
  • In certain embodiments, the pharmaceutical composition is administered in an amount sufficient to provide a Cmax of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a Cmin of approximately at least 33% of the Cmax over a 12 hour period. In certain embodiments, the pharmaceutical composition is administered in an amount sufficient to provide a Cmax of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a Cmin of approximately at least 50% of the Cmax over a 12 hour period. In certain embodiments, the pharmaceutical composition is administered in an amount sufficient to provide a Cmax of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a Cmin of approximately between about at least 33% -50% of the Cmax over a 12 hour period.
  • In certain embodiments, the pharmaceutical composition is administered to a subject in an amount that provides a Cmax of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a Cmin of between about 5 ng/mL to about 30 ng/mL plasma over a 24 hour period. In certain embodiments, the pharmaceutical composition is administered to a subject in an amount that provides a Cmax of R,R,R-DHTBZ of about 15 ng/mL to about 60 ng/mL plasma and a Cmin of between about 7.5 ng/mL to about 30 ng/mL plasma over a 24 hour period.
  • In certain embodiments, a method for treating neurological or psychiatric diseases or disorders is provided herein that comprises administering to a subject a pharmaceutical composition comprising the VMAT2 inhibitor, as an active pharmaceutical ingredient, in an amount sufficient to provide: (i) a therapeutic concentration range of about 15 ng to about 60 ng of R,R,R-DHTBZ per mL plasma; and (ii) a threshold concentration of at least 15 ng R,R,R-DHTBZ per mL plasma over a period of about 8 hours to about 24 hours.
  • In certain embodiments, the therapeutic concentration range is about 15 ng to about 35 ng, to about 40 ng, to about 45 ng, to about 50 ng, or to about 55 ng R,R,R-DHTBZ per mL plasma.
  • In certain embodiments, the threshold concentration of R,R,R-DHTBZ is about 15 ng/mL, about 20 ng/mL, about 25 ng/mL, about 30 ng/mL, about 35 ng/mL, about 40 ng/mL, about 45 ng/mL, about 50 ng/mL, about 55 ng/mL or about 60 ng/mL plasma, over a period of about 8 hrs, about 12 hrs, about 16 hrs, about 20 hrs, about 24 hrs, about 28 hrs, or about 32 hrs. In certain embodiments, the threshold concentration of R,R,R-DHTBZ is between about 15 ng/mL to about 35 ng/mL over a period of about 8 hours to about 24 hours.
  • Plasma concentrations may be measured by methods known in the art and generally by tandem mass spectroscopy.
  • In certain embodiments, the therapeutically effective amount of the VMAT2 inhibitor is 10-90% less than the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor.
  • In certain embodiments, the therapeutically effective amount of the VMAT2 inhibitor is 20-80% less than the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor.
  • In certain embodiments, the therapeutically effective amount of the VMAT2 inhibitor is 30-70% less than the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor.
  • In certain embodiments, the therapeutically effective amount of the VMAT2 inhibitor is 40-60% less than the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor.
  • In certain embodiments, the therapeutically effective amount of the VMAT2 inhibitor is about 50% less than the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor.
  • For example, where the dosage administered to a patient who is not also being administered a strong CYP3A4 inhibitor is 40 mg per day, an individual may receive a reduced dosage of 4-36 mg per day, e.g., 8-32 mg per day, such as 12-28 mg per day, for example, 16-24 mg per day, or in certain embodiments, about 20 mg per day. Likewise, the dosage administered to a patient who is not also being administered a strong CYP3A4 inhibitor is 80 mg per day, an individual may receive a reduced dosage of 8-72 mg per day, e.g., 16-64 mg per day, such as 24-56 mg per day, for example, 32-48 mg per day, or in certain embodiments, about 24 mg per day.
  • In certain embodiments, the dose of VMAT2 inhibitor administered to a patient is reduced to, for example, 75% or less, 50% or less, or 25% or less of the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor. For example, where the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor is 40 mg per day, an individual may receive a reduced dosage of 30, 20, or 10 mg per day. Likewise, where the amount that would be administered to a patient who is not also being administered a strong CYP3A4 inhibitor is 80 mg per day, an individual may receive a reduced dosage of 60, 40, or 20 mg per day.
  • Also provided is a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof wherein the patient has previously been determined to have been administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering the VMAT2 inhibitor in an amount equivalent to about 40 mg of valbenazine free base once daily to the patient.
  • Also provided is a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof, said method comprising: determining whether the patient has been administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor; selecting the patient for treatment where the patient has been administered a strong CYP3A4 inhibitor; and administering the VMAT2 inhibitor to the selected patient in an amount equivalent to about 40 mg of valbenazine free base once daily.
  • Also provided is a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof, comprising: administering to the patient a therapeutically effective amount of the VMAT2 inhibitor, subsequently determining that the patient is to begin treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor, selecting the patient for treatment where the patient is to begin treatment with a strong CYP3A4 inhibitor, and administering the VMAT2 inhibitor to the selected patient in an amount equivalent to about 40 mg of valbenazine free base once daily.
  • Also provided is a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof wherein the patient has previously been determined to have been administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering the VMAT2 inhibitor to the patient in an amount that is less than the amount that would be administered to a patient who is not being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • Also provided is a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof, said method comprising: determining whether the patient has been administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor; selecting the patient for treatment where the patient has been administered a strong CYP3A4 inhibitor; and administering the VMAT2 inhibitor to the selected patient in an amount that is less than the amount that would be administered to a patient who is not being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • Also provided is a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof, comprising: administering to the patient a therapeutically effective amount of the VMAT2 inhibitor, subsequently determining that the patient is to begin treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor, selecting the patient for treatment where the patient is to begin treatment with a strong CYP3A4 inhibitor, and administering the VMAT2 inhibitor to the selected patient in an amount that is less than the amount that would be administered to a patient who is not being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • Also provided is a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof, wherein the patient has previously been determined to have been administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering to the patient a therapeutically effective amount of the VMAT2 inhibitor, subsequently selecting the patient that is not able to tolerate one or more exposure-related adverse reactions, and administering a reduced amount of VMAT2 inhibitor, such as 40 mg once daily, to the patient.
  • Also provided is a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof, wherein the patient has previously been determined to have been administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering to the patient a therapeutically effective amount of the VMAT2 inhibitor, subsequently selecting the patient that is able to tolerate one or more exposure-related adverse reactions, and continuing administering the therapeutically effective amount of the VMAT2 inhibitor to the patient.
  • Also provided is a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof wherein the patient has previously been determined to have been administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering the VMAT2 inhibitor in an amount sufficient to produce a mean valbenazine Cmax that is about 1 to about 2 fold higher than the mean valbenazine Cmax for a patient who is not being administered a strong CYP3A4 inhibitor and/or a mean valbenazine AUC0-∞ that is about 1.5 to about 2.5 fold higher than the mean valbenazine AUC0-∞ for a patient who is not being administered a strong CYP3A4 inhibitor.
  • Also provided is a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, for use in a method of treating a neurological or psychiatric disease or disorder in a patient in need thereof wherein the patient has previously been determined to have been administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: administering the VMAT2 inhibitor in an amount sufficient to produce a mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol Cmax that is about 1 to about 2 fold higher than the mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol Cmax for a patient who is not being administered a strong CYP3A4 inhibitor and/or a mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol AUC0-∞ that is about 1.5 to about 2.5 fold higher than the mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol AUC0-∞ for a patient who is not being administered a strong CYP3A4 inhibitor.
  • Valbenazine can be prepared according to U.S. Pat. Nos. 8,039,627 and 8,357,697, the disclosure of each of which is incorporated herein by reference in its entirety. Tetrabenazine may be administered by a variety of methods including the formulations disclosed in PCT Publications WO 2010/018408, WO 2011/019956, and WO 2014/047167, the disclosure of each of which is incorporated herein by reference in its entirety. In certain embodiments, the valbenazine for use in the compositions and methods provided herein is in polymorphic Form I as disclosed in U.S. Ser. No. 15/338,214, the disclosure of which is incorporated herein by reference in its entirety.
  • Pharmaceutical Compositions
  • Also provided is a composition for treating a patient in need of a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising the VMAT2 inhibitor, characterized in that the composition is administered in an amount equivalent to about 40 mg of valbenazine free base of the VMAT2 inhibitor once daily to the patient.
  • Also provided is a composition for treating a patient in need of a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising the VMAT2 inhibitor, characterized in that the therapeutically effective amount of the VMAT2 inhibitor is less than the amount that would be administered to a patient who is not being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • Also provided is a composition for treating a patient in need of a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, comprising the VMAT2 inhibitor, characterized in that the composition comprising the VMAT2 inhibitor in an amount equivalent to about 40 mg of valbenazine free base is administered once daily to the patient subsequently determined to begin treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor following administration of the composition comprising a therapeutically effective amount of the VMAT2 inhibitor.
  • Also provided is a composition for treating a patient in need of a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, comprising the VMAT2 inhibitor, characterized in that the composition comprising the VMAT2 inhibitor in an amount that would be less than that administered to a patient who has not begun treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor is administered to the patient subsequently determined to have begun treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor following administration of the composition comprising a therapeutically effective amount of the VMAT2 inhibitor.
  • Also provided is a composition for treating a patient in need of a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, and being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising:, comprising a therapeutically effective amount of the VMAT2 inhibitor, wherein administration of the composition produces a mean valbenazine Cmax that is about 1 to about 2 fold higher than the mean valbenazine Cmax for a patient who is not being administered a strong CYP3A4 inhibitor and/or a mean valbenazine AUC0-∞ that is about 1.5 to about 2.5 fold higher than the mean valbenazine AUC0-∞ for a patient who is not being administered a strong CYP3A4 inhibitor.
  • Also provided is a composition for treating a patient in need of a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, and being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising:, comprising a therapeutically effective amount of the VMAT2 inhibitor, wherein administration of the composition produces a mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol Cmax that is about 1 to about 2 fold higher than the mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol Cmax for a patient who is not being administered a strong CYP3A4 inhibitor and/or a mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol AUC0-∞ that is about 1.5 to about 2.5 fold higher than the mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol AUC0-∞ for a patient who is not being administered a strong CYP3A4 inhibitor.
  • In certain embodiments, the patient or a medical care worker is informed that administration of the VMAT2 inhibitor to patients who is also being administered a strong CYP3A4 inhibitor in higher exposure of valbenazine and/or (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol than administration of the composition to a patient who is not being administered a strong CYP3A4 inhibitor.
  • In certain embodiments, the patient or a medical care worker is informed that administration of the VMAT2 inhibitor to a patient who is also being administered a strong CYP3A4 inhibitor may result in increased risk of one or more exposure-related adverse reactions than administration of the composition to a patient who is not being administered a strong CYP3A4 inhibitor.
  • In certain embodiments, the one or more exposure-related adverse reactions is chosen from somnolence, anticholinergic effects, balance disorders or falls, headache, akathisia, vomiting, nausea, arthralgia, QT prolongation, increase in blood glucose, increase in weight, respiratory infections, drooling, dyskinesia, extrapyramidal symptoms (non-akathisia), anxiety, insomnia, increase in prolactin, increase in alkaline phosphatase, and increase in bilirubin.
  • In certain embodiments, the one or more exposure-related adverse reactions is chosen from somnolence, anticholinergic effects, balance disorders or falls, headache, akathisia, vomiting, nausea, arthralgia, and QT prolongation.
  • In certain embodiments, the one or more exposure-related adverse reactions is chosen from somnolence and QT prolongation.
  • In certain embodiments, the patient or a medical care worker is informed that co-administration of the composition and the CYP3A4 inhibitor may prolong the patient's QT interval.
  • In certain embodiments, the patient or a medical care worker is informed that administration of the composition to a patient who is also being administered a strong CYP3A4 inhibitor may prolong the patient's QT interval than administration of the composition to a patient who is also being administered a strong CYP3A4 inhibitor.
  • In certain embodiments, the strong CYP3A4 inhibitor is chosen from clarithromycin, chloramphenicol, cobicistat, indinavir, itraconazole, ketoconazole, nefazodone, nelfinavir, ritonavir, saquinavir, and telithromycin. In certain embodiments, the strong CYP3A4 inhibitor is chosen from clarithromycin, itraconazole, and ketoconazole. In certain embodiments, the strong CYP3A4 inhibitor is ketoconazole.
  • In certain embodiments, the composition is for treating a neurological or psychiatric disease or disorder.
  • In certain embodiments, the composition is administered orally.
  • In certain embodiments, the composition is administered in the form of a tablet or capsule.
  • In certain embodiments, the composition is administered with or without food.
  • In certain embodiments, the VMAT2 inhibitor is valbenazine or a pharmaceutically acceptable salt and/or isotopic variant thereof.
  • In certain embodiments, the VMAT2 inhibitor is valbenazine or a pharmaceutically acceptable salt thereof.
  • In certain embodiments, the VMAT2 inhibitor is a valbenazine tosylate salt.
  • In certain embodiments, the VMAT2 inhibitor is a ditosylate salt of valbenazine.
  • In certain embodiments, the VMAT2 inhibitor is an isotopic variant that is L-Valine, (2R, 3R,11 bR)-1,3,4,6,7,11b-hexahydro-9,10-di(methoxy-d3)-3-(2-methylpropyl)-2H-benzo[α]quinolizin-2-yl ester or a pharmaceutically acceptable salt thereof.
  • In certain embodiments, the VMAT2 inhibitor is (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof.
  • In certain embodiments, the VMAT2 inhibitor is (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt thereof.
  • In certain embodiments, the VMAT2 inhibitor is an isotopic variant that is (+)-α-3-isobutyl-9,10-di(methoxy-d3)-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol or a pharmaceutically acceptable salt thereof.
  • In certain embodiments, the composition is administered in an amount equivalent to between about 20 mg and about 160 mg of valbenazine free base. In certain embodiments, the composition is administered in an amount equivalent to about 20 mg of valbenazine free base. In certain embodiments, the composition is administered in an amount equivalent to about 40 mg of valbenazine free base. In certain embodiments, the composition is administered in an amount equivalent to about 60 mg of valbenazine free base. In certain embodiments, the composition is administered in an amount equivalent to about 80 mg of valbenazine free base. In certain embodiments, the composition is administered in an amount equivalent to about 120 mg of valbenazine free base.
  • In certain embodiments, the composition is administered for a first period of time in a first amount and then the amount is increased to a second amount. In certain embodiments, the first period of time is a week. In certain embodiments, the first amount is equivalent to about 40 mg of valbenazine free base. In certain embodiments, the the second amount is equivalent to about 80 mg of valbenazine free base.
  • In certain embodiments, the composition is administered in an amount sufficient to achieve a maximal blood plasma concentration (Cmax) of (+)-α-DHTBZ of between about 15 ng to about 60 ng per mL plasma and a minimal blood plasma concentration (Cmin) of (+)-α-DHTBZ of at least 15 ng per mL plasma over an 8 hour period.
