US20110166215A1 - Methods of improving the pharmacokinetics of doxepin - Google Patents

Methods of improving the pharmacokinetics of doxepin Download PDF

Info

Publication number
US20110166215A1
US20110166215A1 US13/007,334 US201113007334A US2011166215A1 US 20110166215 A1 US20110166215 A1 US 20110166215A1 US 201113007334 A US201113007334 A US 201113007334A US 2011166215 A1 US2011166215 A1 US 2011166215A1
Authority
US
United States
Prior art keywords
doxepin
food
patient
max
administering
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
US13/007,334
Inventor
Cara Baron Casseday
Elizabeth Ludington
Michael Skinner
Susan Dubé
Roberta L. Rogowski
Philip Jochelson
Robert Mansbach
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.)
Somaxon Pharmaceuticals Inc
Original Assignee
Somaxon Pharmaceuticals 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=38658703&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20110166215(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Somaxon Pharmaceuticals Inc filed Critical Somaxon Pharmaceuticals Inc
Priority to US13/007,334 priority Critical patent/US20110166215A1/en
Publication of US20110166215A1 publication Critical patent/US20110166215A1/en
Priority to US13/653,213 priority patent/US9572814B2/en
Priority to US15/436,293 priority patent/US10653660B2/en
Priority to US16/876,492 priority patent/US11110074B2/en
Priority to US17/467,360 priority patent/US20210393567A1/en
Abandoned legal-status Critical Current

Links

Images

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
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • 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
    • A61P25/20Hypnotics; Sedatives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to methods of improving the pharmacokinetics of doxepin in a patient.
  • food effect refers to a somewhat unpredictable phenomenon that can influence the absorption of drugs from the gastrointestinal tract following oral administration.
  • a food effect can be designated “negative” when absorption is decreased, or “positive” when absorption is increased and manifested as an increase in oral bioavailability (as reflected by total exposure, usually defined as AUC).
  • food effects can refer to changes in maximum concentration (C max ), or the time to reach maximum concentration (T max ), independently of overall absorption. As a result, some drugs have to be taken in either fasted or fed conditions to achieve the optimum effect.
  • patients may be instructed to take a drug with a meal, before a meal (e.g., one hour before a meal), or after a meal (e.g., two hours after a meal).
  • a meal e.g., one hour before a meal
  • a meal e.g., two hours after a meal
  • drugs are unaffected by food, and thus, can be taken in either a fasted or a fed condition.
  • Doxepin is a tricyclic compound currently approved for treatment of depression and anxiety.
  • the recommended daily oral dose for the treatment of depression or anxiety ranges from 75 milligrams to 300 milligrams.
  • U.S. Pat. Nos. 5,502,047 and 6,211,229 describe the use of doxepin for the treatment chronic and non-chronic (e.g., transient/short term) insomnia.
  • Doxepin unlike most FDA-approved products for the treatment of insomnia, is not a Schedule IV controlled substance. Historically, doxepin pharmacokinetics have not been known to be affected by food.
  • a patient may have a set window of time within which they desire that their sleep occur.
  • it can be useful to minimize the amount of time required to attain a maximum concentration of a drug in order to receive the therapeutic benefit of the drug as soon as possible during the desired treatment period.
  • Some embodiments are based upon the surprising discovery about the food effects of doxepin. For example, as described more fully below, it has been discovered that administration of doxepin without food decreases the time to achieve maximum blood concentration or T max for doxepin. In one experiment, the administration of doxepin without food resulted in achieving T max three hours more quickly than when doxepin was administered with food. As another example, it has been discovered that administration of doxepin with food increases the overall bioavailability of doxepin and results in a higher maximum concentration (C max ) of doxepin. In one experiment, the administration of doxepin with food resulted in a 41% increase in bioavailability (AUC 0- ⁇ and a 15% increase in C max compared to administration in a fasted state.
  • One embodiment relates to a method of shortening the time required to achieve a maximum plasma concentration of doxepin in a patient receiving doxepin therapy, which method can include administering to the patient a therapeutically effective amount of doxepin in a pharmaceutical composition without food.
  • the methods can have various benefits, including more rapid onset of drug action, shorter duration of effect, etc.
  • the administration to the patient can occur, for example, at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours or at least about 4 hours, or more after consuming food. Also, administration to the patient can occur at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours, or at least about 4 hours, or more prior to consuming food, for example.
  • a desirable sleep medication preferably can have the ability to affect the onset of drug action and the duration of drug activity (e.g., to avoid hangover, etc.).
  • a person will desire to fall asleep as soon as possible, to stay asleep for about 8 hours, and to wake up without hangover or extra sedation at the end of the 8 hours.
  • the administration of doxepin without food resulted in achieving a maximum concentration of the drug three hours sooner compared to administration with food.
  • doxepin can be taken without food in order to achieve earlier onset of drug action and/or a shorter duration of drug action.
  • some embodiments relate to a method of shortening the time required to achieve sleep onset, which method can include administering to the patient a therapeutically effective amount of doxepin in a pharmaceutical composition without food.
  • another embodiment relates to a method of treating a sleep disorder comprising providing a patient with a therapeutically effective amount of doxepin and providing the patient with instructions to take the doxepin without food.
  • the doxepin can be provided to the patient at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours or at least about 4 hours or more after consuming food, or at least about 30 minutes, or at least about 1 hour prior to consuming food.
  • Still a further embodiment relates to a method of treating a sleep disorder comprising providing a patient with a therapeutically effective amount of doxepin and providing the patient with information regarding a doxepin food effect.
  • the information can be provided orally or in written form.
  • Some exemplary written forms include a label associated with the drug, on the container for the drug, packaged with the drug, or separately given to the patient apart from the drug.
  • Still some embodiments relate to the use of doxepin in the preparation of a medicament for treatment of a sleep disorder, said medicament for administration without food.
  • the use can further be for shortening the time required to achieve a maximal plasma concentration of doxepin by administration without food.
  • the administration without food can occur when the patient is in a fasted state.
  • the administration without food can occur, for example, at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours or at least about 4 hours, or more after consuming food. Also, it can occur, for example, at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours, or at least about 4 hours, or more prior to consuming food.
  • the administration of the doxepin can occur, for example, at least about 30 minutes, at least about 1 hour, or more prior to consuming food.
  • Some embodiments relate to a method of increasing the maximum plasma concentration of doxepin in a patient receiving doxepin therapy comprising administering to the patient a therapeutically effective amount of doxepin in a pharmaceutical composition with food.
  • some embodiments relate to methods of increasing the oral bioavailability of doxepin, including by administering to a patient an amount of a pharmaceutical oral dosage form of doxepin with food. Further embodiments relate to methods of increasing the oral bioavailability of doxepin to a patient receiving doxepin therapy, which methods can include administering to the patient an amount of a pharmaceutical oral dosage form of doxepin with food, wherein the administration results in an AUC 0- ⁇ that is greater than that achieved by the administration of the same amount of doxepin without food. In such methods, the doxepin can be administered as part of a chronic doxepin therapy, for example.
  • Still another embodiment relates to methods of treating depression or anxiety. It is worth noting that improved pharmacokinetics of doxepin in the context of depression or anxiety can be beneficial, for example by, leading to more safe and effective dosing.
  • the methods of treating depression or anxiety can include administering a therapeutically effective amount of doxepin preferably with food. In some embodiments the depression or anxiety can be treated by administering doxepin without food. Also, some embodiments relate to methods of treating depression or anxiety, including by providing a patient with a therapeutically effective amount of doxepin and providing the patient with instructions to preferably take the doxepin with food. In some alternative embodiments, the instructions can specify taking the doxepin without food.
  • Another embodiment relates to a method of treating depression or anxiety comprising providing a patient with a therapeutically effective amount of doxepin and providing the patient with information regarding a doxepin food effect.
  • some embodiments relate to the use of doxepin in the preparation of a medicament for treatment of a psychological disorder, said medicament for administration with food.
  • the disorder can preferably be depression or anxiety.
  • the use also can be for the preparation of a medicament for increasing the oral bioavailability of doxepin by administration with food; for increasing the oral bioavailability of doxepin to a patient receiving doxepin therapy by administering an amount of a pharmaceutical oral dosage form of doxepin with food, wherein the administration results in an AUC 0- ⁇ that is greater than that achieved by the administration of the same amount of doxepin without food; for increasing the time required to achieve a maximal plasma concentration of doxepin by administration with food; for minimizing side effects associated with a doxepin treatment of the psychological disorder, by administering a therapeutically effective amount of doxepin with food, which can result in the patient receiving or in a physician prescribing a lower dosage of doxepin compared the dosage that is taken by the patient without food; or for improving the consistency
  • some embodiments relate to methods of decreasing the oral bioavailability of doxepin. Decreasing oral bioavailability can be beneficial in some contexts. For example, as mentioned above for sleep therapies, it can be beneficial to shorten the duration of the drug action in order to minimize hangover or other effects.
  • the methods of decreasing bioavailability can include administering to a patient an amount of a pharmaceutical oral dosage form of doxepin without food.
  • some embodiments relate to methods of decreasing the oral bioavailability of doxepin to a patient receiving doxepin therapy, comprising administering to the patient an amount of a pharmaceutical oral dosage form of doxepin without food, wherein the administration results in an AUC 0- ⁇ that is less than that achieved by the administration of the same amount of doxepin with food.
  • doxepin can be administered as part of a chronic doxepin therapy, for example.
  • administration to the patient can occur, for example, at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours or at least about 4 hours, or more after consuming food.
  • administration to the patient can occur at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours, or at least about 4 hours, or more prior to consuming food, for example.
  • Some embodiments relate to methods of minimizing side effects associated with a doxepin therapy.
  • the methods can include, for example, administering a therapeutically effective amount of doxepin to a patient with food. Also, the administration of doxepin with food can result in a patient receiving or a physician prescribing a lower dose of doxepin compared to the dosage prescribed without food.
  • This disclosure includes a method for improving the consistency of pharmacokinetics associated with doxepin therapy, in which a patient receives multiple doxepin dosages over multiple days, comprising administering the doxepin to the patient in a fixed temporal relationship to food intake by the patient.
  • This can, for example, include providing written or verbal instructions to the patient to take the doxepin in a fixed temporal relationship to food intake by the patient. Taking doxepin in a fixed relationship to food can help assure more consistent efficacy of the medication.
  • some embodiments relate to methods of alleviating a doxepin food effect or altering a doxepin pharmacokinetic parameter in a patient, for example, by administering doxepin to a patient in need thereof, wherein the patient is in a non-fasted state or in a fasted state.
  • the amount of doxepin may advantageously be, for example, about 0.001 milligram to about 350 milligrams, preferably about 50 milligrams to about 300 milligrams or more preferably about 75 milligrams to about 300 milligrams, or any amount or sub-range within those ranges.
  • the effective amount may be about 0.5 milligrams to about 20 milligrams, more preferably about 1 milligram to about 6 milligrams.
  • the pharmaceutical composition of doxepin can be, for example, a tablet, capsule or liquid.
  • the doxepin can be provided or administered as a unit dosage form.
  • the doxepin can be provided or administered as an oral dosage form.
  • kits or a product that includes doxepin and written instructions or information associated therewith to take the doxepin without food.
  • the instructions can specify that doxepin be administered to the patient at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours, at least about 4 hours or more after consuming food, or at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours, at least about 4 hours or more prior to consuming food, for example.
  • the kits or product can include instructions that describe administration to the patient preferably at least about 30 minutes or at least about 1 hour prior to consuming food.
  • the kit or product can include information regarding a food effect associated with doxepin.
  • it includes a kit or a product comprising doxepin and written instructions associated therewith to take the doxepin with food.
  • the product or kit can include doxepin in amount of about 0.5 milligram to about 350 milligrams, preferably about 50 milligrams to about 300 milligrams or more preferably about 75 milligrams to about 300 milligrams.
  • the amount may be about 0.5 milligrams to about 20 milligrams, more preferably about 1 milligram to about 6 milligrams. Some preferred amounts are about 1 milligram, about 3 milligrams and about 6 milligrams.
  • the product or kit can include doxepin as a tablet, a capsule, a liquid, a unite dosage form or an oral dosage form, for example.
  • FIG. 1 illustrates the design of a study of food effect associated with doxepin.
  • Embodiments described herein relate to the novel and unexpected discovery of a food effect associated with doxepin.
  • a single-center, phase 1, randomized, open-label, single dose, two-way crossover study was conducted in 16 healthy young adult male and female volunteers.
  • a food effect was observed on the pharmacokinetics of doxepin.
  • pharmacokinetic parameters (AUC 0- ⁇ and C max ) of doxepin were approximately 41% and 15% higher, respectively, compared to the fasted state, and median T max was delayed by approximately 3.0 hours in the fed state.
  • the increase in AUC was statistically significant and demonstrates a positive food effect on exposure.
  • the increased T max in the fed state suggests that food slows the absorption of the drug.
  • some embodiments relate to methods of improving the pharmacokinetics of doxepin in a patient.
  • administration of doxepin in a fasted state can result in decreasing the time required to achieve a maximum plasma concentration of doxepin.
  • the time to reach maximum concentration (T max ) of doxepin can be minimized by administering the drug without food.
  • administration of doxepin with food can increase the overall bioavailability of the drug. For example, the time to reach maximum concentration can be increased by administering doxepin with food and the bioavailability of the drug can be increased.
  • the total effective amount of drug that the patient receives can be maximized by administering doxepin with food, while in other embodiments the oral bioavailability of doxepin can be decreased by administering the doxepin without food.
  • plasma concentrations and half-lives of doxepin are already known to vary from patient to patient, knowledge of the doxepin food effect can help patients and physicians to eliminate this additional source of dosing uncertainty, to improve safety and tolerability, and improve therapies that utilize doxepin.
  • doxepin can be taken with food; it can be taken after the patient has gone without food for a period of time; and/or it can be taken some period of time prior to consuming food.
  • doxepin preferably can be taken without food; in order to increase the bioavailability of the drug where rapid onset and shorter duration are not issues, doxepin preferably can be taken with food; and to assure more consistent efficacy, doxepin preferably can be taken in a fixed relationship to food consumption, regardless of whether the drug is taken with our without food.
  • Short term exposure with more rapid onset can be preferable when treating, for example, a sleep disorder, while increased bioavailability and/or consistent kinetics can be preferable for treating conditions such as depression and anxiety.
  • Doxepin HCl is a tricyclic compound currently approved for treatment of depression and anxiety.
  • the recommended daily dose for the treatment of depression or anxiety ranges from 75 milligrams to 300 milligrams.
  • U.S. Pat. Nos. 5,502,047 and 6,211,229 the entire contents of which are incorporated herein by reference, describe the use of doxepin for the treatment chronic and non-chronic (e.g., transient/short term) insomnias at dosages below those used to treat depression.
  • doxepin for the treatment chronic and non-chronic (e.g., transient/short term) insomnias at dosages below those used to treat depression.
  • a food effect associated with doxepin treatment of currently approved disorders or sleep disorders has not previously been reported.
  • Some embodiments relate to methods of improving the pharmacokinetics of doxepin, including by administering doxepin with or without food.
  • a number of pharmacokinetic parameters can be affected by taking doxepin with or without food, including for example, T max , C max , and the area under the curve (AUC).
  • various therapeutic regimens can be utilized to take advantage of the doxepin food effects.
  • T max maximum plasma concentration
  • Some embodiments relate to methods of shortening the time required to achieve a maximum plasma concentration or a T max of doxepin in a patient receiving doxepin therapy, which methods can include administering to the patient a therapeutically effective amount of doxepin in a pharmaceutical composition without food.
  • Some embodiments relate to methods of preventing a doxepin food effect in order to minimize the time required for onset of action of the drug.
  • the methods can include administering doxepin to a patient in need thereof, wherein the patient is in a fasted state or has not eaten or will not eat within a particular time period.
  • the methods further can include providing instructions to take the doxepin without food or in a fasted state.
  • some embodiments relate to improved methods of treating a sleep disorder.
  • the methods can include providing a patient with a therapeutically effective amount of doxepin and providing the patient with instructions to take the doxepin without food.
  • doxepin can be administered without food in order to minimize the amount of time to achieve sleep onset or to otherwise minimize the amount of time before drug action occurs.
  • the information regarding the doxepin food effect can be provided to the patient.
  • the information can include, for example, instructions that may be provided to patients receiving doxepin therapy or health care professionals involved in treatment of those patients that the doxepin should be administered without food, preferably separated from food for the time periods discussed above.
  • such instructions could be provided orally or in written form.
  • Some exemplary written forms include a label associated with the drug, on the container for the drug, packaged with the drug, or separately given to the patient apart from the drug, including providing the patient with access to a website or other electronic information with the instructions/information.
  • the maximum plasma concentration or overall bioavailability of doxepin can be affected by food or a lack thereof. Increasing concentration or bioavailability can be desirable in some circumstances.