  • In certain embodiments, the composition is administered in an amount sufficient to achieve a maximal blood plasma concentration (Cmax) of (+)-α-DHTBZ of between about 15 ng to about 60 ng per mL plasma and a minimal blood plasma concentration (Cmin) of approximately between about at least 33% -50% of the Cmax over a 12 hour period.
  • In certain embodiments, the composition is administered in an amount sufficient to achieve: (i) a therapeutic concentration range of about 15 ng to about 60 ng of (+)-α-DHTBZ per mL plasma; and (ii) a threshold concentration of at least 15 ng (+)-α-DHTBZ per mL plasma over a period of about 8 hours to about 24 hours.
  • In certain embodiments, the therapeutically effective amount of the VMAT2 inhibitor is 10-90% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • In certain embodiments, the therapeutically effective amount of the VMAT2 inhibitor is 20-80% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • In certain embodiments, the therapeutically effective amount of the VMAT2 inhibitor is 30-70% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • In certain embodiments, the therapeutically effective amount of the VMAT2 inhibitor is 40-60% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • In certain embodiments, the therapeutically effective amount of the VMAT2 inhibitor is about 50% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
  • Also provided herein is a pharmaceutical composition for use in treating neurological or psychiatric diseases or disorders, comprising the VMAT2 inhibitor as an active pharmaceutical ingredient, in combination with one or more pharmaceutically acceptable carriers or excipients.
  • The choice of excipient, to a large extent, depends on factors, such as the particular mode of administration, the effect of the excipient on the solubility and stability of the active ingredient, and the nature of the dosage form.
  • The pharmaceutical compositions provided herein may be provided in unit dosage forms or multiple-dosage forms. Unit-dosage forms, as used herein, refer to physically discrete units suitable for administration to human and animal subjects and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of the active ingredient(s) sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carriers or excipients. Examples of unit-dosage forms include ampoules, syringes, and individually packaged tablets and capsules. Unit dosage forms may be administered in fractions or multiples thereof. A multiple-dosage form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dosage form. Examples of multiple-dosage forms include vials, bottles of tablets or capsules, or bottles of pints or gallons.
  • The pharmaceutical compositions provided herein may be administered alone, or in combination with one or more other compounds provided herein, one or more other active ingredients. The pharmaceutical compositions provided herein may be formulated in various dosage forms for oral, parenteral, and topical administration. The pharmaceutical compositions may also be formulated as a modified release dosage form, including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms. These dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art). The pharmaceutical compositions provided herein may be administered at once, or multiple times at intervals of time. It is understood that the precise dosage and duration of treatment may vary with the age, weight, and condition of the patient being treated, and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test or diagnostic data. It is further understood that for any particular individual, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the formulations.
  • Oral Administration
  • The pharmaceutical compositions provided herein may be provided in solid, semisolid, or liquid dosage forms for oral administration. As used herein, oral administration also includes buccal, lingual, and sublingual administration. Suitable oral dosage forms include, but are not limited to, tablets, capsules, pills, troches, lozenges, pastilles, cachets, pellets, medicated chewing gum, granules, bulk powders, effervescent or non-effervescent powders or granules, solutions, emulsions, suspensions, solutions, wafers, sprinkles, elixirs, and syrups. In addition to the active ingredient(s), the pharmaceutical compositions may contain one or more pharmaceutically acceptable carriers or excipients, including, but not limited to, binders, fillers, diluents, disintegrants, wetting agents, lubricants, glidants, coloring agents, dye-migration inhibitors, sweetening agents, and flavoring agents.
  • Binders or granulators impart cohesiveness to a tablet to ensure the tablet remaining intact after compression. Suitable binders or granulators include, but are not limited to, starches, such as corn starch, potato starch, and pre-gelatinized starch (e.g., STARCH 1500); gelatin; sugars, such as sucrose, glucose, dextrose, molasses, and lactose; natural and synthetic gums, such as acacia, alginic acid, alginates, extract of Irish moss, Panwar gum, ghatti gum, mucilage of isabgol husks, carboxymethylcellulose, methylcellulose, polyvinylpyrrolidone (PVP), Veegum, larch arabogalactan, powdered tragacanth, and guar gum; celluloses, such as ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose, methyl cellulose, hydroxyethylcellulose (HEC), hydroxypropylcellulose (HPC), hydroxypropyl methyl cellulose (HPMC); microcrystalline celluloses, such as AVICEL-PH-101, AVICEL-PH-103, AVICEL RC-581, AVICEL-PH-105 (FMC Corp., Marcus Hook, Pa.); and mixtures thereof. Suitable fillers include, but are not limited to, talc, calcium carbonate, microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pregelatinized starch, and mixtures thereof. The binder or filler may be present from about 50 to about 99% by weight in the pharmaceutical compositions provided herein.
  • Suitable diluents include, but are not limited to, dicalcium phosphate, calcium sulfate, lactose, sorbitol, sucrose, inositol, cellulose, kaolin, mannitol, sodium chloride, dry starch, and powdered sugar. Certain diluents, such as mannitol, lactose, sorbitol, sucrose, and inositol, when present in sufficient quantity, can impart properties to some compressed tablets that permit disintegration in the mouth by chewing. Such compressed tablets can be used as chewable tablets.
  • Suitable disintegrants include, but are not limited to, agar; bentonite; celluloses, such as methylcellulose and carboxymethylcellulose; wood products; natural sponge; cation-exchange resins; alginic acid; gums, such as guar gum and Vee gum HV; citrus pulp; cross-linked celluloses, such as croscarmellose; cross-linked polymers, such as crospovidone; cross-linked starches; calcium carbonate; microcrystalline cellulose, such as sodium starch glycolate; polacrilin potassium; starches, such as com starch, potato starch, tapioca starch, and pre-gelatinized starch; clays; aligns; and mixtures thereof. The amount of disintegrant in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art. The pharmaceutical compositions provided herein may contain from about 0.5 to about 15% or from about 1 to about 5% by weight of a disintegrant.
  • Suitable lubricants include, but are not limited to, calcium stearate; magnesium stearate; mineral oil; light mineral oil; glycerin; sorbitol; mannitol; glycols, such as glycerol behenate and polyethylene glycol (PEG); stearic acid; sodium lauryl sulfate; talc; hydrogenated vegetable oil, including peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, com oil, and soybean oil; zinc stearate; ethyl oleate; ethyl laureate; agar; starch; lycopodium; silica or silica gels, such as AEROSIL®200 (W.R. Grace Co., Baltimore, Md.) and CAB-0-SIL® (Cabot Co. of Boston, Mass.); and mixtures thereof. The pharmaceutical compositions provided herein may contain about 0.1 to about 5% by weight of a lubricant. Suitable glidants include colloidal silicon dioxide, CAB-0-SIL® (Cabot Co. of Boston, Mass.), and asbestos-free talc. Coloring agents include any of the approved, certified, water soluble FD&C dyes, and water insoluble FD&C dyes suspended on alumina hydrate, and color lakes and mixtures thereof. A color lake is the combination by adsorption of a water-soluble dye to a hydrous oxide of a heavy metal, resulting in an insoluble form of the dye. Flavoring agents include natural flavors extracted from plants, such as fruits, and synthetic blends of compounds which produce a pleasant taste sensation, such as peppermint and methyl salicylate. Sweetening agents include sucrose, lactose, mannitol, syrups, glycerin, and artificial sweeteners, such as saccharin and aspartame. Suitable emulsifying agents include gelatin, acacia, tragacanth, bentonite, and surfactants, such as polyoxyethylene sorbitan monooleate (TWEEN® 20), polyoxyethylene sorbitan monooleate 80 (TWEEN® 80), and triethanolamine oleate. Suspending and dispersing agents include sodium carboxymethylcellulose, pectin, tragacanth, Veegum, acacia, sodium carbomethylcellulose, hydroxypropyl methylcellulose, and polyvinylpyrolidone. Preservatives include glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol. Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate, and polyoxyethylene lauryl ether. Solvents include glycerin, sorbitol, ethyl alcohol, and syrup. Examples of non-aqueous liquids utilized in emulsions include mineral oil and cottonseed oil. Organic acids include citric and tartaric acid. Sources of carbon dioxide include sodium bicarbonate and sodium carbonate.
  • It should be understood that many carriers and excipients may serve several functions, even within the same formulation. The pharmaceutical compositions provided herein may be provided as compressed tablets, tablet triturates, chewable lozenges, rapidly dissolving tablets, multiple compressed tablets, or enteric-coating tablets, sugar-coated, or film-coated tablets. Enteric coated tablets are compressed tablets coated with substances that resist the action of stomach acid but dissolve or disintegrate in the intestine, thus protecting the active ingredients from the acidic environment of the stomach. Enteric-coatings include, but are not limited to, fatty acids, fats, phenylsalicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalates. Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which may be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation. Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material. Film coatings include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coating imparts the same general characteristics as sugar coating. Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets.
  • The tablet dosage forms may be prepared from the active ingredient in powdered, crystalline, or granular forms, alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, controlled-release polymers, lubricants, diluents, and/or colorants. Flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges.
  • The pharmaceutical compositions provided herein may be provided as soft or hard capsules, which can be made from gelatin, methylcellulose, starch, or calcium alginate. The hard gelatin capsule, also known as the dry-filled capsule (DFC), consists of two sections, one slipping over the other, thus completely enclosing the active ingredient. The soft elastic capsule (SEC) is a soft, globular shell, such as a gelatin shell, which is plasticized by the addition of glycerin, sorbitol, or a similar polyol. The soft gelatin shells may contain a preservative to prevent the growth of microorganisms. Suitable preservatives are those as described herein, including methyl- and propyl-parabens, and sorbic acid. The liquid, semisolid, and solid dosage forms provided herein may be encapsulated in a capsule. Suitable liquid and semisolid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils, or triglycerides. The capsules may also be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient.
  • The pharmaceutical compositions provided herein may be provided in liquid and semisolid dosage forms, including emulsions, solutions, suspensions, elixirs, and syrups. An emulsion is a two-phase system, in which one liquid is dispersed in the form of small globules throughout another liquid, which can be oil-in-water or water-in-oil. Emulsions may include a pharmaceutically acceptable non-aqueous liquids or solvent, emulsifying agent, and preservative. Suspensions may include a pharmaceutically acceptable suspending agent and preservative. Aqueous alcoholic solutions may include a pharmaceutically acceptable acetal, such as a di(lower alkyl) acetal of a lower alkyl aldehyde (the term “lower” means an alkyl having between 1 and 6 carbon atoms), e.g., acetaldehyde diethyl acetal; and a water-miscible solvent having one or more hydroxyl groups, such as propylene glycol and ethanol. Elixirs are clear, sweetened, and hydroalcoholic solutions. Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may also contain a preservative. For a liquid dosage form, for example, a solution in a polyethylene glycol may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be measured conveniently for administration.
  • Other useful liquid and semisolid dosage forms include, but are not limited to, those containing the active ingredient(s) provided herein, and a dialkylated mono- or polyalkylene glycol, including, 1,2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether, wherein 350, 550, and 750 refer to the approximate average molecular weight of the polyethylene glycol. These formulations may further comprise one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.
  • The pharmaceutical compositions provided herein for oral administration may be also provided in the forms of liposomes, micelles, microspheres, or nanosystems.
  • The pharmaceutical compositions provided herein may be provided as noneffervescent or effervescent, granules and powders, to be reconstituted into a liquid dosage form. Pharmaceutically acceptable carriers and excipients used in the non-effervescent granules or powders may include diluents, sweeteners, and wetting agents. Pharmaceutically acceptable carriers and excipients used in the effervescent granules or powders may include organic acids and a source of carbon dioxide. Coloring and flavoring agents can be used in all of the above dosage forms. The pharmaceutical compositions provided herein may be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms.
  • The pharmaceutical compositions provided herein may be co-formulated with other active ingredients which do not impair the desired therapeutic action, or with substances that supplement the desired action, such as antacids, proton pump inhibitors, and Hz-receptor antagonists.
  • The pharmaceutical compositions provided herein may be administered parenterally by injection, infusion, or implantation, for local or systemic administration. Parenteral administration, as used herein, include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial, and subcutaneous administration.
  • Parenteral Administration
  • The pharmaceutical compositions provided herein may be formulated in any dosage forms that are suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems, and solid forms suitable for solutions or suspensions in liquid prior to injection. Such dosage forms can be prepared according to conventional methods known to those skilled in the art of pharmaceutical science.
  • The pharmaceutical compositions intended for parenteral administration may include one or more pharmaceutically acceptable carriers and excipients, including, but not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, cryoprotectants, lyoprotectants, thickening agents, pH adjusting agents, and inert gases.
  • Suitable aqueous vehicles include, but are not limited to, water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringers injection, isotonic dextrose injection, sterile water injection, dextrose and lactated Ringers injection. Non-aqueous vehicles include, but are not limited to, fixed oils of vegetable origin, castor oil, com oil, cottonseed oil, olive oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oils, hydrogenated soybean oil, and medium-chain triglycerides of coconut oil, and palm seed oil. Water-miscible vehicles include, but are not limited to, ethanol, 1,3-butanediol, liquid polyethylene glycol (e.g., polyethylene glycol 300 and polyethylene glycol 400), propylene glycol, glycerin, N-methyl-2-pyrrolidone, dimethylacetamide, and dimethylsulfoxide.
  • Suitable antimicrobial agents or preservatives include, but are not limited to, phenols, cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl phydroxybenzates, thimerosal, benzalkonium chloride, benzethonium chloride, methyl- and propylparabens, and sorbic acid. Suitable isotonic agents include, but are not limited to, sodium chloride, glycerin, and dextrose. Suitable buffering agents include, but are not limited to, phosphate and citrate. Suitable antioxidants are those as described herein, including bisulfite and sodium metabisulfite. Suitable local anesthetics include, but are not limited to, procaine hydrochloride. Suitable suspending and dispersing agents are those as described herein, including sodium carboxymethylcelluose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone. Suitable emulsifying agents include those described herein, including polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate 80, and triethanolamine oleate. Suitable sequestering or chelating agents include, but are not limited to EDTA. Suitable pH adjusting agents include, but are not limited to, sodium hydroxide, hydrochloric acid, citric acid, and lactic acid. Suitable complexing agents include, but are not limited to, cyclodextrins, including alpha-cyclodextrin, beta-cyclodextrin, hydroxypropyl-beta-cyclodextrin, sulfobutylether-beta-cyclodextrin, and sulfobutylether 7-beta-cyclodextrin (CAPTISOL®, CyDex, Lenexa, Kans.).
  • The pharmaceutical compositions provided herein may be formulated for single or multiple dosage administration. The single dosage formulations are packaged in an ampule, a vial, or a syringe. The multiple dosage parenteral formulations must contain an antimicrobial agent at bacteriostatic or fungistatic concentrations. All parenteral formulations must be sterile, as known and practiced in the art.
  • In certain embodiments, the pharmaceutical compositions are provided as ready-to-use sterile solutions. In certain embodiments, the pharmaceutical compositions are provided as sterile dry soluble products, including lyophilized powders and hypodermic tablets, to be reconstituted with a vehicle prior to use. In certain embodiments, the pharmaceutical compositions are provided as ready-to-use sterile suspensions. In certain embodiments, the pharmaceutical compositions are provided as sterile dry insoluble products to be reconstituted with a vehicle prior to use. In certain embodiments, the pharmaceutical compositions are provided as ready-to-use sterile emulsions.
  • The pharmaceutical compositions provided herein may be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms.