  • Some embodiments relate to methods of increasing the oral bioavailability of doxepin to a patient receiving doxepin therapy.
  • the methods can include administering to the patient a pharmaceutical oral dosage form of doxepin with food.
  • the administration can result in a greater AUC than if the drug is taken without food.
  • the methods can include administering to the patient a therapeutically effective amount of doxepin in a pharmaceutical composition with food or within a relatively short time of consuming food (e.g., 15 minutes, 30 minutes, one hour, etc.).
  • some embodiments relate to a method of increasing the maximum plasma concentration of doxepin in a patient receiving doxepin therapy comprising administering to the patient a therapeutically effective amount of doxepin in a pharmaceutical composition with food.
  • Doxepin has been used for several decades in the treatment of depression and anxiety. Several side effects have been reported in connection with the use of doxepin to treat depression or anxiety. The studies disclosed herein show that by taking doxepin with food, AUC was increased by 41% compared to taking doxepin in a fasted state. Because AUC (bioavailability) is increased by taking doxepin with food, a patient can take a lower dose compared to when a patient takes doxepin without food. Increasing the oral bioavailability or AUC of doxepin, and decreasing dosage required for treatment can minimize or alleviate side effects and improve the safety and tolerability of doxepin therapy.
  • doxepin for depression or anxiety should take the drug with food, or in close proximity to eating.
  • Some embodiments relate to improved methods of treating depression and anxiety.
  • the methods can include providing a patient with a therapeutically effective amount of doxepin and providing the patient with instructions to take the doxepin with food.
  • administering doxepin with food can result in an increase in the bioavailability of doxepin.
  • a patient can take less doxepin, which can be safer and more tolerable for the patient.
  • information may be provided to patients receiving doxepin therapy or health care professionals involved in treatment of those patients regarding a doxepin food effect and/or that the doxepin should be administered with food, or at least in relatively close proximity to eating food or eating a meal (for example, within one hour or less).
  • information or instructions could be provided orally or in written form.
  • Some exemplary written forms include a label associated with the drug, on the container for the drug, packaged with the drug, or separately given to the patient apart from the drug, including providing the patient with access to a website or other electronic information with the instructions/information.
  • the invention further includes a package of doxepin with such written instructions associated therewith.
  • aspects of the invention also relate to methods of treating depression or anxiety by administering doxepin without food. Such embodiments can also include instructions to take the medication without food.
  • Still further embodiments relate to methods for improving the consistency of pharmacokinetics associated with doxepin therapy, in which a patient receives a multiple doxepin dosages over multiple days.
  • the methods can include administering the doxepin to the patient in a fixed temporal relationship to food intake by the patient.
  • the method can further include providing instructions to the patient to take the doxepin in a fixed temporal relationship to food intake by the patient.
  • taking doxepin in fixed or consistent temporal relationship to food can lead to improved safety and tolerability for the patient, for example, due to less variability in the drug kinetics in the patient.
  • kits and products for a therapy that includes the use of doxepin.
  • the kits and products can include doxepin and instructions to take the doxepin without food or in a fasted state, or to take the doxepin with food or within a predetermined period of eating food.
  • the instructions or information regarding a food effect can be provided orally or verbally, or could be in written form.
  • Some exemplary written forms include a label associated with the drug, on the container for the drug, packaged with the drug, or separately given to the patient apart from the drug, including providing the patient with access to a web site or other electronic information with the instructions/information.
  • the invention further includes a package of doxepin with such written instructions associated therewith or with information on where to access the instructions/information (e.g., a website).
  • doxepin, a pharmaceutically acceptable salt of doxepin, or prodrug of doxepin can be administered in any suitable oral form.
  • doxepin, or a pharmaceutically acceptable salt or a prodrug thereof can be administered to a patient.
  • administer and its variants are intended to cover both self-administration and administration by another person or by a device.
  • Doxepin can be administered without food or in a fasted state.
  • doxepin can be administered at least about 30 minutes to about 4, 5, 6 or more hours after consuming food. More preferably, doxepin can be taken at least about 1 hour to about 6 hours after consuming food. In some aspects doxepin can be taken at least about 1, 2, 3, 4, 5 6 or more hours after consuming food.
  • doxepin can be administered at least about 30 minutes to about 6 hours before consuming any food, or more preferably, at least about 1 hour to about 3 hours before consuming food. In some aspects, doxepin can be administered about 1, 2, 3 or more hours before food is consumed.
  • instructions may be provided to patients receiving doxepin therapy or health care professionals involved in treatment of those patients that the doxepin should be administered without food, preferably separated from food for the time periods discussed above. Also, the patient can be provided with information regarding a doxepin food effect. In other embodiments of the invention, such as when doxepin is used to treat depression or anxiety, instructions may be provided to patients receiving doxepin therapy or health care professionals involved in treatment of those patients that the doxepin should be administered with food, or at least in relatively close proximity to eating food or eating a meal (for example, within one hour or less).
  • Such instructions or information regarding a food effect could be provided orally or verbally, or could be in written form.
  • Some exemplary written forms include a label associated with the drug, on the container for the drug, packaged with the drug, or separately given to the patient apart from the drug, including providing the patient with access to a website or other electronic information with the instructions/information.
  • the invention further includes a package of doxepin with such written instructions associated therewith.
  • the food effect disclosed herein can appropriately play a part in designing customized dosing regimens that reflect the preferences of a particular healthcare professional or their patient.
  • doxepin therapy e.g., chronic doxepin therapy
  • doxepin is administered without food, even though more drug will be required to achieve the same effect. In such cases, consistency is important to maintaining equivalent pharmacological effects.
  • one embodiment includes administering doxepin in a consistent relationship to food intake, regardless of whether it is or is not taken with food.
  • Suitable routes of administration of doxepin can include any route in which significant quantities of drug reach the stomach, including oral, buccal, and sublingual administration.
  • the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Administration though oral pathways can be accomplished, for example, using a capsule, a tablet, a granule, a spray, a syrup, a liquid, powder, granules, pastes (e.g., for application to the tongue).
  • Oral administration can be accomplished using fast-melt formulations, for example.
  • rapidly-melting strips or sheets that include the drug and suitable excipients can be prepared that dissolve quickly in the mouth, using well-known formulation technology.
  • the compositions may take any suitable form, for example, tablets or lozenges.
  • Pharmaceutical preparations for oral use can be obtained by mixing one or more solid excipient with pharmaceutical combination of the invention, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • compositions which can be used orally include for example, liquid solutions, powders, and suspensions in bulk or unit dosage forms.
  • the oral formulations can include, for example, pills, tablets, granules, sprays, syrups, pastes, powders, boluses, pre-measured ampules or syringes, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
  • doxepin can be used alone or in combination with other substances, such as for example, other insomnia or sleep medications, or with other medications that treat a primary illness.
  • Doxepin alone or in combination with other drugs can be included as part of a composition.
  • the compounds and compositions can include any suitable form of the compound for pharmaceutical delivery, as discussed in further detail herein.
  • compositions and formulations disclosed herein also can include one or more pharmaceutically acceptable carrier materials or excipients. Such compositions can be prepared for storage and for subsequent administration. Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in the incorporated material of Remington: The Science and Practice of Pharmacy (20 th ed, Lippincott Williams & Wilkens Publishers (2003)), which is incorporated herein by reference in its entirety.
  • carrier material or “excipient” herein can mean any substance, not itself a therapeutic agent, used as a carrier and/or diluent and/or adjuvant, or vehicle for delivery of a therapeutic agent to a subject or added to a pharmaceutical composition to improve its handling or storage properties or to permit or facilitate formation of a dose unit of the composition into a discrete article such as a capsule or tablet suitable for oral administration.
  • Excipients can include, by way of illustration and not limitation, diluents, disintegrants, binding agents, adhesives, wetting agents, polymers, lubricants, glidants, substances added to mask or counteract a disagreeable taste or odor, flavors, dyes, fragrances, and substances added to improve appearance of the composition.
  • Acceptable excipients include sugars (such as lactose, sucrose, mannitol, sorbitol), starch powder, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, or derivatives thereof, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, gelatin, acacia gum, sodium alginate, polyvinyl-pyrrolidone (PVP), and/or polyvinyl alcohol, saline, dextrose, lecithin, albumin, sodium glutamate, cysteine hydrochloride, and the like.
  • sugars such as lactose, sucrose, mannitol, sorbitol
  • starch powder maize starch, wheat starch, rice starch
  • suitable excipients for soft gelatin capsules include vegetable oils, waxes, fats, semisolid and liquid polyols.