  • The pharmaceutical compositions may be formulated as a suspension, solid, semi-solid, or thixotropic liquid, for administration as an implanted depot. In certain embodiments, the pharmaceutical compositions provided herein are dispersed in a solid inner matrix, which is surrounded by an outer polymeric membrane that is insoluble in body fluids but allows the active ingredient in the pharmaceutical compositions diffuse through.
  • Suitable inner matrixes include polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinylacetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers, such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol, and cross-linked partially hydrolyzed polyvinyl acetate.
  • Suitable outer polymeric membranes include polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinylacetate copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinylchloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer.
  • Topical Administration
  • The pharmaceutical compositions provided herein may be administered topically to the skin, orifices, or mucosa. The topical administration, as used herein, include (intra)dermal, conjuctival, intracorneal, intraocular, ophthalmic, auricular, transdermal, nasal, vaginal, uretheral, respiratory, and rectal administration.
  • The pharmaceutical compositions provided herein may be formulated in any dosage forms that are suitable for topical administration for local or systemic effect, including emulsions, solutions, suspensions, creams, gels, hydrogels, ointments, dusting powders, dressings, elixirs, lotions, suspensions, tinctures, pastes, foams, films, aerosols, irrigations, sprays, suppositories, bandages, dermal patches. The topical formulation of the pharmaceutical compositions provided herein may also comprise liposomes, micelles, microspheres, nanosystems, and mixtures thereof.
  • Pharmaceutically acceptable carriers and excipients suitable for use in the topical formulations provided herein include, but are not limited to, aqueous vehicles, water miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, penetration enhancers, cryopretectants, lyoprotectants, thickening agents, and inert gases.
  • The pharmaceutical compositions may also be administered topically by electroporation, iontophoresis, phonophoresis, sonophoresis and microneedle or needle-free injection, such as POWDERJECT™ (Chiron Corp., Emeryville, Calif.), and BIOJECT™ (Bioject Medical Technologies Inc., Tualatin, Oreg.).
  • The pharmaceutical compositions provided herein may be provided in the forms of ointments, creams, and gels. Suitable ointment vehicles include oleaginous or hydrocarbon bases, including such as lard, benzoinated lard, olive oil, cottonseed oil, and other oils, white petrolatum; emulsifiable or absorption bases, such as hydrophilic petrolatum, hydroxystearin sulfate, and anhydrous lanolin; water-removable bases, such as hydrophilic ointment; water-soluble ointment bases, including polyethylene glycols of varying molecular weight; emulsion bases, either water-in-oil (W/O) emulsions or oil-in-water (O/W) emulsions, including cetyl alcohol, glyceryl monostearate, lanolin, and stearic acid. These vehicles are emollient but generally require addition of antioxidants and preservatives.
  • Suitable cream base can be oil-in-water or water-in-oil. Cream vehicles may be water-washable, and contain an oil phase, an emulsifier, and an aqueous phase. The oil phase is also called the “internal” phase, which is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol. The aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant. The emulsifier in a cream formulation may be a nonionic, anionic, cationic, or amphoteric surfactant.
  • Gels are semisolid, suspension-type systems. Single-phase gels contain organic macromolecules distributed substantially uniformly throughout the liquid carrier. Suitable gelling agents include crosslinked acrylic acid polymers, such as carbomers, carboxypolyalkylenes, Carbopol®; hydrophilic polymers, such as polyethylene oxides, polyoxyethylene-polyoxypropylene copolymers, and polyvinylalcohol; cellulosic polymers, such as hydroxypropyl cellulose, hydroxyethyl cellulose, hydroxypropyl methylcellulose, hydroxypropyl methylcellulose phthalate, and methylcellulose; gums, such as tragacanth and xanthan gum; sodium alginate; and gelatin. In order to prepare a uniform gel, dispersing agents such as alcohol or glycerin can be added, or the gelling agent can be dispersed by trituration, mechanical mixing, and/or stirring.
  • The pharmaceutical compositions provided herein may be administered rectally, urethrally, vaginally, or perivaginally in the forms of suppositories, pessaries, bougies, poultices or cataplasm, pastes, powders, dressings, creams, plasters, contraceptives, ointments, solutions, emulsions, suspensions, tampons, gels, foams, sprays, or enemas. These dosage forms can be manufactured using conventional processes.
  • Rectal, urethral, and vaginal suppositories are solid bodies for insertion into body orifices, which are solid at ordinary temperatures but melt or soften at body temperature to release the active ingredient(s) inside the orifices. Pharmaceutically acceptable carriers utilized in rectal and vaginal suppositories include vehicles, such as stiffening agents, which produce a melting point in the proximity of body temperature, when formulated with the pharmaceutical compositions provided herein; and antioxidants as described herein, including bisulfite and sodium metabisulfite. Suitable vehicles include, but are not limited to, cocoa butter (theobroma oil), glycerin-gelatin, carbowax (polyoxyethylene glycol), spermaceti, paraffin, white and yellow wax, and appropriate mixtures of mono-, di- and triglycerides of fatty acids, hydrogels, such as polyvinyl alcohol, hydroxyethyl methacrylate, polyacrylic acid; glycerinated gelatin. Combinations of the various vehicles may be used. Rectal and vaginal suppositories may be prepared by the compressed method or molding. The typical weight of a rectal and vaginal suppository is about 2 to 3 g.
  • The pharmaceutical compositions provided herein may be administered ophthalmically in the forms of solutions, suspensions, ointments, emulsions, gel-forming solutions, powders for solutions, gels, ocular inserts, and implants.
  • The pharmaceutical compositions provided herein may be administered intranasally or by inhalation to the respiratory tract. The pharmaceutical compositions may be provided in the form of an aerosol or solution for delivery using a pressurized container, pump, spray, atomizer, such as an atomizer using electrohydrodynamics to produce a fine mist, or nebulizer, alone or in combination with a suitable propellant, such as 1,1,1,2-tetrafluoroethane or 1,1,1,2,3,3,3-heptafluoropropane. The pharmaceutical compositions may also be provided as a dry powder for insufflation, alone or in combination with an inert carrier such as lactose or phospholipids; and nasal drops. For intranasal use, the powder may comprise a bioadhesive agent, including chitosan or cyclodextrin.
  • Solutions or suspensions for use in a pressurized container, pump, spray, atomizer, or nebulizer may be formulated to contain ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilizing, or extending release of the active ingredient provided herein, a propellant as solvent; and/or a surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
  • The pharmaceutical compositions provided herein may be micronized to a size suitable for delivery by inhalation, such as 50 micrometers or less, or 10 micrometers or less. Particles of such sizes may be prepared using a comminuting method known to those skilled in the art, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenization, or spray drying.
  • Capsules, blisters and cartridges for use in an inhaler or insufflator may be formulated to contain a powder mix of the pharmaceutical compositions provided herein; a suitable powder base, such as lactose or starch; and a performance modifier, such as /-leucine, mannitol, or magnesium stearate. The lactose may be anhydrous or in the form of the monohydrate. Other suitable excipients include dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose, and trehalose. The pharmaceutical compositions provided herein for inhaled/intranasal administration may further comprise a suitable flavor, such as menthol and levomenthol, or sweeteners, such as saccharin or saccharin sodium.
  • The pharmaceutical compositions provided herein for topical administration may be formulated to be immediate release or modified release, including delayed-, sustained-, pulsed-, controlled-, targeted, and programmed release.
  • Modified Release
  • The pharmaceutical compositions provided herein may be formulated as a modified release dosage form. As used herein, the term “modified release” refers to a dosage form in which the rate or place of release of the active ingredient(s) is different from that of an immediate dosage form when administered by the same route. Modified release dosage forms include delayed-, extended-, prolonged-, sustained-, pulsatile- or pulsed-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms.
  • The pharmaceutical compositions in modified release dosage forms can be prepared using a variety of modified release devices and methods known to those skilled in the art, including, but not limited to, matrix controlled release devices, osmotic controlled release devices, multiparticulate controlled release devices, ion-exchange resins, enteric coatings, multilayered coatings, microspheres, liposomes, and combinations thereof. The release rate of the active ingredient(s) can also be modified by varying the particle sizes and polymorphorism of the active ingredient(s).
  • The pharmaceutical compositions provided herein in a modified release dosage form may be fabricated using a matrix controlled release device known to those skilled in the art.
  • In certain embodiments, the pharmaceutical compositions provided herein in a modified release dosage form is formulated using an erodible matrix device, which is water swellable, erodible, or soluble polymers, including synthetic polymers, and naturally occurring polymers and derivatives, such as polysaccharides and proteins.
  • Materials useful in forming an erodible matrix include, but are not limited to, chitin, chitosan, dextran, and pullulan; gum agar, gum arabic, gum karaya, locust bean gum, gum tragacanth, carrageenans, gum ghatti, guar gum, xanthan gum, and scleroglucan; starches, such as dextrin and maltodextrin; hydrophilic colloids, such as pectin; phosphatides, such as lecithin; alginates; propylene glycol alginate; gelatin; collagen; and cellulosics, such as ethyl cellulose (EC), methylethyl cellulose (MEC), carboxymethyl cellulose (CMC), CMEC, hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), cellulose acetate (CA), cellulose propionate (CP), cellulose butyrate (CB), cellulose acetate butyrate (CAB), CAP, CAT, hydroxypropyl methyl cellulose (HPMC), HPMCP, HPMCAS, hydroxypropyl methyl cellulose acetate trimellitate (HPMCAT), and ethylhydroxy ethylcellulose (EHEC); polyvinyl pyrrolidone; polyvinyl alcohol; polyvinyl acetate; glycerol fatty acid esters; polyacrylamide; polyacrylic acid; copolymers of ethacrylic acid or methacrylic acid (EUDRAGIT®, Rohm America, Inc., Piscataway, N.J.); poly(2-hydroxyethyl-methacrylate); polylactides; copolymers of L-glutamic acid and ethyl-L-glutamate; degradable lactic acidglycolic acid copolymers; poly-D-(−)-3-hydroxybutyric acid; and other acrylic acid derivatives, such as homopolymers and copolymers of butylmethacrylate, methylmethacrylate, ethylmethacrylate, ethylacrylate, (2-dimethylaminoethyl)methacrylate, and (trimethylaminoethyl)methacrylate chloride.
  • In certain embodiments, the pharmaceutical compositions are formulated with a non-erodible matrix device. The active ingredient(s) is dissolved or dispersed in an inert matrix and is released primarily by diffusion through the inert matrix once administered. Materials suitable for use as a non-erodible matrix device included, but are not limited to, insoluble plastics, such as polyethylene, polypropylene, polyisoprene, polyisobutylene, polybutadiene, polymethylmethacrylate, polybutylmethacrylate, chlorinated polyethylene, polyvinylchloride, methyl acrylate-methyl methacrylate copolymers, ethylene-vinylacetate copolymers, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, vinylchloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer, polyvinyl chloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, and; hydrophilic polymers, such as ethyl cellulose, cellulose acetate, crospovidone, and cross-linked partially hydrolyzed polyvinyl acetate,; and fatty compounds, such as camauba wax, microcrystalline wax, and triglycerides.
  • In a matrix controlled release system, the desired release kinetics can be controlled, for example, via the polymer type employed, the polymer viscosity, the particle sizes of the polymer and/or the active ingredient(s), the ratio of the active ingredient(s) versus the polymer, and other excipients in the compositions.
  • The pharmaceutical compositions provided herein in a modified release dosage form may be prepared by methods known to those skilled in the art, including direct compression, dry or wet granulation followed by compression, melt-granulation followed by compression.
  • The pharmaceutical compositions provided herein in a modified release dosage form may be fabricated using an osmotic controlled release device, including one-chamber system, two-chamber system, asymmetric membrane technology (AMT), and extruding core system (ECS). In general, such devices have at least two components: (a) the core which contains the active ingredient(s); and (b) a semipermeable membrane with at least one delivery port, which encapsulates the core. The semipermeable membrane controls the influx of water to the core from an aqueous environment of use so as to cause drug release by extrusion through the delivery port(s).
  • In addition to the active ingredient(s), the core of the osmotic device optionally includes an osmotic agent, which creates a driving force for transport of water from the environment of use into the core of the device. One class of osmotic agents waterswellable hydrophilic polymers, which are also referred to as “osmopolymers” and “hydrogels,” including, but not limited to, hydrophilic vinyl and acrylic polymers, polysaccharides such as calcium alginate, polyethylene oxide (PEO), polyethylene glycol (PEG), polypropylene glycol (PPG), poly(2-hydroxyethyl methacrylate), poly(acrylic) acid, poly(methacrylic) acid, polyvinylpyrrolidone (PVP), crosslinked PVP, polyvinyl alcohol (PVA), PVA/PVP copolymers, PVA/PVP copolymers with hydrophobic monomers such as methyl methacrylate and vinyl acetate, hydrophilic polyurethanes containing large PEO blocks, sodium croscarmellose, carrageenan, hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), carboxymethyl cellulose (CMC) and carboxyethyl, cellulose (CEC), sodium alginate, polycarbophil, gelatin, xanthan gum, and sodium starch glycolate.
  • The other class of osmotic agents is osmogens, which are capable of imbibing water to affect an osmotic pressure gradient across the barrier of the surrounding coating. Suitable osmogens include, but are not limited to, inorganic salts, such as magnesium sulfate, magnesium chloride, calcium chloride, sodium chloride, lithium chloride, potassium sulfate, potassium phosphates, sodium carbonate, sodium sulfite, lithium sulfate, potassium chloride, and sodium sulfate; sugars, such as dextrose, fructose, glucose, inositol, lactose, maltose, mannitol, raffinose, sorbitol, sucrose, trehalose, and xylitol,; organic acids, such as ascorbic acid, benzoic acid, fumaric acid, citric acid, maleic acid, sebacic acid, sorbic acid, adipic acid, edetic acid, glutamic acid, p-tolunesulfonic acid, succinic acid, and tartaric acid; urea; and mixtures thereof.
  • Osmotic agents of different dissolution rates may be employed to influence how rapidly the active ingredient(s) is initially delivered from the dosage form. For example, amorphous sugars, such as Mannogeme EZ (SPI Pharma, Lewes, Del.) can be used to provide faster delivery during the first couple of hours to promptly produce the desired therapeutic effect, and gradually and continually release of the remaining amount to maintain the desired level of therapeutic or prophylactic effect over an extended period of time. In this case, the active ingredient(s) is released at such a rate to replace the amount of the active ingredient metabolized and excreted.
  • The core may also include a wide variety of other excipients and carriers as described herein to enhance the performance of the dosage form or to promote stability or processing.
  • Materials useful in forming the semipermeable membrane include various grades of acrylics, vinyls, ethers, polyamides, polyesters, and cellulosic derivatives that are water-permeable and water-insoluble at physiologically relevant pHs, or are susceptible to being rendered water-insoluble by chemical alteration, such as crosslinking. Examples of suitable polymers useful in forming the coating, include plasticized, unplasticized, and reinforced cellulose acetate (CA), cellulose diacetate, cellulose triacetate, CA propionate, cellulose nitrate, cellulose acetate butyrate (CAB), CA ethyl carbamate, CAP, CA methyl carbamate, CA succinate, cellulose acetate trimellitate (CAT), CA dimethylaminoacetate, CAethyl carbonate, CA chloroacetate, CA ethyl oxalate, CA methyl sulfonate, CA butyl sulfonate, CA p-toluene sulfonate, agar acetate, amylose triacetate, beta glucan acetate, beta glucan triacetate, acetaldehyde dimethyl acetate, triacetate of locust bean gum, hydroxlated ethylene-vinylacetate, EC, PEG, PPG, PEG/PPG copolymers, PVP, HEC, HPC, CMC, CMEC, HPMC, HPMCP, HPMCAS, HPMCAT, poly(acrylic) acids and esters and poly(methacrylic) acids and esters and copolymers thereof, starch, dextran, dextrin, chitosan, collagen, gelatin, polyalkenes, polyethers, polysulfones, polyethersulfones, polystyrenes, polyvinyl halides, polyvinyl esters and ethers, natural waxes, and synthetic waxes.
  • Semipermeable membrane may also be a hydrophobic microporous membrane, wherein the pores are substantially filled with a gas and are not wetted by the aqueous medium but are permeable to water, as disclosed in U.S. Pat. No. 5,798,119. Such hydrophobic but water-permeable membrane are typically composed of hydrophobic polymers such as polyalkenes, polyethylene, polypropylene, polytetrafluoroethylene, polyacrylic acid derivatives, polyethers, polysulfones, polyethersulfones, polystyrenes, polyvinyl halides, polyvinylidene fluoride, polyvinyl esters and ethers, natural waxes, and synthetic waxes. The delivery port(s) on the semipermeable membrane may be formed postcoating by mechanical or laser drilling. Delivery port(s) may also be formed in situ by erosion of a plug of water-soluble material or by rupture of a thinner portion of the membrane over an indentation in the core. In addition, delivery ports may be formed during coating process.
  • The total amount of the active ingredient(s) released and the release rate can substantially by modulated via the thickness and porosity of the semipermeable membrane, the composition of the core, and the number, size, and position of the delivery ports.
  • The pharmaceutical compositions in an osmotic controlled-release dosage form may further comprise additional conventional excipients as described herein to promote performance or processing of the formulation.