  • suitable excipients for the preparation of solutions and syrups include, without limitation, water, polyols, sucrose, invert sugar and glucose.
  • Suitable excipients for injectable solutions include, without limitation, water, alcohols, polyols, glycerol, and vegetable oils.
  • the compositions can include disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • compositions for oral administration can be formulated according to conventional pharmaceutical practice as described in the incorporated material in Remington: The Science and Practice of Pharmacy (20 th ed, Lippincott Williams & Wilkens Publishers (2003)). For example, dissolution or suspension of the active compound in a vehicle such as water or naturally occurring vegetable oil like sesame, peanut, or cottonseed oil or a synthetic fatty vehicle like ethyl oleate or the like may be desired. Buffers, preservatives, antioxidants and the like can be incorporated according to accepted pharmaceutical practice.
  • the compound can also be made in microencapsulated form.
  • sustained release forms or from sustained release drug delivery systems.
  • sustained release materials can be found in the incorporated materials in Remington: The Science and Practice of Pharmacy (20 th ed, Lippincott Williams & Wilkens Publishers (2003)).
  • the selected dosage level can depend upon, for example, the condition being treated, the route of administration, the severity of the condition being treated, and the condition and prior medical history of the patient being treated. However, it is within the skill of the art to start doses of the compound at levels lower than required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. It will be understood, however, that the specific dose level for any particular patient can depend upon a variety of factors including the genetic makeup, body weight, general health, diet, time and route of administration, combination with other drugs and the particular condition being treated, and its severity. For the treatment of insomnia, preferably one dose is administered prior to bedtime.
  • the preferable dosage can be between about 0.001 milligrams and about 350 milligrams.
  • the dosage can be about 50 milligrams to about 350 milligrams. More preferably, the dosage can be between about 75 milligrams and 300 milligrams.
  • the dosage can be between about 0.1 milligrams and 20 milligrams or between about 0.5 milligrams and 10 milligrams.
  • the dosage also can be between about 1 milligram and about 6 milligrams.
  • the dosage can be about 0.5 milligrams, 1 milligram, about 2 milligrams, about 3 milligrams, about 4 milligrams, about 5 milligrams or about 6 milligrams.
  • the dosage can be about 7 milligrams, about 8 milligrams, about 9 milligrams, or about 10 milligrams.
  • the lower dosage ranges are particularly desirable for sleep-related indications, while the higher dosage ranges are particularly desirable for depression and anxiety-related indications.
  • PK pharmacokinetics
  • Subjects received a single 6 milligram dose of doxepin in the morning under either fed or fasted conditions on 2 dosing days (Day 1 and Day 8). There were approximately 7 days between each dose. Enrolled subjects were admitted to the study center on the evening before study drug dosing (Day 0 and Day 7) and remained at the study center for approximately 5 days. All subjects were dosed under both fed and fasted conditions during the study as illustrated in FIG. 1 .
  • Subjects being dosed under fasted conditions were required to fast overnight for at least 10 hours prior to study drug administration and for 4 hours after study drug administration. Fluids were restricted from 1 hour predose to 1 hour postdose, except for water taken at the time of dosing. Subjects being dosed under fed conditions were dosed approximately 5 minutes after eating a high-fat, high-calorie standardized breakfast (to be ingested within 25 minutes). Subjects were required to ingest the entire contents of the breakfast. All subjects were required to remain in bed for approximately 4 hours after dosing.
  • the total amount of protein, fat, and carbohydrate that made up this meal was approximately 33, 55, and 58 g, respectively.
  • the total calorie content was approximately 850 kcal.
  • PK profiles were evaluated on Days 1 through 5 and Days 8 through 12. Blood samples were collected at predose (0 hour) and at 0.08, 0.17, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 4, 6, 8, 12, 24, 36, 48, 60, 72, and 96 hours postdose. The samples were analyzed for doxepin and doxepin metabolite concentrations in plasma. Plasma concentrations of doxepin were measured using validated high performance liquid chromatography coupled to tandem mass spectrometry. The lower limit of quantification for doxepin was 0.05 ng/mL. The following PK parameters were estimated by noncompartmental methods using actual elapsed time from dosing:
  • Table 2 shows the estimates of clearance and volume of distribution for doxepin.
  • Mean CL/F and Vd/F were 43% and 14% lower in the fed state compared to the fasted state, respectively.
  • Mean CL/F values were lower in the fed (477 L/h) versus the fasted (837 L/h) states.
  • Mean Vd/F remained almost unchanged for fed (10280 L) and fasted (11930 L) states.

Abstract

Methods of improving the pharmacokinetics of doxepin in a patient.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 11/781,165, filed Jul. 20, 2007, which claims priority to U.S. Provisional Application Nos. 60/832,727 and 60/833,617, respectively filed on Jul. 20, 2006 and Jul. 24, 2006, both entitled METHODS OF IMPROVING THE PHARMACOKINETICS OF DOXEPIN. The disclosure of each of the above-described applications is hereby incorporated by reference in its entirety.
  • FIELD OF THE INVENTION
  • The present invention relates to methods of improving the pharmacokinetics of doxepin in a patient.
  • BACKGROUND OF THE INVENTION
  • The term “food effect” refers to a somewhat unpredictable phenomenon that can influence the absorption of drugs from the gastrointestinal tract following oral administration. A food effect can be designated “negative” when absorption is decreased, or “positive” when absorption is increased and manifested as an increase in oral bioavailability (as reflected by total exposure, usually defined as AUC). Alternatively, food effects can refer to changes in maximum concentration (Cmax), or the time to reach maximum concentration (Tmax), independently of overall absorption. As a result, some drugs have to be taken in either fasted or fed conditions to achieve the optimum effect. For example, patients may be instructed to take a drug with a meal, before a meal (e.g., one hour before a meal), or after a meal (e.g., two hours after a meal). However, many drugs are unaffected by food, and thus, can be taken in either a fasted or a fed condition.
  • Doxepin is a tricyclic compound currently approved for treatment of depression and anxiety. The recommended daily oral dose for the treatment of depression or anxiety ranges from 75 milligrams to 300 milligrams. Also, U.S. Pat. Nos. 5,502,047 and 6,211,229 describe the use of doxepin for the treatment chronic and non-chronic (e.g., transient/short term) insomnia. Doxepin, unlike most FDA-approved products for the treatment of insomnia, is not a Schedule IV controlled substance. Historically, doxepin pharmacokinetics have not been known to be affected by food.
  • In treating depression, anxiety and sleep disorders it is beneficial to optimize the pharmacokinetics of the administered medication in a patient. For example, in the case of sleep disorders a patient may have a set window of time within which they desire that their sleep occur. Thus, it can be useful to minimize the amount of time required to attain a maximum concentration of a drug in order to receive the therapeutic benefit of the drug as soon as possible during the desired treatment period.
  • SUMMARY OF THE INVENTION
  • Some embodiments are based upon the surprising discovery about the food effects of doxepin. For example, as described more fully below, it has been discovered that administration of doxepin without food decreases the time to achieve maximum blood concentration or Tmax for doxepin. In one experiment, the administration of doxepin without food resulted in achieving Tmax three hours more quickly than when doxepin was administered with food. As another example, it has been discovered that administration of doxepin with food increases the overall bioavailability of doxepin and results in a higher maximum concentration (Cmax) of doxepin. In one experiment, the administration of doxepin with food resulted in a 41% increase in bioavailability (AUC0-∞ and a 15% increase in Cmax compared to administration in a fasted state.
  • As a result of the various discoveries related to the food effects of doxepin and depending upon the type of therapy and the desired overall result of that therapy, a patient can benefit from a number of different therapeutic regimens. Disclosed are various therapeutic regimens influenced by the food effects observed with doxepin.
  • Achieving a More Rapid Maximum Concentration (Tmax)
  • In some circumstances, more rapid onset of doxepin action may be desired. One embodiment relates to a method of shortening the time required to achieve a maximum plasma concentration of doxepin in a patient receiving doxepin therapy, which method can include administering to the patient a therapeutically effective amount of doxepin in a pharmaceutical composition without food. The methods can have various benefits, including more rapid onset of drug action, shorter duration of effect, etc. The administration to the patient can occur, for example, at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours or at least about 4 hours, or more after consuming food. Also, administration to the patient can occur at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours, or at least about 4 hours, or more prior to consuming food, for example.