  • The osmotic controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art.
  • In certain embodiments, the pharmaceutical compositions provided herein are formulated as AMT controlled-release dosage form, which comprises an asymmetric osmotic membrane that coats a core comprising the active ingredient(s) and other pharmaceutically acceptable excipients. The AMT controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art, including direct compression, dry granulation, wet granulation, and a dip-coating method.
  • In certain embodiments, the pharmaceutical compositions provided herein are formulated as ESC controlled-release dosage form, which comprises an osmotic membrane that coats a core comprising the active ingredient(s), hydroxylethyl cellulose, and other pharmaceutically acceptable excipients.
  • The pharmaceutical compositions provided herein in a modified release dosage form may be fabricated a multiparticulate controlled release device, which comprises a multiplicity of particles, granules, or pellets, ranging from about 10 pm to about 3 mm, about 50 pm to about 2.5 mm, or from about 100 pm to 1 mm in diameter. Such multiparticulates may be made by the processes know to those skilled in the art, including wet-and dry-granulation, extrusion/spheronization, roller-compaction, melt-congealing, and by spray-coating seed cores.
  • Other excipients as described herein may be blended with the pharmaceutical compositions to aid in processing and forming the multiparticulates. The resulting particles may themselves constitute the multiparticulate device or may be coated by various film forming materials, such as enteric polymers, water-swellable, and water-soluble polymers. The multiparticulates can be further processed as a capsule or a tablet.
  • Targeted Delivery
  • The pharmaceutical compositions provided herein may also be formulated to be targeted to a particular tissue, receptor, or other area of the body of the subject to be treated, including liposome-, resealed erythrocyte-, and antibody-based delivery systems.
  • Dosages
  • In the treatment, prevention, or amelioration of one or more symptoms of tic disorders or other conditions, disorders or diseases associated with VMAT2 inhibition, an appropriate dosage level generally is about 0.001 to 100 mg per kg patient body weight per day (mg/kg per day), about 0.01 to about 80 mg/kg per day, about 0.1 to about 50 mg/kg per day, about 0.5 to about 25 mg/kg per day, or about 1 to about 20 mg/kg per day, which may be administered in single or multiple doses. Within this range the dosage may be 0.005 to 0.05, 0.05 to 0.5, or 0.5 to 5.0, 1 to 15, 1 to 20, or 1 to 50 mg/kg per day. In certain embodiments, the dosage level is about 0.001 to 100 mg/kg per day.
  • In certain embodiments, the dosage level is about from 25 to 100 mg/kg per day. In certain embodiments, the dosage level is about 0.01 to about 40 mg/kg per day. In certain embodiments, the dosage level is about 0.1 to about 80 mg/kg per day. In certain embodiments, the dosage level is about 0.1 to about 50 mg/kg per day. In certain embodiments, the dosage level is about 0.1 to about 40 mg/kg per day. In certain embodiments, the dosage level is about 0.5 to about 80 mg/kg per day. In certain embodiments, the dosage level is about 0.5 to about 40 mg/kg per day. In certain embodiments, the dosage level is about 0.5 to about 25 mg/kg per day. In certain embodiments, the dosage level is about 1 to about 80 mg/kg per day. In certain embodiments, the dosage level is about 1 to about 75 mg/kg per day. In certain embodiments, the dosage level is about 1 to about 50 mg/kg per day. In certain embodiments, the dosage level is about 1 to about 40 mg/kg per day. In certain embodiments, the dosage level is about 1 to about 25 mg/kg per day.
  • In certain embodiments, the dosage level is about from 5.0 to 150 mg per day, and in certain embodiments from 10 to 100 mg per day. In certain embodiments, the dosage level is about 80 mg per day. In certain embodiments, the dosage level is about 40 mg per day.
  • For oral administration, the pharmaceutical compositions can be provided in the form of tablets containing 1.0 to 1,000 mg of the active ingredient, particularly about 1, about 5, about 10, about 15, about 20, about 25, about 30, about 40, about 45, about 50, about 75, about 80, about 100, about 150, about 200, about 250, about 300, about 400, about 500, about 600, about 750, about 800, about 900, and about 1,000 mg of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. In certain embodiments, the pharmaceutical compositions can be provided in the form of tablets containing about 100 mg of the active ingredient. In certain embodiments, the pharmaceutical compositions can be provided in the form of tablets containing about 80 mg of the active ingredient. In certain embodiments, the pharmaceutical compositions can be provided in the form of tablets containing about 75 mg of the active ingredient. In certain embodiments, the pharmaceutical compositions can be provided in the form of tablets containing about 50 mg of the active ingredient. In certain embodiments, the pharmaceutical compositions can be provided in the form of tablets containing about 40 mg of the active ingredient. In certain embodiments, the pharmaceutical compositions can be provided in the form of tablets containing about 25 mg of the active ingredient. The compositions may be administered on a regimen of 1 to 4 times per day, including once, twice, three times, and four times per day.
  • It will be understood, however, that the specific dose level and frequency of dosage for any particular patient may be varied and will depend upon a variety of factors including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the age, body weight, general health, sex, diet, mode and time of administration, rate of excretion, drug combination, the severity of the particular condition, and the host undergoing therapy.
  • The compounds provided herein may also be combined or used in combination with other agents useful in the treatment, prevention, or amelioration of one or more symptoms of the diseases or conditions for which the compounds provided herein are useful, including tic disorders and other conditions commonly treated with antipsychotic medication.
  • In certain embodiments, the compounds provided herein may also be combined or used in combination with a typical antipsychotic drug. In certain embodiments, the typical antipsychotic drug is fluphenazine, haloperidol, loxapine, molindone, perphenazine, pimozide, sulpiride, thioridazine, or trifluoperazine. In certain embodiments, the antipsychotic drug is an atypical antipsychotic drug. In certain embodiments, the atypical antipsychotic drug is aripiprazole, asenapine, clozapine, iloperidone, olanzapine, paliperidone, quetiapine, risperidone, or ziprasidone. In certain embodiments, the atypical antipsychotic drug is clozapine.
  • Such other agents, or drugs, may be administered, by a route and in an amount commonly used thereof, simultaneously or sequentially with the compounds provided herein. When compounds provided herein are used contemporaneously with one or more other drugs, a pharmaceutical composition containing such other drugs in addition to the compounds provided herein may be utilized, but is not required. Accordingly, the pharmaceutical compositions provided herein include those that also contain one or more other active ingredients or therapeutic agents, in addition to the compounds provided herein.
  • The weight ratio of the compounds provided herein to the second active ingredient may be varied, and will depend upon the effective dose of each ingredient. Generally, an effective dose of each will be used. Thus, for example, when the compounds provided herein are used in combination with the second drug, or a pharmaceutical composition containing such other drug, the weight ratio of the particulates to the second drug may range from about 1,000:1 to about 1: 1,000, or about 200:1 to about 1:200.
  • Combinations of the particulates provided herein and other active ingredients will generally also be within the aforementioned range, but in each case, an effective dose of each active ingredient should be used.
  • Examples of embodiments of the present disclosure are provided in the following examples. The following examples are presented only by way of illustration and to assist one of ordinary skill in using the disclosure. The examples are not intended in any way to otherwise limit the scope of the disclosure.
  • EXAMPLES Example 1 A Phase 1, Open-Label Study to Assess the Effect of Ketoconazole on the Pharmacokinetics of NBI-98854 in Healthy Subjects
  • This was a Phase 1, single-center, open-label, drug interaction study conducted in 24 healthy male and female subjects.
  • After providing informed consent, subjects were screened for eligibility to participate in the study. Screening started 21 days before Day 1 (the first day of study drug administration). Subjects who met the eligibility criteria were admitted to the research unit on Day −1 (the day prior to dosing) and remained at the study center until the end of the study (Day 10 or early termination). On Day −1, a blood sample was collected to determine cytochrome P450 2D6 (CYP2D6) status.
  • All subjects received the same treatment. Subjects received a single dose of 50 mg NBI-98854 on Days 1 and 6 at approximately 0800 hours. In addition, subjects received 200 mg ketoconazole twice daily (bid) on Days 5 through 9 at approximately 0800 and 2000 hours. Subjects were required to fast overnight from approximately 10 hours before until 4 hours after NBI-98854 dose on Days 1 and 6. Subjects were given meals upon completion of study assessments/dosing and at regular meal times during their stay in the research unit.
  • Blood samples for pharmacokinetic (PK) analyses of NBI-98854 and its metabolites were collected at set time intervals up to 96 hours post NBI-98854 dosing. Blood specimens to determine ketoconazole plasma concentrations were collected predose through Day 9. Safety and tolerability was assessed through Day 10.
  • NBI-98854 was administered as a 50 mg capsule orally as a single dose, twice during the treatment period on Days 1 and 6.
  • Ketoconazole was administered as a 200 mg tablet orally as a single dose, twice daily on Days 5 through 9.
  • Pharmacokinetics
  • Blood samples for PK analyses of NBI-98854 and its metabolites were collected before and after the Days 1 and 6 dosing: at 45 minutes prior to NBI-98854 dosing, and at 15, 30, 45 minutes, and 1, 1.5, 2, 3, 4, 8, 12, 18, 24, 48, 72 and 96 hours after NBI-98854 dosing or at early termination.
  • Blood samples to determine ketoconazole plasma concentrations were collected at 45 minutes pre- and 1 hour post-morning ketoconazole dosing on Day 5, at 1 and 8 hours post-morning ketoconazole dosing on Day 6, and at 1 hour post-morning ketoconazole dosing on Days 7-9 or at early termination.
  • The following PK parameters were calculated for NBI-98854 and the NBI-98782 and NBI-136110 metabolites:
      • area under the plasma concentration versus time curve (AUC) from 0 to 24 hours postdose (AUC0-24)
      • AUC from time 0 to the time of the last quantifiable concentration (AUC0-tlast)
      • AUC extrapolated to infinity (AUC0-∞), maximum plasma concentration (Cmax)
      • time to maximum plasma concentration (tmax)
      • time prior to the first measurable concentration (Tlag)
      • apparent terminal half-life (t1/2)
      • apparent terminal rate constant (λz)
      • apparent mean residence time (MRT) and
      • molar ratio of the metabolites NBI-98782 and NBI-136110 to the parent drug NBI-98854.
  • No PK parameters were calculated for ketoconazole.
  • Plasma concentrations of NBI-98854, its metabolites, and ketoconazole were summarized with descriptive statistics and in figures. An analysis of variance (ANOVA) model was used to compare AUC0-∞ and Cmax for NBI-98854 administered with ketoconazole (“test”) versus AUC0-∞ and Cmax for NBI-98854 alone (“reference”). The tmax was analyzed using non-parametric methods. The PK parameters for metabolites of NBI-98854 were also evaluated. Safety data were summarized using descriptive statistics.
  • The PK data for tmax, Tlag, t1/2, MRT, and Vz/F were rounded to 2 significant figures and all other parameters (AUC0-24, AUC0-tlast, AUC0-∞, Cmax, and CL/F) were rounded to 3 significant figures. The last significant figure was rounded up if the digit to the right of it was ≥5, and was rounded down if the digit to the right of it was ≤4.
  • Pharmacokinetic Results
  • NBI-98854 was readily absorbed after oral administration alone or in combination with ketoconazole. Mean plasma concentrations were higher after combination treatment compared to NBI-98854 alone.
  • PK parameters for NBI-98854 after treatment with NBI-98854 alone or in combination with ketoconazole are summarized below.
  • Mean AUC0-∞ for NBI-98854 after treatment with ketoconazole was approximately 2.1-fold greater than that after treatment with NBI-98854 alone. Mean Cmax for NBI-98854 after treatment with NBI-98854 plus ketoconazole was also approximately 1.5-fold greater than that after treatment with NBI-98854 alone. Median tmax values were the same after treatment with NBI-98854 plus ketoconazole and NBI-98854 alone (0.75 hours) and mean t1/2 values were higher after treatment with NBI-98854 plus ketoconazole than after treatment with NBI-98854 alone (20 and 16 hours, respectively).
  • The geometric CV % for NBI-98854 after treatment with NBI-98854 plus ketoconazole and NBI-98854 alone was generally similar for AUC0-∞, Cmax, t1/2, and MRT. The geometric mean ratios and associated 90% confidence intervals (CIs) for AUC0-∞ and Cmax for NBI-98854 after treatment with NBI-98854 alone or in combination with ketoconazole are provided below.
  • Geometric mean ratios for AUC0-∞ and Cmax for NBI-98854 after treatment with NBI-98854 alone or in combination with ketoconazole were 213.7% and 151.1%, respectively. The corresponding upper and lower 90% CI bounds for AUC0-∞ (2.04 to 2.24) and Cmax(1.41 to 1.62) were outside the ‘no effect’ range of 80% to 125%, indicating an effect of treatment with ketoconazole on NBI-98854 AUC0-∞ and Cmax.
  • Summary of Pharmacokinetic Parameters for NBI-98854 (PK Analysis Set)
  • NBI-98854 (80 mg) +
    Parameter NBI-98854 (50 mg) Ketoconazole (200 mg)
    Statistic (N = 24) (N = 24)
    AUC0-24 (ng hr/mL)
    Mean (SD) 2580 (943) 4820 (1450)
    Geometric CV % 38.1 33.1
    AUC0-tlast (ng hr/mL)
    Mean (SD) 3420 (1230) 7100 (2260)
    Geometric CV % 37.4 33.7
    AUC0-∞ (ng hr/mL)
    Mean (SD) 3470 (1240) 7340 (2360)
    Geometric CV % 37.0 33.9
    Cmax (ng/mL)
    Mean (SD) 425 (176) 624 (221)
    Geometric CV % 47.7 3i.7
    tmax (hours)
    Median (min, max) 0.75 (0.48, 1.5) 0.75 (0.50, 2.0)
    Tlag (hours)
    Mean (SD) 0.11 (0.13) 0.02 (0.07)
    t1/2 (hours)
    Mean (SD) 16 (2.2) 20 (3.1)
    Geometric CV % 15 16
    MRT (hours)
    Mean (SD) 17 (2.8) 22 (4.2)
    Geometric CV % 17 19
    CL/F (L/hr)
    Mean (SD) 16.3 (5.94) 7.96 (2.75)
    Geometric CV % 37.0 33.9
    Vz/F (L)
    Mean (SD) 370 (136) 220 (88.9)
    Geometric CV % 37.1 36.2
  • NBI-98854 Geometric Mean Ratios for Pharmacokinetic Exposure Parameters (PK Analysis Set)
  • Ratioa (%) (NBI-98854 with 90% Confidence
    Parameter ketoconazole vs. NBI-98854 alone) Intervalb
    AUC0-∞ 213.7 203.6, 224.3
    Cmax (ng/mL) 151.1 140.6, 162.3
    aRatio of geometric least-squares means was based on a mixed model using log-transformed (base 10) data.
    bThe 90% confidence interval for geometric mean ratio was based on least-squares means using log-transformed (base 10) data
  • Plasma concentrations for the active metabolite NBI-98782 increased after oral administration of NBI-98854 alone or in combination with ketoconazole.
  • PK parameters for NBI-98782 after treatment with NBI-98854 alone or in combination with ketoconazole are below.
  • Mean AUC0-∞ for NBI-98782 after treatment with NBI-98854 in combination with ketoconazole was approximately 2.1-fold greater than mean AUC after treatment with NBI-98854 alone. Mean Cmax for NBI-98782 after treatment with NBI-98854 plus ketoconazole was approximately 1.6-fold greater than that after treatment with NBI-98854 alone. Median tmax values were the same after treatment with NBI-98854 plus ketoconazole and NBI-98854 alone (4.0 hours) and mean t1/2 was higher after treatment with NBI-98854 plus ketoconazole than after treatment with NBI-98854 alone (21 and 18 hours, respectively). The variability in PK (ie, geometric CV %) for NBI-98782 after treatment with NBI-98854 alone and in combination with ketoconazole was generally similar for AUC, Cmax, t1/2, and MRT.
  • The NBI-98782 geometric mean ratios and associated 90% CIs for AUC0-∞ and Cmax after treatment with NBI-98854 alone versus in combination with ketoconazole are provided below.
  • Geometric mean ratios for AUC0-∞ and Cmax for NBI-98782 after treatment with NBI-98854 alone versus in combination with ketoconazole were 206.7% and 162.9%, respectively, which were similar to the observed ratios for NBI-98854. The corresponding upper and lower 90% CI bounds for both parameters were outside the ‘no effect’ range of 80% to 125%, indicating an effect of treatment with ketoconazole on NBI-98782 AUC0-∞ and Cmax.
  • Summary of Pharmacokinetic Parameters for NBI-98854 (PK Analysis Set)
  • NBI-98854 (50 mg) +
    Parameter NBI-98854 (50 mg) Ketoconazole (600 mg)
    Statistic (N = 24) (N = 24)
    AUC0-24 (ngxhr/mL)
    Mean (SD) 237 (110) 419 (188)
    Geometric CV % 37.8 38.7
    AUC0-tlast (ngxhr/mL)
    Mean (SD) 435 (236) 880 (488)
    Geometric CV % 40.6 42.8
    AUC0-∞ (ngxhr/mL)
    Mean (SD) 450 (262) 935 (534)
    Geometric CV % 41.3 43.8
    Cmax (ng/mL)
    Mean (SD) 14.1 (5.48) 23.4 (10.4)
    Geometric CV % 35.0 39.6
    tmax (hr)
    Median (min, max) 4.0 (4.0, 1.8) 4.0 (4.0, 2.4)
    Tlag (hr)
    Mean (SD) 0.24 (0.051) 0.00 (0.00)
    t1/2 (hr)
    Mean (SD) 18 (2.4) 21 (3.3)
    Geometric CV % 13 16
    MRT (hr)
    Mean (SD) 28 (0.4) 34 (5.3)
    Geometric CV % 14 16
  • NBI-98854 Geometric Mean Ratios for Pharmacokinetic Exposure Parameters (PK Analysis Set)
  • Ratioa (%) (NBI-98854 with 90% Confidence
    Parameter ketoconazole vs. NBI-98854 alone) Intervalb
    AUC0-∞ 206.7 198.4, 215.4
    (ngxhr/mL)
    Cmax (ng/mL) 162.9 153.8, 172.5
    aRatio of geometric least-squares means was based on a mixed model using log-transformed (base 10) data.
    bThe 90% confidence interval for geometric mean ratio was based on least-squares means using log-transformed (base 10) data
  • Plasma concentrations for the metabolite NBI-136110 decreased after oral administration of NBI-98854 in combination with ketoconazole compared with administration of NBI-98854 alone. PK parameters for NBI-136110 after treatment with NBI-98854 alone or in combination with ketoconazole are summarized below.
  • Mean AUC0-tlast for NBI-136110 after treatment with NBI-98854 in combination with ketoconazole was 46% of the observed value after treatment with NBI-98854 alone.