  • Sleep Therapy:
  • Another embodiment relates to methods of treating sleep disorders. A desirable sleep medication preferably can have the ability to affect the onset of drug action and the duration of drug activity (e.g., to avoid hangover, etc.). Generally, a person will desire to fall asleep as soon as possible, to stay asleep for about 8 hours, and to wake up without hangover or extra sedation at the end of the 8 hours. As mentioned above, surprisingly, the administration of doxepin without food resulted in achieving a maximum concentration of the drug three hours sooner compared to administration with food. As a result, when treating sleep, doxepin can be taken without food in order to achieve earlier onset of drug action and/or a shorter duration of drug action.
  • Thus, some embodiments relate to a method of shortening the time required to achieve sleep onset, which method can include administering to the patient a therapeutically effective amount of doxepin in a pharmaceutical composition without food. Also, another embodiment relates to a method of treating a sleep disorder comprising providing a patient with a therapeutically effective amount of doxepin and providing the patient with instructions to take the doxepin without food. The doxepin can be provided to the patient at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours or at least about 4 hours or more after consuming food, or at least about 30 minutes, or at least about 1 hour prior to consuming food.
  • Still a further embodiment relates to a method of treating a sleep disorder comprising providing a patient with a therapeutically effective amount of doxepin and providing the patient with information regarding a doxepin food effect. The information can be provided orally or in written form. Some exemplary written forms include a label associated with the drug, on the container for the drug, packaged with the drug, or separately given to the patient apart from the drug.
  • Still some embodiments relate to the use of doxepin in the preparation of a medicament for treatment of a sleep disorder, said medicament for administration without food. Also, the use can further be for shortening the time required to achieve a maximal plasma concentration of doxepin by administration without food. The administration without food can occur when the patient is in a fasted state. The administration without food can occur, for example, at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours or at least about 4 hours, or more after consuming food. Also, it can occur, for example, at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours, or at least about 4 hours, or more prior to consuming food. In some aspects, the administration of the doxepin can occur, for example, at least about 30 minutes, at least about 1 hour, or more prior to consuming food.
  • Achieving a Greater Cmax or a Greater Bioavailability Greater Cmax:
  • Some embodiments relate to a method of increasing the maximum plasma concentration of doxepin in a patient receiving doxepin therapy comprising administering to the patient a therapeutically effective amount of doxepin in a pharmaceutical composition with food.
  • Greater Bioavailability:
  • Also, some embodiments relate to methods of increasing the oral bioavailability of doxepin, including by administering to a patient an amount of a pharmaceutical oral dosage form of doxepin with food. Further embodiments relate to methods of increasing the oral bioavailability of doxepin to a patient receiving doxepin therapy, which methods can include administering to the patient an amount of a pharmaceutical oral dosage form of doxepin with food, wherein the administration results in an AUC0-∞ that is greater than that achieved by the administration of the same amount of doxepin without food. In such methods, the doxepin can be administered as part of a chronic doxepin therapy, for example.
  • Anxiety/Depression Therapy:
  • Still another embodiment relates to methods of treating depression or anxiety. It is worth noting that improved pharmacokinetics of doxepin in the context of depression or anxiety can be beneficial, for example by, leading to more safe and effective dosing. The methods of treating depression or anxiety can include administering a therapeutically effective amount of doxepin preferably with food. In some embodiments the depression or anxiety can be treated by administering doxepin without food. Also, some embodiments relate to methods of treating depression or anxiety, including by providing a patient with a therapeutically effective amount of doxepin and providing the patient with instructions to preferably take the doxepin with food. In some alternative embodiments, the instructions can specify taking the doxepin without food.
  • Another embodiment relates to a method of treating depression or anxiety comprising providing a patient with a therapeutically effective amount of doxepin and providing the patient with information regarding a doxepin food effect.
  • Also, some embodiments relate to the use of doxepin in the preparation of a medicament for treatment of a psychological disorder, said medicament for administration with food. The disorder can preferably be depression or anxiety. The use also can be for the preparation of a medicament for increasing the oral bioavailability of doxepin by administration with food; for increasing the oral bioavailability of doxepin to a patient receiving doxepin therapy by administering an amount of a pharmaceutical oral dosage form of doxepin with food, wherein the administration results in an AUC0-∞ that is greater than that achieved by the administration of the same amount of doxepin without food; for increasing the time required to achieve a maximal plasma concentration of doxepin by administration with food; for minimizing side effects associated with a doxepin treatment of the psychological disorder, by administering a therapeutically effective amount of doxepin with food, which can result in the patient receiving or in a physician prescribing a lower dosage of doxepin compared the dosage that is taken by the patient without food; or for improving the consistency of pharmacokinetics associated with doxepin therapy, in which a patient receives a multiple doxepin dosages over multiple days, comprising administering the doxepin in a fixed temporal relationship to food intake by the patient.
  • Decreasing Bioavailability:
  • It should be noted that some embodiments relate to methods of decreasing the oral bioavailability of doxepin. Decreasing oral bioavailability can be beneficial in some contexts. For example, as mentioned above for sleep therapies, it can be beneficial to shorten the duration of the drug action in order to minimize hangover or other effects. The methods of decreasing bioavailability can include administering to a patient an amount of a pharmaceutical oral dosage form of doxepin without food. Furthermore, some embodiments relate to methods of decreasing the oral bioavailability of doxepin to a patient receiving doxepin therapy, comprising administering to the patient an amount of a pharmaceutical oral dosage form of doxepin without food, wherein the administration results in an AUC0-∞ that is less than that achieved by the administration of the same amount of doxepin with food. In such methods, doxepin can be administered as part of a chronic doxepin therapy, for example. Also, in such methods administration to the patient can occur, for example, at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours or at least about 4 hours, or more after consuming food. Also, administration to the patient can occur at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours, or at least about 4 hours, or more prior to consuming food, for example.
  • Minimizing Doxepin Side Effects and Improving Pharmacokinetic Consistency
  • Some embodiments relate to methods of minimizing side effects associated with a doxepin therapy. The methods can include, for example, administering a therapeutically effective amount of doxepin to a patient with food. Also, the administration of doxepin with food can result in a patient receiving or a physician prescribing a lower dose of doxepin compared to the dosage prescribed without food.
  • This disclosure includes a method for improving the consistency of pharmacokinetics associated with doxepin therapy, in which a patient receives multiple doxepin dosages over multiple days, comprising administering the doxepin to the patient in a fixed temporal relationship to food intake by the patient. This can, for example, include providing written or verbal instructions to the patient to take the doxepin in a fixed temporal relationship to food intake by the patient. Taking doxepin in a fixed relationship to food can help assure more consistent efficacy of the medication.
  • Other Food Effect Methods
  • Also, some embodiments relate to methods of alleviating a doxepin food effect or altering a doxepin pharmacokinetic parameter in a patient, for example, by administering doxepin to a patient in need thereof, wherein the patient is in a non-fasted state or in a fasted state.
  • Further embodiments relate to a method of treating a disorder with doxepin comprising providing a patient with a therapeutically effective amount of doxepin and providing the patient with information regarding a doxepin food effect. The information can include, for example, instructions to take the doxepin with or without food, or to ensure that doxepin is consistently taken either with or without food. The information can be in an oral or written form. Some exemplary written forms include a label associated with the drug, on the container for the drug, packaged with the drug, separately given to the patient apart from the drug, or provided in manner that the patient can independently obtain the information (e.g., a web site).
  • Dosage and Compositions
  • Again, in the various disclosed embodiments, the amount of doxepin, including the therapeutically effective amount, may advantageously be, for example, about 0.001 milligram to about 350 milligrams, preferably about 50 milligrams to about 300 milligrams or more preferably about 75 milligrams to about 300 milligrams, or any amount or sub-range within those ranges. Alternatively, the effective amount may be about 0.5 milligrams to about 20 milligrams, more preferably about 1 milligram to about 6 milligrams.
  • Also, in the various disclosed embodiments, the pharmaceutical composition of doxepin can be, for example, a tablet, capsule or liquid. Furthermore, the doxepin can be provided or administered as a unit dosage form. Preferably, the doxepin can be provided or administered as an oral dosage form.
  • Doxepin Kits and Products
  • Finally, the present disclosure also includes a kit or a product that includes doxepin and written instructions or information associated therewith to take the doxepin without food. For example, the instructions can specify that doxepin be administered to the patient at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours, at least about 4 hours or more after consuming food, or at least about 30 minutes, at least about 1 hour, at least about 2 hours, at least about 3 hours, at least about 4 hours or more prior to consuming food, for example. In some aspects, the kits or product can include instructions that describe administration to the patient preferably at least about 30 minutes or at least about 1 hour prior to consuming food. The kit or product can include information regarding a food effect associated with doxepin. Alternatively, it includes a kit or a product comprising doxepin and written instructions associated therewith to take the doxepin with food.
  • The product or kit can include doxepin in amount of about 0.5 milligram to about 350 milligrams, preferably about 50 milligrams to about 300 milligrams or more preferably about 75 milligrams to about 300 milligrams. Alternatively, the amount may be about 0.5 milligrams to about 20 milligrams, more preferably about 1 milligram to about 6 milligrams. Some preferred amounts are about 1 milligram, about 3 milligrams and about 6 milligrams.
  • Finally, the product or kit can include doxepin as a tablet, a capsule, a liquid, a unite dosage form or an oral dosage form, for example.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 illustrates the design of a study of food effect associated with doxepin.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Embodiments described herein relate to the novel and unexpected discovery of a food effect associated with doxepin. As described more fully below, a single-center, phase 1, randomized, open-label, single dose, two-way crossover study was conducted in 16 healthy young adult male and female volunteers. A food effect was observed on the pharmacokinetics of doxepin. In the fed state, pharmacokinetic parameters (AUC0-∞ and Cmax) of doxepin were approximately 41% and 15% higher, respectively, compared to the fasted state, and median Tmax was delayed by approximately 3.0 hours in the fed state. The increase in AUC was statistically significant and demonstrates a positive food effect on exposure. The increased Tmax in the fed state suggests that food slows the absorption of the drug.
  • Accordingly, some embodiments relate to methods of improving the pharmacokinetics of doxepin in a patient. As discussed above and more fully elsewhere herein, administration of doxepin in a fasted state can result in decreasing the time required to achieve a maximum plasma concentration of doxepin. In particular, the time to reach maximum concentration (Tmax) of doxepin can be minimized by administering the drug without food. Also, as discussed above and more fully elsewhere herein, administration of doxepin with food can increase the overall bioavailability of the drug. For example, the time to reach maximum concentration can be increased by administering doxepin with food and the bioavailability of the drug can be increased.
  • In addition, in a different embodiment, the total effective amount of drug that the patient receives can be maximized by administering doxepin with food, while in other embodiments the oral bioavailability of doxepin can be decreased by administering the doxepin without food. Because plasma concentrations and half-lives of doxepin are already known to vary from patient to patient, knowledge of the doxepin food effect can help patients and physicians to eliminate this additional source of dosing uncertainty, to improve safety and tolerability, and improve therapies that utilize doxepin. For example, as described more fully elsewhere herein, depending on the effect desired, doxepin can be taken with food; it can be taken after the patient has gone without food for a period of time; and/or it can be taken some period of time prior to consuming food.
  • As a result of the food effect discovery, various improved therapeutic methods are provided, including: where short term exposure is desired, with a more rapid onset and shorter duration of effect, doxepin preferably can be taken without food; in order to increase the bioavailability of the drug where rapid onset and shorter duration are not issues, doxepin preferably can be taken with food; and to assure more consistent efficacy, doxepin preferably can be taken in a fixed relationship to food consumption, regardless of whether the drug is taken with our without food. Short term exposure with more rapid onset can be preferable when treating, for example, a sleep disorder, while increased bioavailability and/or consistent kinetics can be preferable for treating conditions such as depression and anxiety.
  • Doxepin HCl is a tricyclic compound currently approved for treatment of depression and anxiety. The recommended daily dose for the treatment of depression or anxiety ranges from 75 milligrams to 300 milligrams. Also, U.S. Pat. Nos. 5,502,047 and 6,211,229, the entire contents of which are incorporated herein by reference, describe the use of doxepin for the treatment chronic and non-chronic (e.g., transient/short term) insomnias at dosages below those used to treat depression. A food effect associated with doxepin treatment of currently approved disorders or sleep disorders has not previously been reported.
  • Methods of Improving the Pharmacokinetics of Doxepin
  • Some embodiments relate to methods of improving the pharmacokinetics of doxepin, including by administering doxepin with or without food. A number of pharmacokinetic parameters can be affected by taking doxepin with or without food, including for example, Tmax, Cmax, and the area under the curve (AUC). Furthermore, various therapeutic regimens can be utilized to take advantage of the doxepin food effects.
  • Affecting Maximum Concentration (Tmax):
  • As discussed above, administration of doxepin without food or in a fasted state can result in shortening the time required to achieve a maximum plasma concentration (Tmax) of doxepin. Achieving a shorter Tmax can be desirable since onset of drug action can be more rapid and the duration of drug action may be shortened. Some embodiments relate to methods of shortening the time required to achieve a maximum plasma concentration or a Tmax of doxepin in a patient receiving doxepin therapy, which methods can include administering to the patient a therapeutically effective amount of doxepin in a pharmaceutical composition without food.
  • Also, it should be noted that other embodiments relate to methods of increasing the time required to achieve a maximum concentration of doxepin in a patient by administering doxepin with food.
  • Some embodiments relate to methods of preventing a doxepin food effect in order to minimize the time required for onset of action of the drug. The methods can include administering doxepin to a patient in need thereof, wherein the patient is in a fasted state or has not eaten or will not eat within a particular time period. The methods further can include providing instructions to take the doxepin without food or in a fasted state.
  • Use of Doxepin in Sleep-Related Indications:
  • Knowledge of the food effect disclosed herein is useful in determining an optimum regimen for providing doxepin sleep therapy. In patients receiving sleep therapy, the onset of action is an important consideration. The studies disclosed herein demonstrate that taking doxepin with food can significantly increase Tmax. As a result, sleep patients who take doxepin without food would be expected to have faster sleep onset or faster drug action in comparison to those who take doxepin with food.
  • Thus, some embodiments relate to improved methods of treating a sleep disorder. The methods can include providing a patient with a therapeutically effective amount of doxepin and providing the patient with instructions to take the doxepin without food. In some aspects, doxepin can be administered without food in order to minimize the amount of time to achieve sleep onset or to otherwise minimize the amount of time before drug action occurs.
  • The information regarding the doxepin food effect can be provided to the patient. The information can include, for example, instructions that may be provided to patients receiving doxepin therapy or health care professionals involved in treatment of those patients that the doxepin should be administered without food, preferably separated from food for the time periods discussed above. By way of example, such instructions could be provided orally or in written form. Some exemplary written forms include a label associated with the drug, on the container for the drug, packaged with the drug, or separately given to the patient apart from the drug, including providing the patient with access to a website or other electronic information with the instructions/information.
  • Affecting Overall Concentration (Cmax) or Bioavailability:
  • The maximum plasma concentration or overall bioavailability of doxepin can be affected by food or a lack thereof. Increasing concentration or bioavailability can be desirable in some circumstances. Some embodiments relate to methods of increasing the oral bioavailability of doxepin to a patient receiving doxepin therapy. The methods can include administering to the patient a pharmaceutical oral dosage form of doxepin with food. The administration can result in a greater AUC than if the drug is taken without food. The methods can include administering to the patient a therapeutically effective amount of doxepin in a pharmaceutical composition with food or within a relatively short time of consuming food (e.g., 15 minutes, 30 minutes, one hour, etc.).
  • Also, some embodiments relate to a method of increasing the maximum plasma concentration of doxepin in a patient receiving doxepin therapy comprising administering to the patient a therapeutically effective amount of doxepin in a pharmaceutical composition with food.
  • It should be noted that other embodiments relate to methods of decreasing the oral bioavailability or AUC of doxepin by administering doxepin without food.
  • Use of Doxepin in Depression and Anxiety
  • Doxepin has been used for several decades in the treatment of depression and anxiety. Several side effects have been reported in connection with the use of doxepin to treat depression or anxiety. The studies disclosed herein show that by taking doxepin with food, AUC was increased by 41% compared to taking doxepin in a fasted state. Because AUC (bioavailability) is increased by taking doxepin with food, a patient can take a lower dose compared to when a patient takes doxepin without food. Increasing the oral bioavailability or AUC of doxepin, and decreasing dosage required for treatment can minimize or alleviate side effects and improve the safety and tolerability of doxepin therapy. Thus, in some aspects it can be preferred that patients receiving doxepin for depression or anxiety should take the drug with food, or in close proximity to eating. Some embodiments relate to improved methods of treating depression and anxiety. The methods can include providing a patient with a therapeutically effective amount of doxepin and providing the patient with instructions to take the doxepin with food. As discussed more fully herein, administering doxepin with food can result in an increase in the bioavailability of doxepin. As a result a patient can take less doxepin, which can be safer and more tolerable for the patient.