  • Similarly, mean Cmax for NBI-136110 after treatment with NBI-98854 plus ketoconazole was 22% of the observed value with NBI-98854 alone. Median tmax values were approximately 4-fold higher after treatment with NBI-98854 plus ketoconazole versus NBI-98854 alone. Mean t1/2 for NBI-136110 after treatment with NBI-98854 with ketoconazole was higher than for NBI-98854 alone; however, because the calculated t1/2 of NBI-136110 after treatment with NBI-98854 plus ketoconazole was longer than the blood collection period of 96 hours (120 hours), this value is of uncertain accuracy. Consequently, the AUC0-∞ and MRT of NBI-136110 after treatment of NBI-98854 plus ketoconazole are also of uncertain accuracy.
  • NBI-136110 Geometric Mean Ratios for Pharmacokinetic Exposure Parameters (PK Analysis Set)
  • NBI-98854 (50 mg) +
    Parameter NBI-98854 (50 mg) Ketoconazole (200 mg)
    Statistic (N = 24) (N = 24)
    AUC0-24 (ngxhr/mL)
    Mean (SD) 588 (177) 161 (64.0)
    Geometric CV % 35.8 38.2
    AUC0-tlast (ngxhr/mL)
    Mean (SD) 1210 (333) 567 (223)
    Geometric CV % 29.7 38.8
    AUC0-∞ (ngxhr/mL)
    Mean (SD) 1330 (376) Not applicable
    Geometric CV % 29.0
    Cmax (ng/mL)
    Mean (SD) 37.8 (14.0) 8.05 (3.31)
    Geometric CV % 47.4 39.3
    tmax (hr)
    Median (min, max) 4.0 (1.5, 24) 18 (2.0, 48)
    Tlag (hr)
    Mean (SD) 0.18 (0.12) 0.00 (0.00)
    t1/2 (hr)
    Mean (SD) 28 (5.3) Not applicable
    Geometric CV % 19
    MRT (hr)
    Mean (SD) 40 (7.5) Not applicable
    Geometric CV % 19
  • The NBI-136110 geometric mean ratios and associated 90% CIs for AUC0-∞ and Cmax after treatment with NBI-98854 alone or in combination with ketoconazole are provided below. Geometric mean ratios for AUC0-tlast and Cmax for NBI-136110 after treatment with NBI-98854 alone or in combination with ketoconazole were 45.6% and 21.6%, respectively. The 90% CI for AUC0-tlast and Cmax were outside the ‘no effect’ range of 0.80 to 1.25.
  • NBI-136110 Geometric Mean Ratios for Pharmacokinetic Exposure Parameters (PK Analysis Set)
  • Ratioa (%) (NBI-98854 with 90% Confidence
    Parameter ketoconazole vs. NBI-98854 alone) Intervalb
    AUC0-∞ 45.6 41.8, 49.7
    Cmax (ng/mL) 21.6 18.8, 24.8
    aRatio of geometric least-squares means was based on a mixed model using log-transformed (base 10) data.
    bThe 90% confidence interval for geometric mean ratio was based on least-squares means using log-transformed (base 10) data
  • Ketoconazole plasma concentrations were measured on Day 5 at 45 minutes before and 1 hour after the morning ketoconazole dose, on Day 6 at 1 and 8 hours after the morning ketoconazole dose, and on Days 7 to 9 at 1 hour after the morning ketoconazole dose, or upon early termination._Measurable concentrations of ketoconazole were present in all postdose plasma samples analyzed. Mean concentrations of ketoconazole ranged from 1,643 to 5,177 ng/mL on Days 5 to 9 at 1 hour postdose.
  • Administration of NBI-98854 plus ketoconazole led to a 1.5-fold increase in peak plasma concentration (Cmax) and a 2.1-fold increase in total exposure (AUC0-∞) of NBI-98854 compared with administration of NBI-98854 alone. The 90% CI for the geometric mean ratios (1.41 to 1.62 for Cmax and 2.04 to 2.24 for AUC0-∞) were outside the ‘no effect’ range of 0.80 to 1.25.
  • Administration of NBI-98854 plus ketoconazole also led to a 1.6-fold increase in Cmax and a 2.1-fold increase in AUC0-∞ of the metabolite NBI-98782 compared with administration of NBI-98854 alone. The 90% CI for the geometric mean ratios were outside the ‘no effect’ range of 0.80 to 1.25.
  • For the metabolite NBI-136110, mean AUC0-tlast and Cmax after treatment with NBI-98854 plus ketoconazole was 46% and 22%, respectively, of the observed value after treatment with NBI-98854 alone. The 90% CI for the geometric mean ratio was outside the ‘no effect’ range of 0.80 to 1.25.
  • Median tmax was the same after treatment with NBI-98854 plus ketoconazole and after treatment with NBI-98854 alone for NBI-98854 (0.75 hours) and NBI-98782 (4.0 hours). For NBI-136110, median tmax was higher after treatment with NBI-98854 plus ketoconazole than after treatment with NBI-98854 alone (18 and 4.0 hours, respectively).
  • Mean t1/2 of NBI-98854 increased from 16 to 20 hours when NBI-98854 was administered with ketoconazole. Similarly, mean t1/2 of NBI-98782 increased from 18 to 21 hours when NBI-98854 was administered with ketoconazole. The mean tin of NBI-136110 was also increased, but the magnitude of change is of uncertain accuracy.
  • Administration of NBI-98854 plus ketoconazole led to a 1.5-fold increase in peak plasma concentration (Cmax) and a 2.1-fold increase in total exposure (AUC0-∞) of NBI-98854 compared with administration of NBI-98854 alone. The 90% confidence intervals (CI) for the geometric mean ratios (1.41 to 1.62 for Cmax and 2.04 to 2.24 for AUC0-∞) were outside the ‘no effect’ range of 0.80 to 1.25.
  • Administration of NBI-98854 plus ketoconazole also led to a 1.6-fold increase in Cmax and a 2.1-fold increase in AUC0-∞ of the metabolite NBI-98782 compared with administration of NBI-98854 alone. The 90% CI for the geometric mean ratios were outside the ‘no effect’ range of 0.80 to 1.25.
  • For the metabolite NBI-136110, mean AUC0-∞ and Cmax after administration of NBI-98854 plus ketoconazole were 46% and 22%, respectively, of the observed values after administration of NBI-98854 alone. The 90% CI for the geometric mean ratios were outside the ‘no effect’ range of 0.80 to 1.25.
  • Median tmax was the same after treatment with NBI-98854 plus ketoconazole and after treatment with NBI-98854 alone for NBI-98854 (0.75 hours) and NBI-98782 (4.0 hours). For NBI-136110, median tmax was higher after treatment with NBI-98854 plus ketoconazole than after treatment with NBI-98854 alone (18 and 4.0 hours, respectively).
  • Mean t1/2 of NBI-98854 increased from 16 to 20 hours when it was administered with ketoconazole. Similarly, mean tin of NBI-98782 increased from 18 to 21 hours when NBI-98854 was administered with ketoconazole. The mean t1/2 of NBI-136110 was also increased, but the magnitude of change is of uncertain accuracy.
  • Safety
  • Safety was assessed based on adverse events (AEs), clinical laboratory tests (hematology, serum chemistry, and urinalysis), vital signs (including orthostatic blood pressure), physical examinations, and 12-lead electrocardiograms (ECGs).
  • Safety Results
  • AEs were assigned to a treatment based on the time of the most recent treatment administered prior to the AE onset. TEAEs are summarized below. There were no pretreatment AEs. There were no deaths, SAEs, or discontinuations due to an AE reported in this study. Five subjects (20.8%) experienced 6 AEs during the study; all AEs occurred after the initial treatment with NBI-98854 alone. All AEs had an onset on Day 1 or 2. The only AE that occurred in >1 subject was headache (2 subjects, 8.3%). All AEs were mild in intensity. Two events were considered possibly related to study drug, 1 event of headache and the event of dizziness postural. The other event of headache was considered unlikely related to study drug.
  • The number and percentage of subjects who experienced TEAEs are summarized below.
  • Number and Percentage of Subjects Who Experienced a Treatment-Emergent Adverse Event (Safety Analysis Set)
  • NBI-98854 (50 mg) +
    NBI-98854 (50 mg) Ketoconazole (200 mg) (n,
    Preferred (n, %) %)
    Term N = 24 N = 24
    Overall  5 (20.8) 0
    Gastrointestinal Disorders 2 (8.3) 0
    Constipation 1 (4.2) 0
    Nausea 1 (4.2) 0
    Injury, Poisoning and 1 (4.2) 0
    Procedural Complications 1 (4.2) 0
    Procedural dizziness
    Nervous System Disorders  3 (12.5) 0
    Dizziness postural 1 (4.2) 0
    Headache 2 (8.3) 0
  • There were no deaths, SAEs, or discontinuations due to an AE. Five subjects (20.8%) experienced 6 AEs during the study; all AEs occurred after the initial treatment with NBI-98854 alone, with an onset on Day 1 or 2. The only AE that occurred in >1 subject was headache (2 subjects, 8.3%). Two events were considered possibly related to study drug, 1 event of headache and the event of dizziness postural, and the other event of headache was considered unlikely related to study drug; all were mild in intensity.
  • Mean clinical laboratory values were similar pre- and postdose, and there were no results considered clinically significant or documented as an AE by the investigator for any hematology, serum chemistry, or urinalysis value. No clinically relevant postdose mean vital signs changes, including orthostatic changes, were observed. No abnormal physical examination findings occurred during the study, and there were no important changes in body weight. There were no abnormal ECGs that were assessed as clinically significant during the study.
  • Conclusions
  • Administration of NBI-98854 plus ketoconazole led to a 1.5-fold increase in Cmax and a 2.1-fold increase in AUC0-∞ of NBI-98854 compared with administration of NBI-98854 alone.
  • Administration of NBI-98854 plus ketoconazole also led to a 1.6-fold increase in Cmax and a 2.1-fold increase in AUC0-∞ of the metabolite NBI-98782 compared with administration of NBI-98854 alone.
  • Mean AUC0-tlast and Cmax for the metabolite NBI-136110 after treatment with NBI-98854 plus ketoconazole were 46% and 22%, respectively, of the observed values after treatment with NBI-98854 alone.
  • The 90% CI for all the above parameters were outside the 90% CI range of 0.80 to 1.25.
  • NBI-98854 was well tolerated when administered alone and concomitantly with ketoconazole in healthy volunteers.
  • Example 2 Pharmacologic Characterization of Valbenazine, Tetrabenazine, and Metabolite Thereof
  • Upon oral administration, TBZ is reduced to form four discrete isomeric secondary alcohol metabolites, collectively referred to as dihydrotetrabenazine (DHTBZ), which contains three asymmetric carbon centers (C-2, C-3, and C-11β), which could hypothetically result in eight stereoisomers. However, because the C-3 and C-11β carbons have fixed relative configurations, only four stereoisomers are possible: (R,R,R-DHTBZ or (+)-α-DHTBZ (alternate nomenclature) or NBI-98782 (laboratory nomenclature); S,S,S-DHTBZ or (−)-α-DHTBZ or NBI-98771; S,R,R-DHTBZ or (+)-β-DHTBZ or NBI-98795; and R,S,S-DHTBZ or (−)-β-DHTBZ or NBI-98772.
  • The affinity of each compound was measured by inhibition of [3H]-DHTBZ binding to rat forebrain membranes. The affinities relative to R,R,R-DHTBZ were also calculated and are presented. Data are reported as both the negative logarithm of the Ki (pKi) for statistical calculation with the normally distributed binding parameter used to determine the mean and SEM. The Ki value was determined from the mean pKi as 10(-pKi). The R,R,R-DHTBZ stereoisomer binds with the highest affinity to both rat and human VMAT2 (Ki=1.0 to 4.2 nM). In comparison, the remaining three DHTBZ stereoisomers (S,R,R-DHTBZ, S,S,S-DHTBZ, R,S,S-DHTBZ) bind to VMAT2 with a Ki values of 9.7, 250, and 690 nM, respectively.
  • In Vitro VMAT2 Binding Affinity in Rat Forebrain
  • VMAT2
    Affinity
    pKi mean Relative to
    Compound Ki,, nm (SEM) N R,R,R-DHTBZa
    R,R,R-DHTBZ 4.2 8.38 (0.42) 27 1.0
    S,R,R-DHTBZ 9.7 8.01 (0.32)  6 2.3
    S,S,S-DHTBZ 250 6.60 (0.22)  4 60
    R,S,S-DHTBZ 690 6.16 (0.05)  5 160
    aAffinity relative to R,R,R-DHTBZ was calculated using the Ki value determined in the same study
  • The primary metabolic clearance pathways of valbenazine (VBZ, NBI-98854) are hydrolysis (to form R,R,R-DHTBZ) and mono-oxidation (to form the metabolite NBI-136110). R,R,R-DHTBZ and NBI-136110, the two most abundant circulating metabolites of VBZ, are formed gradually and their plasma concentrations decline with half-lives similar to VBZ.
  • VBZ and its metabolites, R,R,R-DHTBZ and NBI-136110, were tested for their ability to inhibit the binding of [3H]-DHTBZ to VMAT2 in cell lines or native tissues. The affinity of each compound was measured by inhibition of [3H]-DHTBZ binding to either human platelets or rat striatal membranes. The affinities relative to R,R,R-DHTBZ were also calculated and are presented. Data are reported as both the negative logarithm of the Ki (pKi) for statistical calculation with the normally distributed binding parameter used to determine the mean and SEM (n=4 for each compound in each tissue). The Ki value was determined from the mean pKi as 10(−pKi). The primary metabolite R,R,R-DHTBZ, was the most potent inhibitor of VMAT2 in rat striatum and human platelet homogenates.
  • In Vitro VMAT2 Binding Affinity of Valbenazine and its Metabolites
  • Rat Striatum Human Platelets
    Affinity Affinity
    Relative Relative
    Ki, pKi mean to R,R,R- Ki, pKi mean to R,R,R-
    Compound nm (SEM) DHTBZ nm (SEM) DHTBZ
    Valbenazine 110 6.95 (0.02) 39 150 6.82 45
    (0.02)
    R,R,R- 1.98 8.70 (0.09) 1.0 3.1 8.52 1.0
    DHTBZ (0.03)
    NBI-136610 160 6.80 (0.02) 57 220 6.65(0.04) 67
  • VBZ and NBI-136110 had similar effects on VMAT2 inhibition, but with Ki values that were approximately 40-65 times the Ki values (lower affinity) of R,R,R-DHTBZ. These results were corroborated by the radioligand binding assay of DHTBZ stereoisomers (i.e., TBZ metabolites) in the rat forebrain, which also showed R,R,R-DHTBZ to be the most potent inhibitor of VMAT2, followed by S,R,R-DHTBZ. Comparatively, S,S,S-DHTBZ and R,S,S-DHTBZ, the other two primary metabolites of TBZ, were found to be poor VMAT2 inhibitors with affinities approximately 60 and 160 times weaker than R,R,R-DHTBZ.
  • The affinity of VBZ and its metabolites R,R,R-DHTBZ and NBI-136110 for other targets beyond VMAT2 was assessed in an extensive Cerep screen of multiple classes of protein targets including GPCRs, cell-surface monoamine transporters, and ion channels including the cardiac potassium channel, human ether-a-go-go-related gene (HERG).
  • The multi-target activity screen of more than 80 targets for these compounds (Cerep screen) demonstrated that VBZ and its metabolites, R,R,R-DHTBZ and NBI-136110, did not inhibit the binding of cognate ligands to any of the targets by more than 50% at concentrations of 1-10 μM. In contrast, the other three DHTBZ stereoisomers (S,R,R-DHTBZ, S,S,S-DHTBZ, R,S,S-DHTBZ), which are metabolites of TBZ but not VBZ, demonstrated >50% inhibition of ligand binding to a number of receptor subtypes including serotonin, dopamine and adrenergic receptors. Results expressed as percent of control specific binding: (tested compound specific binding/control specific binding)×100. All compounds were tested at 1 or 10 μM final concentration and results are an excerpt of a larger 80 target panel performed as an initial screen at Cerep (n=2 for each compound at each target). Bolded results (>50%) indicate activity at target receptor.
  • In Vitro Activity of Valbenazine and DHTBZ Stereoisomers at Dopamine, Serotonin, and Adrenergic Receptors
  • Receptor R,R,R- S,S,S-DHTBZ/
    Target Valbenazine DHTBZ S,R,R-DHTBZ R,S,S-DHTBZa
    Serotonin5- 26 17 69 96
    HT1A
    Serotonin5-  1 −4  3 84
    HT2A
    Serotonin5-HT7  4  3 80 98
    Dopamine D1  8 −6 −5 82
    Dopamine D2(s)  2  6 25 89
    aFor purposes of the broad panel screen, the S,S,S- and R,S,S-netabolites were tested as a 50/50 mixture.
  • To describe the monoamine systems in greater detail, detailed radioligand binding assays were performed for dopamine, serotonin and adrenergic receptor subtypes as well as the transporters for dopamine (DAT), serotonin (SERT), and norepinephrine (NET) for the common metabolite of TBZ and VBZ (R,R,R-DHTBZ) and the other relevant metabolites unique to TBZ and VBZ. This detailed analysis revealed the high specificity of R,R,R-DHTBZ for the VMAT2 transporter and the non-specific activities of the other TBZ metabolites, including relatively high affinity for dopamine and serotonin receptor subtypes. Interestingly, the R,R,R-DHTBZ metabolite showed the greatest non-selectivity with respect to the monoamine receptors. None of the TBZ or VBZ metabolites had any affinity for the monoamine transporters DAT, SERT or NET. To complete the selectivity profile for VMAT2, the functional activity for the human VMAT1 transporter of these compounds was tested in cells expressing VMAT1. While the non-selective irreversible high-affinity uptake inhibitor of VMAT1, reserpine, substantially inhibited uptake through VMAT1, there was no significant inhibitory activity of TBZ, VBZ, or its metabolites R,R,R-DHTBZ or NBI-136110 at concentrations up to 10 μM. For both VMAT1 and VMAT2, uptake was measured in the untransfected host cells and was found to be similar to transfected cells in the presence of excess reserpine.
  • Radioligand binding assays and the broad panel screen indicate that in addition to varying potency at the VMAT2 transporter, two of the other DHTBZ metabolites of TBZ (S,S,S-DHTBZ and R,S,S-DHTBZ) interact with D1 and D2 receptors. Since VBZ is not metabolized to either of these DHTBZ stereoisomers, its effects on postsynaptic dopamine receptors either directly or indirectly through the metabolites are non-existent.
  • Moreover, results from the broad panel screen indicate that VBZ and its major metabolites (R,R,R-DHTBZ and NBI-136110) have little to no affinity for more than 80 binding sites, including receptors, monoamine transporters, and ion channels. This profile suggests a low potential for off-target pharmacological effects. In addition, uptake studies using TBZ, VBZ and its metabolites, R,R,R-DHTBZ and NBI-136110, confirmed the selectivity of these compounds for VMAT2 as they had no significant effect on the uptake of monoamines through VMAT1 compared to reserpine, a known VMAT1/VMAT2 inhibitor.
  • The selectivity and specificity of VBZ was distinctively demonstrated using two in vivo surrogate measures of pharmacological effects. Ptosis, known to occur via adrenergic activation and prolactin release from the pituitary, modulated through the D2 dopamine receptor, demonstrated the difference between treatment with TBZ and VBZ. TBZ, VBZ and R,R,R-DHTBZ induced ptosis in an equivalent manner. This confirms that the metabolites formed by dosing TBZ or VBZ, or dosing of the active metabolite itself (R,R,R-DHTBZ) all have activity at VMAT2 affecting presynaptic monoamine release, in this case, related to norepinephrine release specifically to induce ptosis. Following similar treatment, but this time using prolactin release as a surrogate for dopaminergic modulation, R,R,R-DHTBZ and VBZ (to a lesser extent) induced a similar increase in serum prolactin levels as TBZ.
  • The various embodiments described above can be combined to provide further embodiments. All of the U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the Application Data Sheet are incorporated herein by reference, in their entirety. Aspects of the embodiments can be modified, if necessary to employ concepts of the various patents, applications and publications to provide yet further embodiments.
  • These and other changes can be made to the embodiments in light of the above-detailed description. In general, in the following claims, the terms used should not be construed to limit the claims to the specific embodiments disclosed in the specification and the claims, but should be construed to include all possible embodiments along with the full scope of equivalents to which such claims are entitled. Accordingly, the claims are not limited by the disclosure