  • In some aspects, information, including instructions may be provided to patients receiving doxepin therapy or health care professionals involved in treatment of those patients regarding a doxepin food effect and/or that the doxepin should be administered with food, or at least in relatively close proximity to eating food or eating a meal (for example, within one hour or less). By way of example, such information or instructions could be provided orally or in written form. Some exemplary written forms include a label associated with the drug, on the container for the drug, packaged with the drug, or separately given to the patient apart from the drug, including providing the patient with access to a website or other electronic information with the instructions/information. The invention further includes a package of doxepin with such written instructions associated therewith.
  • It should be noted that some aspects of the invention also relate to methods of treating depression or anxiety by administering doxepin without food. Such embodiments can also include instructions to take the medication without food.
  • Improved Pharmacokinetic Consistency and Efficacy:
  • Still further embodiments relate to methods for improving the consistency of pharmacokinetics associated with doxepin therapy, in which a patient receives a multiple doxepin dosages over multiple days. The methods can include administering the doxepin to the patient in a fixed temporal relationship to food intake by the patient. Also, the method can further include providing instructions to the patient to take the doxepin in a fixed temporal relationship to food intake by the patient. As discussed more fully herein, taking doxepin in fixed or consistent temporal relationship to food can lead to improved safety and tolerability for the patient, for example, due to less variability in the drug kinetics in the patient.
  • Kits and Products:
  • Furthermore, some embodiments relate to kits and products for a therapy that includes the use of doxepin. The kits and products can include doxepin and instructions to take the doxepin without food or in a fasted state, or to take the doxepin with food or within a predetermined period of eating food.
  • The instructions or information regarding a food effect can be provided orally or verbally, or could be in written form. Some exemplary written forms include a label associated with the drug, on the container for the drug, packaged with the drug, or separately given to the patient apart from the drug, including providing the patient with access to a web site or other electronic information with the instructions/information. The invention further includes a package of doxepin with such written instructions associated therewith or with information on where to access the instructions/information (e.g., a website).
  • Administration of Doxepin
  • In performing the methods, doxepin, a pharmaceutically acceptable salt of doxepin, or prodrug of doxepin can be administered in any suitable oral form. Also, doxepin, or a pharmaceutically acceptable salt or a prodrug thereof can be administered to a patient. (It should be understood that the term “administer” and its variants are intended to cover both self-administration and administration by another person or by a device.).
  • Doxepin can be administered without food or in a fasted state. For example, doxepin can be administered at least about 30 minutes to about 4, 5, 6 or more hours after consuming food. More preferably, doxepin can be taken at least about 1 hour to about 6 hours after consuming food. In some aspects doxepin can be taken at least about 1, 2, 3, 4, 5 6 or more hours after consuming food.
  • Also, doxepin can be administered at least about 30 minutes to about 6 hours before consuming any food, or more preferably, at least about 1 hour to about 3 hours before consuming food. In some aspects, doxepin can be administered about 1, 2, 3 or more hours before food is consumed.
  • In some embodiments of the invention, such as when doxepin is used to facilitate sleep, instructions may be provided to patients receiving doxepin therapy or health care professionals involved in treatment of those patients that the doxepin should be administered without food, preferably separated from food for the time periods discussed above. Also, the patient can be provided with information regarding a doxepin food effect. In other embodiments of the invention, such as when doxepin is used to treat depression or anxiety, instructions may be provided to patients receiving doxepin therapy or health care professionals involved in treatment of those patients that the doxepin should be administered with food, or at least in relatively close proximity to eating food or eating a meal (for example, within one hour or less).
  • Again, by way of example, such instructions or information regarding a food effect could be provided orally or verbally, or could be in written form. Some exemplary written forms include a label associated with the drug, on the container for the drug, packaged with the drug, or separately given to the patient apart from the drug, including providing the patient with access to a website or other electronic information with the instructions/information. The invention further includes a package of doxepin with such written instructions associated therewith.
  • It should be recognized that in some cases, the food effect disclosed herein can appropriately play a part in designing customized dosing regimens that reflect the preferences of a particular healthcare professional or their patient. Thus, for example, in doxepin therapy (e.g., chronic doxepin therapy), such as therapy for depression or anxiety, it is customary to titrate the dosage; in other words, typically start with a lower dose and then increase it to the minimum dose that is sufficiently effective for the patient in question. In this type of situation, there may be instances when doxepin is administered without food, even though more drug will be required to achieve the same effect. In such cases, consistency is important to maintaining equivalent pharmacological effects. In other words, if a patient on chronic doxepin therapy is receiving a titrated dose that has been determined when the patient is consistently taking doxepin with food, or consistently taking doxepin without food, then food-induced variations in doxepin pharmacokinetics should not be a factor in therapeutic response or side effects. Thus, for chronic doxepin therapy, one embodiment includes administering doxepin in a consistent relationship to food intake, regardless of whether it is or is not taken with food.
  • Suitable routes of administration of doxepin can include any route in which significant quantities of drug reach the stomach, including oral, buccal, and sublingual administration.
  • For oral administration, the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Administration though oral pathways can be accomplished, for example, using a capsule, a tablet, a granule, a spray, a syrup, a liquid, powder, granules, pastes (e.g., for application to the tongue). Oral administration can be accomplished using fast-melt formulations, for example. For example, rapidly-melting strips or sheets that include the drug and suitable excipients can be prepared that dissolve quickly in the mouth, using well-known formulation technology. For buccal or sublingual administration, the compositions may take any suitable form, for example, tablets or lozenges. Pharmaceutical preparations for oral use can be obtained by mixing one or more solid excipient with pharmaceutical combination of the invention, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Pharmaceutical preparations which can be used orally include for example, liquid solutions, powders, and suspensions in bulk or unit dosage forms. Also, the oral formulations can include, for example, pills, tablets, granules, sprays, syrups, pastes, powders, boluses, pre-measured ampules or syringes, push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
  • A variety of techniques for formulation and administration can be found in Remington: The Science and Practice of Pharmacy (20th ed., Lippincott Williams & Wilkens Publishers (2003)), which is incorporated herein by reference in its entirety.
  • Compositions
  • As mentioned above, doxepin, pharmaceutically acceptable salts, and/or prodrugs of the same can be used alone or in combination with other substances, such as for example, other insomnia or sleep medications, or with other medications that treat a primary illness. Doxepin alone or in combination with other drugs can be included as part of a composition. The compounds and compositions can include any suitable form of the compound for pharmaceutical delivery, as discussed in further detail herein.
  • The compositions and formulations disclosed herein also can include one or more pharmaceutically acceptable carrier materials or excipients. Such compositions can be prepared for storage and for subsequent administration. Acceptable carriers or diluents for therapeutic use are well known in the pharmaceutical art, and are described, for example, in the incorporated material of Remington: The Science and Practice of Pharmacy (20th ed, Lippincott Williams & Wilkens Publishers (2003)), which is incorporated herein by reference in its entirety. The term “carrier” material or “excipient” herein can mean any substance, not itself a therapeutic agent, used as a carrier and/or diluent and/or adjuvant, or vehicle for delivery of a therapeutic agent to a subject or added to a pharmaceutical composition to improve its handling or storage properties or to permit or facilitate formation of a dose unit of the composition into a discrete article such as a capsule or tablet suitable for oral administration. Excipients can include, by way of illustration and not limitation, diluents, disintegrants, binding agents, adhesives, wetting agents, polymers, lubricants, glidants, substances added to mask or counteract a disagreeable taste or odor, flavors, dyes, fragrances, and substances added to improve appearance of the composition. Acceptable excipients include sugars (such as lactose, sucrose, mannitol, sorbitol), starch powder, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, or derivatives thereof, cellulose esters of alkanoic acids, cellulose alkyl esters, talc, stearic acid, magnesium stearate, magnesium oxide, sodium and calcium salts of phosphoric and sulfuric acids, gelatin, acacia gum, sodium alginate, polyvinyl-pyrrolidone (PVP), and/or polyvinyl alcohol, saline, dextrose, lecithin, albumin, sodium glutamate, cysteine hydrochloride, and the like. Examples of suitable excipients for soft gelatin capsules include vegetable oils, waxes, fats, semisolid and liquid polyols. Suitable excipients for the preparation of solutions and syrups include, without limitation, water, polyols, sucrose, invert sugar and glucose. Suitable excipients for injectable solutions include, without limitation, water, alcohols, polyols, glycerol, and vegetable oils. If desired, the compositions can include disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. The pharmaceutical compositions can additionally include preservatives, solubilizers, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavorings, buffers, coating agents, or antioxidants. Compositions for oral administration can be formulated according to conventional pharmaceutical practice as described in the incorporated material in Remington: The Science and Practice of Pharmacy (20th ed, Lippincott Williams & Wilkens Publishers (2003)). For example, dissolution or suspension of the active compound in a vehicle such as water or naturally occurring vegetable oil like sesame, peanut, or cottonseed oil or a synthetic fatty vehicle like ethyl oleate or the like may be desired. Buffers, preservatives, antioxidants and the like can be incorporated according to accepted pharmaceutical practice. The compound can also be made in microencapsulated form.