Claims (108)

1. A method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof wherein the patient is also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising:
administering the VMAT2 inhibitor in an amount equivalent to about 40 mg of valbenazine free base once daily to the patient.
2. A method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof wherein the patient is also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising:
administering a therapeutically effective amount of the VMAT2 inhibitor to the patient,
wherein the therapeutically effective amount of the VMAT2 inhibitor is less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
3. A method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof, comprising:
administering to the patient a therapeutically effective amount of the VMAT2 inhibitor,
subsequently determining that the patient is to begin treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor, and
administering the VMAT2 inhibitor in an amount equivalent to about 40 mg of valbenazine free base once daily to the patient.
4. A method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof, comprising:
administering a therapeutically effective amount of the VMAT2 inhibitor to the patient,
subsequently determining that the patient is to begin treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor, and
administering the VMAT2 inhibitor in an amount that is less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
5. A method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, to a patient in need thereof wherein the patient is also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising:
administering a therapeutically effective amount of the VMAT2 inhibitor to the patient,
wherein the administration produces a mean valbenazine Cmax that is about 1 to about 2 fold higher than the mean valbenazine Cmax for a patient who is not being administered a strong CYP3A4 inhibitor and/or a mean valbenazine AUC0-∞ that is about 1.5 to about 2.5 fold higher than the mean valbenazine AUC0-∞ for a patient who is not being administered a strong CYP3A4 inhibitor.
6. A method of administering a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof to a patient in need thereof wherein patient is also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising:
administering a therapeutically effective amount of the VMAT2 inhibitor,
wherein the administration produces a mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol Cmax that is about 1 to about 2 fold higher than the mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol Cmax for a patient who is not being administered a strong CYP3A4 inhibitor and/or a mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol AUC0-∞ that is about 1.5 to about 2.5 fold higher than the mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol AUC0-∞ for a patient who is not being administered a strong CYP3A4 inhibitor.
7. The method of any one of claims 1 to 6, further comprising informing the patient or a medical care worker that administration of the VMAT2 inhibitor to patients who is also being administered a strong CYP3A4 inhibitor in higher exposure of valbenazine and/or (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol than administration of the VMAT2 inhibitor to a patient who is not being administered a strong CYP3A4 inhibitor.
8. The method of any one of claims 1 to 7, further comprising informing the patient or a medical care worker that administration of the VMAT2 inhibitor to a patient who is also being administered a strong CYP3A4 inhibitor may result in increased risk of one or more exposure-related adverse reactions than administration of the VMAT2 inhibitor to a patient who is not being administered a strong CYP3A4 inhibitor.
9. The method of claim 8, wherein the one or more exposure-related adverse reactions is chosen from somnolence, anticholinergic effects, balance disorders or falls, headache, akathisia, vomiting, nausea, arthralgia, QT prolongation, increase in blood glucose, increase in weight, respiratory infections, drooling, dyskinesia, extrapyramidal symptoms (non-akathisia), anxiety, insomnia, increase in prolactin, increase in alkaline phosphatase, and increase in bilirubin.
10. The method of claim 9, wherein the one or more exposure-related adverse reactions is chosen from somnolence, anticholinergic effects, balance disorders or falls, headache, akathisia, vomiting, nausea, arthralgia, and QT prolongation.
11. The method of claim 10, wherein the one or more exposure-related adverse reactions is chosen from somnolence and QT prolongation.
12. The method of any one of claims 1 to 11, further comprising informing the patient or a medical care worker that co-administration of the VMAT2 inhibitor and the CYP3A4 inhibitor may prolong the patient's QT interval.
13. The method of any one of claims 1 to 12, further comprising informing the patient or a medical care worker that administration of the VMAT2 inhibitor to a patient who is also being administered a strong CYP3A4 inhibitor may prolong the patient's QT interval than administration of the VMAT2 inhibitor to a patient who is also being administered a strong CYP3A4 inhibitor.
14. The method of any one of claims 1 to 13, wherein the strong CYP3A4 inhibitor is chosen from clarithromycin, chloramphenicol, cobicistat, indinavir, itraconazole, ketoconazole, nefazodone, nelfinavir, ritonavir, saquinavir, and telithromycin.
15. The method of claim 14, wherein the strong CYP3A4 inhibitor is chosen from clarithromycin, itraconazole, and ketoconazole.
16. The method of claim 15, wherein the strong CYP3A4 inhibitor is ketoconazole.
17. The method of any one of claims 1 to 16, wherein the VMAT2 inhibitor is administered to the patient to treat a neurological or psychiatric disease or disorder.
18. The method of claim 17, wherein the neurological or psychiatric disease or disorder is a hyperkinetic movement disorder, mood disorder, bipolar disorder, schizophrenia, schizoaffective disorder, mania in mood disorder, depression in mood disorder, treatment-refractory obsessive compulsive disorder, neurological dysfunction associated with Lesch-Nyhan syndrome, agitation associated with Alzheimer's disease, Fragile X syndrome or Fragile X-associated tremor-ataxia syndrome, autism spectrum disorder, Rett syndrome, or chorea-acanthocytosis.
19. The method of claim 18, wherein the neurological or psychiatric disease or disorder is a hyperkinetic movement disorder.
20. The method of claim 18, wherein the hyperkinetic movement disorder is tardive dyskinesia.
21. The method of claim 18, wherein the hyperkinetic movement disorder is Tourette's syndrome.
22. The method of claim 18, wherein the hyperkinetic movement disorder is Huntington's disease.
23. The method of claim 18, wherein the hyperkinetic movement disorder is tics.
24. The method of claim 18, wherein the hyperkinetic movement disorder is chorea associated with Huntington's disease.
25. The method of claim 18, wherein the hyperkinetic movement disorder is ataxia, chorea, dystonia, Huntington's disease, myoclonus, restless leg syndrome, or tremors.
26. The method of any one of claims 1 to 25, wherein the VMAT2 inhibitor is administered orally.
27. The method of any one of claims 1 to 26, wherein the VMAT2 inhibitor is administered in the form of a tablet or capsule.
28. The method of any one of claims 1 to 27, wherein the VMAT2 inhibitor is administered with or without food.
29. The method of any one of claims 1 to 28, wherein the VMAT2 inhibitor is valbenazine or a pharmaceutically acceptable salt and/or isotopic variant thereof.
30. The method of claim 29, wherein the VMAT2 inhibitor is valbenazine or a pharmaceutically acceptable salt thereof.
31. The method of claim 30, wherein the VMAT2 inhibitor is a valbenazine tosylate salt.
32. The method of claim 31, wherein the VMAT2 inhibitor is a ditosylate salt of valbenazine.
33. The method of claim 29, wherein the VMAT2 inhibitor is an isotopic variant that is L-Valine, (2R,3R,11bR)-1,3,4,6,7,11b-hexahydro-9,10-di(methoxy-d3)-3-(2-methylpropyl)-2H-benzo[a]quinolizin-2-yl ester or a pharmaceutically acceptable salt thereof.
34. The method of any one of claims 1 to 33, wherein the VMAT2 inhibitor is administered in an amount equivalent to between about 20 mg and about 160 mg of valbenazine free base.
35. The method of claim 34, wherein the VMAT2 inhibitor is administered in an amount equivalent to about 20 mg of valbenazine free base.
36. The method of claim 34, wherein the VMAT2 inhibitor is administered in an amount equivalent to about 40 mg of valbenazine free base.
37. The method of claim 34, wherein the VMAT2 inhibitor is administered in an amount equivalent to about 60 mg of valbenazine free base.
38. The method of claim 34, wherein the VMAT2 inhibitor is administered in an amount equivalent to about 80 mg of valbenazine free base.
39. The method of claim 34, wherein the VMAT2 inhibitor is administered in an amount equivalent to about 120 mg of valbenazine free base.
40. The method of any one of claims 1 to 33, wherein the VMAT2 inhibitor is administered for a first period of time in a first amount and then the amount is increased to a second amount.
41. The method of claim 38, wherein the first period of time is a week.
42. The method of claim 38 or 39, wherein the first amount is equivalent to about 40 mg of valbenazine free base.
43. The method of any one of claims 38 to 40, wherein the second amount is equivalent to about 80 mg of valbenazine free base.
44. The method of any one of claims 1 to 33, wherein the VMAT2 inhibitor is administered in an amount sufficient to achieve a maximal blood plasma concentration (Cmax) of (+)-α-DHTBZ of between about 15 ng to about 60 ng per mL plasma and a minimal blood plasma concentration (Cmin) of (+)-α-DHTBZ of at least 15 ng per mL plasma over an 8 hour period.
45. The method of any one of claims 1 to 33, wherein the VMAT2 inhibitor is administered in an amount sufficient to achieve a maximal blood plasma concentration (Cmax) of (+)-α-DHTBZ of between about 15 ng to about 60 ng per mL plasma and a minimal blood plasma concentration (Cmin) of approximately between about at least 33% -50% of the Cmax over a 12 hour period.
46. The method of any one of claims 1 to 33, wherein the VMAT2 inhibitor is administered in an amount sufficient to achieve: (i) a therapeutic concentration range of about 15 ng to about 60 ng of (+)-α-DHTBZ per mL plasma; and (ii) a threshold concentration of at least 15 ng (+)-α-DHTBZ per mL plasma over a period of about 8 hours to about 24 hours.
47. The method of any one of claims 1 to 33, wherein the therapeutically effective amount of the VMAT2 inhibitor is 10-90% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
48. The method of any one of claims 1 to 33, wherein the therapeutically effective amount of the VMAT2 inhibitor is 20-80% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
49. The method of any one of claims 1 to 33, wherein the therapeutically effective amount of the VMAT2 inhibitor is 30-70% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
50. The method of any one of claims 1 to 33, wherein the therapeutically effective amount of the VMAT2 inhibitor is 40-60% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
51. The method of any one of claims 1 to 33, wherein the therapeutically effective amount of the VMAT2 inhibitor is about 50% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
52. The method of any one of claims 1 to 28, wherein the VMAT2 inhibitor is (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof.
53. The method of claim 52, wherein the VMAT2 inhibitor is (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt thereof.
54. The method of claim 52, wherein the VMAT2 inhibitor is an isotopic variant that is (+)-α-3-isobutyl-9,10-di(methoxy-d3)-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol or a pharmaceutically acceptable salt thereof.
55. A composition for treating a patient in need of a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising the VMAT2 inhibitor, characterized in that the composition is administered in an amount equivalent to about 40 mg of valbenazine free base of the VMAT2 inhibitor once daily to the patient.
56. A composition for treating a patient in need of a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising the VMAT2 inhibitor, characterized in that the therapeutically effective amount of the VMAT2 inhibitor is less than the amount that would be administered to a patient who is not being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
57. A composition for treating a patient in need of a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, comprising the VMAT2 inhibitor,
characterized in that the composition comprising the VMAT2 inhibitor in an amount equivalent to about 40 mg of valbenazine free base is administered once daily to the patient subsequently determined to begin treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor following administration of the composition comprising a therapeutically effective amount of the VMAT2 inhibitor.
58. A composition for treating a patient in need of a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, comprising the VMAT2 inhibitor
characterized in that the composition comprising the VMAT2 inhibitor in an amount that would be less than that administered to a patient who has not begun treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor is administered to the patient subsequently determined to have begun treatment with a strong cytochrome P450 3A4 (CYP3A4) inhibitor following administration of the composition comprising a therapeutically effective amount of the VMAT2 inhibitor.
59. A composition for treating a patient in need of a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, and being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising:, comprising a therapeutically effective amount of the VMAT2 inhibitor,
wherein administration of the composition produces a mean valbenazine Cmax that is about 1 to about 2 fold higher than the mean valbenazine Cmax for a patient who is not being administered a strong CYP3A4 inhibitor and/or a mean valbenazine AUC0-∞ that is about 1.5 to about 2.5 fold higher than the mean valbenazine AUC0-∞ for a patient who is not being administered a strong CYP3A4 inhibitor.
60. A composition for treating a patient in need of a vesicular monoamine transport 2 (VMAT2) inhibitor chosen from valbenazine and (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof, and being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor, comprising: comprising a therapeutically effective amount of the VMAT2 inhibitor,
wherein administration of the composition produces a mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol Cmax that is about 1 to about 2 fold higher than the mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol Cmax for a patient who is not being administered a strong CYP3A4 inhibitor and/or a mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol AUC0-∞ that is about 1.5 to about 2.5 fold higher than the mean (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol AUC0-∞ for a patient who is not being administered a strong CYP3A4 inhibitor.
61. The composition of any one of claims 55 to 60, wherein the patient or a medical care worker is informed that administration of the composition to patients who is also being administered a strong CYP3A4 inhibitor in higher exposure of valbenazine and/or (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol than administration of the composition to a patient who is not being administered a strong CYP3A4 inhibitor.
62. The composition of any one of claims 55 to 61, wherein the patient or a medical care worker is informed that administration of the composition to a patient who is also being administered a strong CYP3A4 inhibitor may result in increased risk of one or more exposure-related adverse reactions than administration of the composition to a patient who is not being administered a strong CYP3A4 inhibitor.
63. The composition of claim 62, wherein the one or more exposure-related adverse reactions is chosen from somnolence, anticholinergic effects, balance disorders or falls, headache, akathisia, vomiting, nausea, arthralgia, QT prolongation, increase in blood glucose, increase in weight, respiratory infections, drooling, dyskinesia, extrapyramidal symptoms (non-akathisia), anxiety, insomnia, increase in prolactin, increase in alkaline phosphatase, and increase in bilirubin.
64. The composition of claim 63, wherein the one or more exposure-related adverse reactions is chosen from somnolence, anticholinergic effects, balance disorders or falls, headache, akathisia, vomiting, nausea, arthralgia, and QT prolongation.
65. The composition of claim 64, wherein the one or more exposure-related adverse reactions is chosen from somnolence and QT prolongation.
66. The composition of any one of claims 55 to 65, wherein the patient or a medical care worker is informed that co-administration of the composition and the CYP3A4 inhibitor may prolong the patient's QT interval.
67. The composition of any one of claims 55 to 66, wherein the patient or a medical care worker is informed that administration of the composition to a patient who is also being administered a strong CYP3A4 inhibitor may prolong the patient's QT interval than administration of the composition to a patient who is also being administered a strong CYP3A4 inhibitor.
68. The composition of any one of claims 55 to 67, wherein the strong CYP3A4 inhibitor is chosen from clarithromycin, chloramphenicol, cobicistat, indinavir, itraconazole, ketoconazole, nefazodone, nelfinavir, ritonavir, saquinavir, and telithromycin.
69. The composition of claim 68, wherein the strong CYP3A4 inhibitor is chosen from clarithromycin, itraconazole, and ketoconazole.
70. The composition of claim 69, wherein the strong CYP3A4 inhibitor is ketoconazole.
71. The composition of any one of claims 55 to 70, wherein the composition is for treating a neurological or psychiatric disease or disorder.
72. The composition of claim 71, wherein the neurological or psychiatric disease or disorder is a hyperkinetic movement disorder, mood disorder, bipolar disorder, schizophrenia, schizoaffective disorder, mania in mood disorder, depression in mood disorder, treatment-refractory obsessive compulsive disorder, neurological dysfunction associated with Lesch-Nyhan syndrome, agitation associated with Alzheimer's disease, Fragile X syndrome or Fragile X-associated tremor-ataxia syndrome, autism spectrum disorder, Rett syndrome, or chorea-acanthocytosis.
73. The composition of claim 72, wherein the neurological or psychiatric disease or disorder is a hyperkinetic movement disorder.
74. The composition of claim 73, wherein the hyperkinetic movement disorder is tardive dyskinesia.
75. The composition of claim 73, wherein the hyperkinetic movement disorder is Tourette's syndrome.
76. The composition of claim 73, wherein the hyperkinetic movement disorder is Huntington's disease.
77. The composition of claim 73, wherein the hyperkinetic movement disorder is tics.
78. The composition of claim 73, wherein the hyperkinetic movement disorder is chorea associated with Huntington's disease.
79. The composition of claim 73, wherein the hyperkinetic movement disorder is ataxia, chorea, dystonia, Huntington's disease, myoclonus, restless leg syndrome, or tremors.
80. The composition of any one of claims 55 to 79, characterized in that the composition is administered orally.
81. The composition of any one of claims 55 to 80, characterized in that the composition is administered in the form of a tablet or capsule.
82. The composition of any one of claims 55 to 81, characterized in that the composition is administered with or without food.
83. The composition of any one of claims 55 to 82, wherein the VMAT2 inhibitor is valbenazine or a pharmaceutically acceptable salt and/or isotopic variant thereof.
84. The composition of claim 83, wherein the VMAT2 inhibitor is valbenazine or a pharmaceutically acceptable salt thereof.
85. The composition of claim 84, wherein the VMAT2 inhibitor is a valbenazine tosylate salt.
86. The composition of claim 85, wherein the VMAT2 inhibitor is a ditosylate salt of valbenazine.
87. The composition of claim 83, wherein the VMAT2 inhibitor is an isotopic variant that is L-Valine, (2R, 3R,11bR)-1,3,4,6,7,11b-hexahydro-9,10-di(methoxy-d3)-3-(2-methylpropyl)-2H-benzo[a]quinolizin-2-yl ester or a pharmaceutically acceptable salt thereof.
88. The composition of any one of claims 55 to 87, characterized in that the composition is administered in an amount equivalent to between about 20 mg and about 160 mg of valbenazine free base of the VMAT2 inhibitor.
89. The composition of claim 88, characterized in that the composition is administered in an amount equivalent to about 20 mg of valbenazine free base of the VMAT2 inhibitor.
90. The composition of claim 88, characterized in that the composition is administered in an amount equivalent to about 40 mg of valbenazine free base of the VMAT2 inhibitor.
91. The composition of claim 88, characterized in that the composition is administered in an amount equivalent to about 60 mg of valbenazine free base of the VMAT2 inhibitor.
92. The composition of claim 88, characterized in that the composition is administered in an amount equivalent to about 80 mg of valbenazine free base of the VMAT2 inhibitor.
93. The composition of claim 88, characterized in that the composition is administered in an amount equivalent to about 120 mg of valbenazine free base of the VMAT2 inhibitor.
94. The composition of any one of claims 55 to 87, characterized in that the composition is administered for a first period of time in a first amount of the VMAT2 inhibitor and then the amount is increased to a second amount.
95. The composition of claim 94, wherein the first period of time is a week.
96. The composition of claim 94 or 95, wherein the first amount is equivalent to about 40 mg of valbenazine free base.
97. The composition of any one of claims 94 to 96, wherein the second amount is equivalent to about 80 mg of valbenazine free base.
98. The composition of any one of claims 55 to 87, characterized in that the composition is administered in an amount sufficient to achieve a maximal blood plasma concentration (Cmax) of (+)-α-DHTBZ of between about 15 ng to about 60 ng per mL plasma and a minimal blood plasma concentration (Cmin) of (+)-α-DHTBZ of at least 15 ng per mL plasma over an 8 hour period.
99. The composition of any one of claims 55 to 87, characterized in that the composition is administered in an amount sufficient to achieve a maximal blood plasma concentration (Cmax) of (+)-α-DHTBZ of between about 15 ng to about 60 ng per mL plasma and a minimal blood plasma concentration (Cmin) of approximately between about at least 33% -50% of the Cmax over a 12 hour period.
100. The composition of any one of claims 55 to 87, characterized in that the composition is administered in an amount sufficient to achieve: (i) a therapeutic concentration range of about 15 ng to about 60 ng of (+)-α-DHTBZ per mL plasma; and (ii) a threshold concentration of at least 15 ng (+)-α-DHTBZ per mL plasma over a period of about 8 hours to about 24 hours.
101. The composition of any one of claims 55 to 87, characterized in that the therapeutically effective amount of the VMAT2 inhibitor is 10-90% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
102. The composition of any one of claims 55 to 87, characterized in that the therapeutically effective amount of the VMAT2 inhibitor is 20-80% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
103. The composition of any one of claims 55 to 87, characterized in that the therapeutically effective amount of the VMAT2 inhibitor is 30-70% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
104. The composition of any one of claims 55 to 87, characterized in that the therapeutically effective amount of the VMAT2 inhibitor is 40-60% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
105. The composition of any one of claims 55 to 87, characterized in that the therapeutically effective amount of the VMAT2 inhibitor is about 50% less than the amount that would be administered to a patient who is not also being administered a strong cytochrome P450 3A4 (CYP3A4) inhibitor.
106. The composition of any one of claims 55 to 82, wherein the VMAT2 inhibitor is (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt and/or isotopic variant thereof.
107. The composition of claim 106, wherein the VMAT2 inhibitor is (+)-α-3-isobutyl-9,10-dimethoxy-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol, or a pharmaceutically acceptable salt thereof.
108. The composition of claim 106, wherein the VMAT2 inhibitor is an isotopic variant that is (+)-α-3-isobutyl-9,10-di(methoxy-d3)-1,3,4,6,7,11b-hexahydro-2H-pyrido[2,1-a]isoquinolin-2-ol or a pharmaceutically acceptable salt thereof.
US16/481,037 2017-01-27 2017-10-10 Methods for the Administration of Certain VMAT2 Inhibitors Abandoned US20200093808A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/481,037 US20200093808A1 (en) 2017-01-27 2017-10-10 Methods for the Administration of Certain VMAT2 Inhibitors

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201762451605P 2017-01-27 2017-01-27
US16/481,037 US20200093808A1 (en) 2017-01-27 2017-10-10 Methods for the Administration of Certain VMAT2 Inhibitors
PCT/US2017/055931 WO2018140094A1 (en) 2017-01-27 2017-10-10 Methods for the administration of certain vmat2 inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/055931 A-371-Of-International WO2018140094A1 (en) 2017-01-27 2017-10-10 Methods for the administration of certain vmat2 inhibitors

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/870,423 Continuation US10952997B2 (en) 2017-01-27 2020-05-08 Methods for the administration of certain VMAT2 inhibitors

Publications (1)

Publication Number Publication Date
US20200093808A1 true US20200093808A1 (en) 2020-03-26

Family

ID=62978647

Family Applications (9)

Application Number Title Priority Date Filing Date
US16/481,037 Abandoned US20200093808A1 (en) 2017-01-27 2017-10-10 Methods for the Administration of Certain VMAT2 Inhibitors
US16/481,029 Abandoned US20190381016A1 (en) 2017-01-27 2017-10-10 Methods for the Administration of Certain VMAT2 Inhibitors
US16/481,033 Active 2037-11-24 US10874648B2 (en) 2017-01-27 2017-10-10 Methods for the administration of certain VMAT2 inhibitors
US16/870,572 Active US11040029B2 (en) 2017-01-27 2020-05-08 Methods for the administration of certain VMAT2 inhibitors
US16/870,423 Active US10952997B2 (en) 2017-01-27 2020-05-08 Methods for the administration of certain VMAT2 inhibitors
US16/870,706 Active US10857137B2 (en) 2017-01-27 2020-05-08 Methods for the administration of certain VMAT2 inhibitors
US17/073,304 Active US10912771B1 (en) 2017-01-27 2020-10-17 Methods for the administration of certain VMAT2 inhibitors
US17/080,343 Active US11439629B2 (en) 2017-01-27 2020-10-26 Methods for the administration of certain VMAT2 inhibitors
US17/125,190 Abandoned US20210169862A1 (en) 2017-01-27 2020-12-17 Methods for the Administration of Certain VMAT2 Inhibitors

Family Applications After (8)

Application Number Title Priority Date Filing Date
US16/481,029 Abandoned US20190381016A1 (en) 2017-01-27 2017-10-10 Methods for the Administration of Certain VMAT2 Inhibitors
US16/481,033 Active 2037-11-24 US10874648B2 (en) 2017-01-27 2017-10-10 Methods for the administration of certain VMAT2 inhibitors
US16/870,572 Active US11040029B2 (en) 2017-01-27 2020-05-08 Methods for the administration of certain VMAT2 inhibitors
US16/870,423 Active US10952997B2 (en) 2017-01-27 2020-05-08 Methods for the administration of certain VMAT2 inhibitors
US16/870,706 Active US10857137B2 (en) 2017-01-27 2020-05-08 Methods for the administration of certain VMAT2 inhibitors
US17/073,304 Active US10912771B1 (en) 2017-01-27 2020-10-17 Methods for the administration of certain VMAT2 inhibitors
US17/080,343 Active US11439629B2 (en) 2017-01-27 2020-10-26 Methods for the administration of certain VMAT2 inhibitors
US17/125,190 Abandoned US20210169862A1 (en) 2017-01-27 2020-12-17 Methods for the Administration of Certain VMAT2 Inhibitors

Country Status (12)

Country Link
US (9) US20200093808A1 (en)
JP (11) JP7199359B2 (en)
KR (10) KR20230170135A (en)
CN (6) CN116712434A (en)
AU (7) AU2017395700B2 (en)
CA (5) CA3051832A1 (en)
EA (5) EA201991784A1 (en)
MX (6) MX2019008853A (en)
MY (2) MY195934A (en)
PH (5) PH12019501732A1 (en)
SG (3) SG11201906883SA (en)
WO (5) WO2018140094A1 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10844058B2 (en) 2015-10-30 2020-11-24 Neurocrine Biosciences, Inc. Valbenazine salts and polymorphs thereof
US10857148B2 (en) 2017-10-10 2020-12-08 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
US10857137B2 (en) 2017-01-27 2020-12-08 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
US10906903B2 (en) 2015-12-23 2021-02-02 Neurocrine Biosciences, Inc. Synthetic methods for preparation of (S)-(2R,3R,11bR)-3-isobutyl-9,10-dimethoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1,-a]isoquinolin-2-yl 2-amino-3-methylbutanoate di(4-methylbenzenesulfonate)
US10940141B1 (en) 2019-08-23 2021-03-09 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
US10993941B2 (en) 2017-10-10 2021-05-04 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
US11026939B2 (en) 2017-09-21 2021-06-08 Neurocrine Biosciences, Inc. High dosage valbenazine formulation and compositions, methods, and kits related thereto
US11026931B2 (en) 2018-08-15 2021-06-08 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB201705304D0 (en) 2017-04-01 2017-05-17 Adeptio Pharmaceuticals Ltd Pharmaceutical compositions
GB201705303D0 (en) 2017-04-01 2017-05-17 Adeptio Pharmaceuticals Ltd Pharmaceutical compositions
GB201705302D0 (en) 2017-04-01 2017-05-17 Adeptio Pharmaceuticals Ltd Pharmaceutical compositions
GB201705306D0 (en) 2017-04-01 2017-05-17 Adeptio Pharmaceuticals Ltd Pharmaceutical compositions
JP7212958B2 (en) 2017-12-26 2023-01-26 ▲蘇▼州科睿思制▲葯▼有限公司 Crystal form of valbenadine tosylate, method for producing the same, and use thereof
AU2020270145A1 (en) * 2019-05-09 2021-12-23 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
KR20240027750A (en) 2021-06-30 2024-03-04 뉴로크린 바이오사이언시즈 인코퍼레이티드 Valbenazine for the treatment of dyskinesias due to cerebral palsy
WO2023278987A1 (en) 2021-06-30 2023-01-05 Neurocrine Biosciences, Inc. Valbenazine for use in the add-on treatment of schizophrenia
TW202337465A (en) * 2021-08-16 2023-10-01 逸達生物科技股份有限公司 Polymer depot compositions for sustained release delivery of vmat2 inhibitors
TW202322816A (en) 2021-08-20 2023-06-16 美商紐羅克里生物科學有限公司 Methods of screening for vmat2 inhibitors
WO2023039187A1 (en) * 2021-09-10 2023-03-16 ATAI Life Sciences AG Ibogaine combination treatment
WO2023076568A1 (en) * 2021-10-29 2023-05-04 Neurocrine Biosciences, Inc. Valbenazine compositions
WO2023172849A1 (en) 2022-03-07 2023-09-14 Neurocrine Biosciences, Inc. Valbenazine, a vmat2 inhibitor, as a free base a tosylate or ditosylate salt, for use in the treatment of chorea associated with huntington's disease

Family Cites Families (118)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1068938A (en) 1910-11-07 1913-07-29 Gustav F Schulze Vehicle-apron.
US3536809A (en) 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
JPS5283918A (en) 1976-01-01 1977-07-13 Wellcome Found Pharmaceutical composition
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
JPS5777697A (en) 1980-11-04 1982-05-15 Nippon Zoki Pharmaceut Co Ltd Physiologically active substance nsq
US4410545A (en) 1981-02-13 1983-10-18 Syntex (U.S.A.) Inc. Carbonate diester solutions of PGE-type compounds
US4328245A (en) 1981-02-13 1982-05-04 Syntex (U.S.A.) Inc. Carbonate diester solutions of PGE-type compounds
JPS57209225A (en) 1981-06-18 1982-12-22 Yamasa Shoyu Co Ltd Antitumor activity intensifier
US4409239A (en) 1982-01-21 1983-10-11 Syntex (U.S.A.) Inc. Propylene glycol diester solutions of PGE-type compounds
JPS6028987A (en) 1983-07-28 1985-02-14 Chugai Pharmaceut Co Ltd Novel digoxin derivative
KR890002631B1 (en) 1984-10-04 1989-07-21 몬산토 캄파니 Composition of prolonged release of biologically active somatotropin
IE58110B1 (en) 1984-10-30 1993-07-14 Elan Corp Plc Controlled release powder and process for its preparation
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
US5612059A (en) 1988-08-30 1997-03-18 Pfizer Inc. Use of asymmetric membranes in delivery devices
IT1229203B (en) 1989-03-22 1991-07-25 Bioresearch Spa USE OF 5 METHYLTHETRAHYDROPHOLIC ACID, 5 FORMYLTHETRAHYDROPHOLIC ACID AND THEIR PHARMACEUTICALLY ACCEPTABLE SALTS FOR THE PREPARATION OF PHARMACEUTICAL COMPOSITIONS IN THE FORM OF CONTROLLED RELEASE ACTIVE IN THE THERAPY OF MENTAL AND ORGANIC DISORDERS.
PH30995A (en) 1989-07-07 1997-12-23 Novartis Inc Sustained release formulations of water soluble peptides.
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US5585112A (en) 1989-12-22 1996-12-17 Imarx Pharmaceutical Corp. Method of preparing gas and gaseous precursor-filled microspheres
IT1246382B (en) 1990-04-17 1994-11-18 Eurand Int METHOD FOR THE TARGETED AND CONTROLLED DELIVERY OF DRUGS IN THE INTESTINE AND PARTICULARLY IN THE COLON
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5204329A (en) 1990-06-11 1993-04-20 Du Pont Merck Pharmaceutical Company Treatment of organ transplantation rejection
US5543390A (en) 1990-11-01 1996-08-06 State Of Oregon, Acting By And Through The Oregon State Board Of Higher Education, Acting For And On Behalf Of The Oregon Health Sciences University Covalent microparticle-drug conjugates for biological targeting
US5580578A (en) 1992-01-27 1996-12-03 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
TW333456B (en) 1992-12-07 1998-06-11 Takeda Pharm Ind Co Ltd A pharmaceutical composition of sustained-release preparation the invention relates to a pharmaceutical composition of sustained-release preparation which comprises a physiologically active peptide.
US5591767A (en) 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US6274552B1 (en) 1993-03-18 2001-08-14 Cytimmune Sciences, Inc. Composition and method for delivery of biologically-active factors
US5985307A (en) 1993-04-14 1999-11-16 Emory University Device and method for non-occlusive localized drug delivery
US5523092A (en) 1993-04-14 1996-06-04 Emory University Device for local drug delivery and methods for using the same
US6087324A (en) 1993-06-24 2000-07-11 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6004534A (en) 1993-07-23 1999-12-21 Massachusetts Institute Of Technology Targeted polymerized liposomes for improved drug delivery
IT1270594B (en) 1994-07-07 1997-05-07 Recordati Chem Pharm CONTROLLED RELEASE PHARMACEUTICAL COMPOSITION OF LIQUID SUSPENSION MOGUISTEIN
US5759542A (en) 1994-08-05 1998-06-02 New England Deaconess Hospital Corporation Compositions and methods for the delivery of drugs by platelets for the treatment of cardiovascular and other diseases
US5660854A (en) 1994-11-28 1997-08-26 Haynes; Duncan H Drug releasing surgical implant or dressing material
US6316652B1 (en) 1995-06-06 2001-11-13 Kosta Steliou Drug mitochondrial targeting agents
US5798119A (en) 1995-06-13 1998-08-25 S. C. Johnson & Son, Inc. Osmotic-delivery devices having vapor-permeable coatings
AU6242096A (en) 1995-06-27 1997-01-30 Takeda Chemical Industries Ltd. Method of producing sustained-release preparation
TW448055B (en) 1995-09-04 2001-08-01 Takeda Chemical Industries Ltd Method of production of sustained-release preparation
JP2909418B2 (en) 1995-09-18 1999-06-23 株式会社資生堂 Delayed release microsphere of drug
US6039975A (en) 1995-10-17 2000-03-21 Hoffman-La Roche Inc. Colon targeted delivery system
US5980945A (en) 1996-01-16 1999-11-09 Societe De Conseils De Recherches Et D'applications Scientifique S.A. Sustained release drug formulations
TW345603B (en) 1996-05-29 1998-11-21 Gmundner Fertigteile Gmbh A noise control device for tracks
US6264970B1 (en) 1996-06-26 2001-07-24 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6419961B1 (en) 1996-08-29 2002-07-16 Takeda Chemical Industries, Ltd. Sustained release microcapsules of a bioactive substance and a biodegradable polymer
ZA977967B (en) 1996-09-23 1999-03-04 Lilly Co Eli Combination therapy for treatment of psychoses
KR100353304B1 (en) 1996-10-01 2002-09-30 에스알아이 인터내셔널 Taste-masked microcapsule compositions and methods of manufacture
CA2217134A1 (en) 1996-10-09 1998-04-09 Sumitomo Pharmaceuticals Co., Ltd. Sustained release formulation
EP0839525B1 (en) 1996-10-31 2004-08-04 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6131570A (en) 1998-06-30 2000-10-17 Aradigm Corporation Temperature controlling device for aerosol drug delivery
EP0946169B1 (en) 1996-12-20 2003-02-26 Takeda Chemical Industries, Ltd. Method of producing a sustained-release preparation
US5891474A (en) 1997-01-29 1999-04-06 Poli Industria Chimica, S.P.A. Time-specific controlled release dosage formulations and method of preparing same
US6120751A (en) 1997-03-21 2000-09-19 Imarx Pharmaceutical Corp. Charged lipids and uses for the same
US6060082A (en) 1997-04-18 2000-05-09 Massachusetts Institute Of Technology Polymerized liposomes targeted to M cells and useful for oral or mucosal drug delivery
US6350458B1 (en) 1998-02-10 2002-02-26 Generex Pharmaceuticals Incorporated Mixed micellar drug deliver system and method of preparation
US6613358B2 (en) 1998-03-18 2003-09-02 Theodore W. Randolph Sustained-release composition including amorphous polymer
US6048736A (en) 1998-04-29 2000-04-11 Kosak; Kenneth M. Cyclodextrin polymers for carrying and releasing drugs
KR19990085365A (en) 1998-05-16 1999-12-06 허영섭 Biodegradable polymer microspheres capable of continuously controlled controlled release and preparation method thereof
WO2000024399A1 (en) 1998-10-23 2000-05-04 Sepracor Inc. Compositions and methods employing r(-) fluoxetine and other active ingredients
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6271359B1 (en) 1999-04-14 2001-08-07 Musc Foundation For Research Development Tissue-specific and pathogen-specific toxic agents and ribozymes
AP2001002264A0 (en) 2000-08-30 2001-09-30 Pfizer Prod Inc Sustained release formulations for growth hormone secretagogues.
EP1513403B1 (en) 2002-05-28 2017-02-15 Bette Pollard Cardiac glycosides to treat cystic fibrosis and other il-8 dependent disorders
WO2004026258A2 (en) 2002-09-19 2004-04-01 University Of Utah Research Foundation Modulating vesicular monoamine transporter trafficking and function: a novel approach for the treatment of parkinson’s disease
US7230097B2 (en) 2003-03-10 2007-06-12 Lupin Ltd. Process for preparation of 7-[α-Amino (4-hydroxyphenyl) acetamido]-3-substituted-3-cephem-4-carboxylic acid
GB0307259D0 (en) 2003-03-28 2003-05-07 Glaxo Group Ltd Process
GB2410947B (en) 2004-02-11 2008-09-17 Cambridge Lab Ltd Pharmaceutical compounds
MXPA06014069A (en) 2004-06-04 2007-04-25 Genentech Inc Method for treating multiple sclerosis.
WO2006039663A2 (en) 2004-09-30 2006-04-13 Vanda Pharmaceuticals, Inc Methods for the administration of iloperidone
CN101039679A (en) 2004-10-28 2007-09-19 惠氏公司 Co-administration of tigecycline and digoxin
WO2006053067A2 (en) 2004-11-09 2006-05-18 Prestwick Pharmaceuticals, Inc. Combination of amantadine and a tetrabenazine compound for treating hyperkinetic disorders
US20110257220A1 (en) 2005-08-06 2011-10-20 Cambridge Laboratories (Ireland) Limited 3,11 b-cis-Dihydrotetrabenazine for the Treatment of Schizophrenia and Other Psychoses
US20100076087A1 (en) 2005-10-06 2010-03-25 Auspex Pharmaceuticals, Inc. Methods of reduction of interpatient variability
WO2008058261A1 (en) 2006-11-08 2008-05-15 Neurocrine Biosciences, Inc. Substituted 3-isobutyl-9, 10-dimethoxy-1,3,4,6,7,11b-hexahydro-2h-pyrido[2,1-a] isoquinolin-2-ol compounds and methods relating thereto
GB0721669D0 (en) 2007-11-02 2007-12-12 Cambridge Lab Ireland Ltd Pharmaceutical compounds
UA101352C2 (en) 2008-01-18 2013-03-25 Ейсей Р Енд Д Менеджмент Ко., Лтд. Normal;heading 1;heading 2;heading 3;FUSED AMINODIHYDROTHIAZINE DERIVATIVE
US20110053866A1 (en) 2008-08-12 2011-03-03 Biovail Laboratories International (Barbados) S.R.L. Pharmaceutical compositions
GB2462611A (en) 2008-08-12 2010-02-17 Cambridge Lab Pharmaceutical composition comprising tetrabenazine
GB2463452A (en) 2008-09-08 2010-03-17 Cambridge Lab Desmethyl derivatives of tetrabenazine and pharmaceutical compositions thereof
GB2463451A (en) 2008-09-08 2010-03-17 Cambridge Lab 3, 11b cis-dihydrotetrabenazine compounds for use in the treatment of dementia
NO3061760T3 (en) 2008-09-18 2018-04-07
US20100096319A1 (en) 2008-10-17 2010-04-22 General Electric Company Separator assembly
HUE030667T2 (en) * 2009-04-06 2017-05-29 Vanda Pharmaceuticals Inc Method of treatment based on polymorphisms of the kcnq1 gene
JP2013501810A (en) 2009-08-12 2013-01-17 ヴァリーント インターナショナル(バルバドス)エスアールエル Pharmaceutical composition
EP2576552A4 (en) 2010-06-01 2013-11-13 Aupex Pharmaceutical Inc Benzoquinolone inhibitors of vmat2
WO2012153341A1 (en) 2011-05-12 2012-11-15 Arch Pharmalabs Limited A process for the preparation of iloperidone and pharmaceutically acceptable salts thereof
WO2014047167A1 (en) 2012-09-18 2014-03-27 Auspex Pharmaceuticals, Inc. Formulations pharmacokinetics of deuterated benzoquinoline inhibitors of vesicular monoamine transporter 2
ES2805376T3 (en) 2012-12-18 2021-02-11 Vanda Pharmaceuticals Inc Tasimelteon for treating circadian rhythm disorders
EA201500801A1 (en) 2013-01-31 2016-01-29 Ауспекс Фармацетикалс, Инк. BENZOHINOLONE INHIBITORS VMAT2
WO2015077521A1 (en) * 2013-11-22 2015-05-28 Auspex Pharmaceuticals, Inc. Benzoquinoline inhibitors of vesicular monoamine transporter 2
WO2015112707A1 (en) 2014-01-27 2015-07-30 Auspex Pharmaceuticals, Inc. Benzoquinoline inhibitors of vesicular monoamine transporter 2
RU2021128797A (en) 2014-02-07 2021-11-26 Ньюрокрайн Байосайенсиз, Инк. PHARMACEUTICAL COMPOSITIONS CONTAINING ANTIPSYCHOTIC DRUG AND VMAT2 INHIBITOR AND THEIR APPLICATION
KR20160117596A (en) 2014-02-07 2016-10-10 오스펙스 파마슈티칼스, 인코포레이티드 Novel pharmaceutical formulations
KR20200133003A (en) 2014-05-06 2020-11-25 뉴로크린 바이오사이언시즈 인코퍼레이티드 Vmat2 inhibitors for the treatment of hyperkinetic movement disorders
WO2016127133A1 (en) 2015-02-06 2016-08-11 Neurocrine Biosciences, Inc. [9,10-dimethoxy-3-(2-methylpropyl)-1h,2h,3h,4h,6h,7h,11bh-pyrido-[2,1-a]isoquinolin-2-yl]methanol and compounds, compositions and methods relating thereto
IL305352A (en) 2015-03-06 2023-10-01 Auspex Pharmaceuticals Inc Methods for the treatment of abnormal involuntary movement disorders
MX2017017015A (en) * 2015-06-23 2018-02-26 Neurocrine Biosciences Inc Vmat2 inhibitors for treating neurological diseases or disorders.
CN115322188A (en) 2015-10-30 2022-11-11 纽罗克里生物科学有限公司 VALBENAZINE salts and polymorphs thereof
CN108925135A (en) 2015-12-23 2018-11-30 纽罗克里生物科学有限公司 Prepare (S)-(2R, 3R, 11bR) -3- isobutyl group -9,10- dimethoxy -2,3,4,6,7,11b- hexahydro -1H- pyrido [2,1-a] isoquinolin-2-yl 2- amino -3 Methylbutanoic acid ester two (4- toluenesulfonate) synthetic method
TW202345829A (en) 2016-12-02 2023-12-01 美商紐羅克里生物科學有限公司 Use of valbenazine for treating schizophrenia or schizoaffective disorder
MX2019008853A (en) 2017-01-27 2019-09-11 Neurocrine Biosciences Inc Methods for the administration of certain vmat2 inhibitors.
WO2018164996A1 (en) 2017-03-06 2018-09-13 Neurocrine Biosciences, Inc. Dosing regimen for valbenazine
US20180280374A1 (en) 2017-04-01 2018-10-04 Adeptio Pharmaceuticals Limited Pharmaceutical compositions
JP7250692B2 (en) 2017-04-01 2023-04-03 アデプティオ ファーマシューティカルズ リミテッド Dihydrotetrabenazine for use in treating movement disorders
JOP20190239A1 (en) 2017-04-19 2019-10-09 Neurocrine Biosciences Inc Vmat2 inhibitor compounds and compositions thereof
WO2018200605A1 (en) 2017-04-26 2018-11-01 Neurocrine Biosciences, Inc. Use of valbenazine for treating levodopa-induced dyskinesia
DK3426250T3 (en) * 2017-05-16 2022-05-16 Bow River LLC TREATMENT OF A PATIENT WITH A CYP3A4 SUBSTRATED MEDICINAL PRODUCT CONTRAINDICATED FOR ADMINISTRATION WITH A STRONG CYP3A4 INHIBITOR
US10857144B2 (en) * 2017-05-16 2020-12-08 Bow River LLC Methods of treatment
MX2020002841A (en) 2017-09-21 2020-07-22 Neurocrine Biosciences Inc High dosage valbenazine formulation and compositions, methods, and kits related thereto.
US20200230127A1 (en) 2017-10-10 2020-07-23 Neurocrine Biosciences, Inc. Methods for the Administration of Certain VMAT2 Inhibitors
US10993941B2 (en) 2017-10-10 2021-05-04 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
WO2019104141A1 (en) 2017-11-22 2019-05-31 Teva Pharmaceuticals Usa, Inc. Solid state form of valbenazine
WO2019241555A1 (en) 2018-06-14 2019-12-19 Neurocrine Biosciences, Inc. Vmat2 inhibitor compounds, compositions, and methods relating thereto
CN112867491A (en) 2018-08-15 2021-05-28 纽罗克里生物科学有限公司 Methods of administering certain VMAT2 inhibitors
AU2020270145A1 (en) 2019-05-09 2021-12-23 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
US10689380B1 (en) 2019-07-30 2020-06-23 Farmhispania S.A. Crystalline forms of valbenazine ditosylate
US10940141B1 (en) 2019-08-23 2021-03-09 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10851103B2 (en) 2015-10-30 2020-12-01 Neurocrine Biosciences, Inc. Valbenazine salts and polymorphs thereof
US10851104B2 (en) 2015-10-30 2020-12-01 Neurocrine Biosciences, Inc. Valbenazine salts and polymorphs thereof
US10844058B2 (en) 2015-10-30 2020-11-24 Neurocrine Biosciences, Inc. Valbenazine salts and polymorphs thereof
US10906903B2 (en) 2015-12-23 2021-02-02 Neurocrine Biosciences, Inc. Synthetic methods for preparation of (S)-(2R,3R,11bR)-3-isobutyl-9,10-dimethoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1,-a]isoquinolin-2-yl 2-amino-3-methylbutanoate di(4-methylbenzenesulfonate)
US10919892B2 (en) 2015-12-23 2021-02-16 Neurocrine Biosciences, Inc. Synthetic methods for preparation of (S)-(2R,3R,11bR)-3-isobutyl-9,10-dimethoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1-a]isoquinolin-2-yl 2-amino-3-methylbutanoate di(4-methylbenzenesulfonate)
US10906902B2 (en) 2015-12-23 2021-02-02 Neurocrine Biosciences, Inc. Synthetic methods for preparation of (S)-(2R,3R,11bR)-3-isobutyl-9,10-dimethoxy-2,3,4,6,7,11b-hexahydro-1H-pyrido[2,1,-a]isoquinolin-2-2-amino-3-methylbutanoate di(4-methylbenzenesulfonate)
US10912771B1 (en) 2017-01-27 2021-02-09 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
US10874648B2 (en) 2017-01-27 2020-12-29 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
US10857137B2 (en) 2017-01-27 2020-12-08 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
US10952997B2 (en) 2017-01-27 2021-03-23 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
US11040029B2 (en) 2017-01-27 2021-06-22 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
US11439629B2 (en) 2017-01-27 2022-09-13 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
US11026939B2 (en) 2017-09-21 2021-06-08 Neurocrine Biosciences, Inc. High dosage valbenazine formulation and compositions, methods, and kits related thereto
US11311532B2 (en) 2017-09-21 2022-04-26 Neurocrine Biosciences, Inc. High dosage valbenazine formulation and compositions, methods, and kits related thereto
US10857148B2 (en) 2017-10-10 2020-12-08 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
US10993941B2 (en) 2017-10-10 2021-05-04 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
US11654142B2 (en) 2017-10-10 2023-05-23 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
US11026931B2 (en) 2018-08-15 2021-06-08 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors
US10940141B1 (en) 2019-08-23 2021-03-09 Neurocrine Biosciences, Inc. Methods for the administration of certain VMAT2 inhibitors

Also Published As

Publication number Publication date
EA201991786A1 (en) 2020-01-16
PH12020550238A1 (en) 2021-02-15
US11439629B2 (en) 2022-09-13
US20190381016A1 (en) 2019-12-19
KR20230169457A (en) 2023-12-15
SG11201906891RA (en) 2019-08-27
CA3051832A1 (en) 2018-08-02
MX2019008868A (en) 2019-09-16
KR20200066662A (en) 2020-06-10
CA3051834A1 (en) 2018-08-02
US20200268725A1 (en) 2020-08-27
MX2019008855A (en) 2019-09-11
MY195934A (en) 2023-02-27
EA201991784A1 (en) 2021-09-23
AU2022203327A1 (en) 2022-06-09
EA201991780A1 (en) 2021-10-26
PH12019501732A1 (en) 2020-03-16
AU2017395700B2 (en) 2022-02-24
AU2017395701A1 (en) 2020-06-18
PH12019501740A1 (en) 2019-10-07
JP2022174343A (en) 2022-11-22
CN110769826A (en) 2020-02-07
JP2020514318A (en) 2020-05-21
US20200268743A1 (en) 2020-08-27
WO2018140096A1 (en) 2018-08-02
KR20190108149A (en) 2019-09-23
US11040029B2 (en) 2021-06-22
PH12019501734A1 (en) 2020-07-06
WO2018140093A1 (en) 2018-08-02
US20210046060A1 (en) 2021-02-18
SG11201906883SA (en) 2019-08-27
KR20190108146A (en) 2019-09-23
JP2023161033A (en) 2023-11-02
SG11201906885TA (en) 2019-08-27
CA3051829A1 (en) 2018-08-02
AU2017395702B2 (en) 2021-09-09
CA3051830A1 (en) 2018-08-02
AU2017395704A1 (en) 2019-08-22
MX2019008854A (en) 2019-09-11
CN110709071A (en) 2020-01-17
CA3078719A1 (en) 2018-08-02
JP7199361B2 (en) 2023-01-05
US10857137B2 (en) 2020-12-08
US20200338066A1 (en) 2020-10-29
AU2022203201A1 (en) 2022-06-02
AU2017395702A1 (en) 2019-08-22
AU2022203201B2 (en) 2023-11-02
JP2020514317A (en) 2020-05-21
JP7107950B2 (en) 2022-07-27
JP2020505408A (en) 2020-02-20
JP2022174344A (en) 2022-11-22
KR20190108147A (en) 2019-09-23
MY191077A (en) 2022-05-30
CN116712434A (en) 2023-09-08
JP2022137219A (en) 2022-09-21
US20200268724A1 (en) 2020-08-27
WO2018140094A1 (en) 2018-08-02
US20210169862A1 (en) 2021-06-10
WO2018140092A1 (en) 2018-08-02
AU2017395701B2 (en) 2024-02-15
AU2017395704B2 (en) 2022-02-24
KR20190108148A (en) 2019-09-23
JP7090151B2 (en) 2022-06-23
KR20230170135A (en) 2023-12-18
KR20230170136A (en) 2023-12-18
CN110740732A (en) 2020-01-31
PH12019501744A1 (en) 2019-09-30
WO2018140095A3 (en) 2018-11-29
US20210030742A1 (en) 2021-02-04
JP2022040408A (en) 2022-03-10
AU2017395703B2 (en) 2021-09-23
MX2022007141A (en) 2022-07-19
JP7199359B2 (en) 2023-01-05
JP2022174345A (en) 2022-11-22
JP2020505407A (en) 2020-02-20
JP2020536905A (en) 2020-12-17
US10874648B2 (en) 2020-12-29
KR20200126430A (en) 2020-11-06
CN111655034A (en) 2020-09-11
AU2017395704B8 (en) 2022-03-17
CN110740731A (en) 2020-01-31
MX2019008853A (en) 2019-09-11
KR20230038601A (en) 2023-03-20
AU2017395700A1 (en) 2019-08-22
WO2018140095A2 (en) 2018-08-02
AU2017395703A1 (en) 2019-08-22
EA202090676A1 (en) 2021-10-08
EA201991782A1 (en) 2021-09-23
JP7199360B2 (en) 2023-01-05
US10912771B1 (en) 2021-02-09
US10952997B2 (en) 2021-03-23
AU2017395704A8 (en) 2022-03-17
MX2020003462A (en) 2020-08-03

Similar Documents

Publication Publication Date Title
US10952997B2 (en) Methods for the administration of certain VMAT2 inhibitors
US11654142B2 (en) Methods for the administration of certain VMAT2 inhibitors
US10993941B2 (en) Methods for the administration of certain VMAT2 inhibitors

Legal Events

Date Code Title Description
AS Assignment

Owner name: NEUROCRINE BIOSCIENCES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:O'BRIEN, CHRISTOPHER F.;BOZIGIAN, HAIG P.;SIGNING DATES FROM 20190526 TO 20190703;REEL/FRAME:050397/0771

AS Assignment

Owner name: NEUROCRINE BIOSCIENCES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:O'BRIEN, CHRISTOPHER F.;BOZIGIAN, HAIG P.;SIGNING DATES FROM 20190529 TO 20190703;REEL/FRAME:050515/0734

STCB Information on status: application discontinuation

Free format text: ABANDONED -- INCOMPLETE APPLICATION (PRE-EXAMINATION)