  • One can also administer the compounds of the invention in sustained release forms or from sustained release drug delivery systems. A description of representative sustained release materials can be found in the incorporated materials in Remington: The Science and Practice of Pharmacy (20th ed, Lippincott Williams & Wilkens Publishers (2003)).
  • Dosage
  • The selected dosage level can depend upon, for example, the condition being treated, the route of administration, the severity of the condition being treated, and the condition and prior medical history of the patient being treated. However, it is within the skill of the art to start doses of the compound at levels lower than required to achieve the desired therapeutic effect and to gradually increase the dosage until the desired effect is achieved. It will be understood, however, that the specific dose level for any particular patient can depend upon a variety of factors including the genetic makeup, body weight, general health, diet, time and route of administration, combination with other drugs and the particular condition being treated, and its severity. For the treatment of insomnia, preferably one dose is administered prior to bedtime.
  • As mentioned above, in some embodiments the preferable dosage can be between about 0.001 milligrams and about 350 milligrams. In some aspects, the dosage can be about 50 milligrams to about 350 milligrams. More preferably, the dosage can be between about 75 milligrams and 300 milligrams. Also, in some aspects, the dosage can be between about 0.1 milligrams and 20 milligrams or between about 0.5 milligrams and 10 milligrams. The dosage also can be between about 1 milligram and about 6 milligrams. Preferably, the dosage can be about 0.5 milligrams, 1 milligram, about 2 milligrams, about 3 milligrams, about 4 milligrams, about 5 milligrams or about 6 milligrams. Further, the dosage can be about 7 milligrams, about 8 milligrams, about 9 milligrams, or about 10 milligrams. The lower dosage ranges are particularly desirable for sleep-related indications, while the higher dosage ranges are particularly desirable for depression and anxiety-related indications.
  • EXAMPLES Example 1 Assessment of the Effect of Food on the Pharmacokinetics of Doxepin
  • A study assessed the effect of food on the pharmacokinetics (PK) of doxepin in healthy subjects. It was a single-center, phase 1, randomized, open-label, single dose, two-way crossover study conducted in sixteen healthy, young adult male and female subjects. Screening procedures were performed within 14 days prior to enrollment.
  • Following screening procedures and baseline assessments, eligible subjects were randomly assigned to one of two treatment sequences (fed-fasted or fasted-fed). Subjects received a single 6 milligram dose of doxepin in the morning under either fed or fasted conditions on 2 dosing days (Day 1 and Day 8). There were approximately 7 days between each dose. Enrolled subjects were admitted to the study center on the evening before study drug dosing (Day 0 and Day 7) and remained at the study center for approximately 5 days. All subjects were dosed under both fed and fasted conditions during the study as illustrated in FIG. 1.
  • Subjects being dosed under fasted conditions were required to fast overnight for at least 10 hours prior to study drug administration and for 4 hours after study drug administration. Fluids were restricted from 1 hour predose to 1 hour postdose, except for water taken at the time of dosing. Subjects being dosed under fed conditions were dosed approximately 5 minutes after eating a high-fat, high-calorie standardized breakfast (to be ingested within 25 minutes). Subjects were required to ingest the entire contents of the breakfast. All subjects were required to remain in bed for approximately 4 hours after dosing.
  • Contents of the high-fat, high-calorie standardized breakfast were:
  • Two eggs fried in butter;
  • Two slices of bacon;
  • 240 mL (8 fl. oz) whole milk;
  • 57 g (2 oz) of hash browned potatoes; and
  • Two slices of toasted white bread with butter.
  • The total amount of protein, fat, and carbohydrate that made up this meal was approximately 33, 55, and 58 g, respectively. The total calorie content was approximately 850 kcal.
  • Subjects were dosed on Day 1 and Day 8. The PK profiles were evaluated on Days 1 through 5 and Days 8 through 12. Blood samples were collected at predose (0 hour) and at 0.08, 0.17, 0.25, 0.5, 0.75, 1, 1.5, 2, 3, 4, 6, 8, 12, 24, 36, 48, 60, 72, and 96 hours postdose. The samples were analyzed for doxepin and doxepin metabolite concentrations in plasma. Plasma concentrations of doxepin were measured using validated high performance liquid chromatography coupled to tandem mass spectrometry. The lower limit of quantification for doxepin was 0.05 ng/mL. The following PK parameters were estimated by noncompartmental methods using actual elapsed time from dosing:
    • Cmax (ng/mL) Maximum observed plasma concentration, obtained directly from the observed concentration versus time data.
    • Tmax (h) Time to maximum plasma concentration, obtained directly from the observed concentration versus time data.
    • AUC0-∞_9 (ng·h/mL) Area under the curve from time zero extrapolated to infinity, calculated by linear up/log down trapezoidal summation and extrapolated to infinity by addition of the last quantifiable concentration divided by the elimination rate constant (AUC0-Tlast+ClastZ). If the extrapolated area (ClastZ) was greater than 30% of AUC0-∞, then AUC0-∞ was set to missing.
    • AUC0-Tlast (ng·h/mL) Area under the curve from time zero to time of last measurable concentration, calculated by linear up/log down trapezoidal summation.
    • AUC0-24 (ng·h/mL) Area under the curve from time zero until 24 hours, calculated by linear up/log down trapezoidal summation. If the 24 h sample was missing or below the lower limit of quantification, AUC0-Tlast was to be reported as AUC0-24.
    • AUC0-48 (ng·h/mL) Area under the curve from time zero until 48 hours, calculated by linear up/log down trapezoidal summation. If the 48 h sample was missing or below the lower limit of quantification, AUC0-Tlast was to be reported as AUC0-48.
    • AUC0-72 (ng·h/mL) Area under the curve from time zero until 72 hours, calculated by linear up/log down trapezoidal summation. If the 72 h sample was missing or below the lower limit of quantification, AUC0-Tlast was to be reported as AUC0-72.
    • AUC0-96 (ng·h/mL) Area under the curve from time zero until 96 hours, calculated by linear up/log down trapezoidal summation. If the 96 h sample was missing or below the lower limit of quantification, AUC0-Tlast was to be reported as AUC0-96.
    • λz (1/h) Elimination rate constant associated with the terminal (log-linear) portion of the curve. This was estimated via linear regression of time versus log concentration. Visual assessment was used to identify the terminal linear phase of the concentration-time profile. A minimum of three data points were used for determination.
    • t1/2 (h) Apparent terminal half-life, determined as ln 2/λz.
    • CL/F (L/h) Apparent oral clearance, calculated as dose divided by AUC0-∞.
    • Vd/F (L) Apparent volume of distribution, calculated as (CL/F)/λz.
    Concentration-Time Profiles
  • With reference to Table 1, following a single 6 milligram dose administration of doxepin in fasted state (Treatment A) and fed state (Treatment B), mean plasma concentrations of doxepin increased, reaching Cmax at 3.0 and 6.0 hours postdose, respectively. Doxepin plasma concentrations, reached mean Cmax values of 0.854 and 0.951 ng/mL in fasted and fed states, respectively. For both treatments, plasma doxepin concentrations declined thereafter and remained quantifiable up to 48 hours postdose (the lower limit of quantification was 0.05 ng/mL). Detectable plasma levels of doxepin were first observed at 30 minutes postdose in both the fed and fasted states (six and five subjects, respectively). All subjects had detectable concentrations of doxepin by 90 minutes postdose. Median Tmax was delayed by approximately 3.0 hours in the fed state (6.0 hours) as compared to the fasted state (3.0 hours). However, the range of values was similar for both treatments. Mean t1/2 values were comparable for fed and fasted states (16.5 versus 14.4 hours, respectively).
  • TABLE 1
    Summary Statistics [arithmetic mean (CV %)] of Plasma
    Doxepin Pharmacokinetic Parameters
    Treatment [a]
    Parameter (unit) Fed (N = 16) Fasted (N = 15)
    Cmax (ng/mL) 0.951 (58.8)  0.854 (63.2) 
    Tmax (h) [b]   6.0 (2.0-6.0)   3.0 (1.5-6.0)
    AUC0-∞ (ng · h/mL) 18.6 (70.2) 14.1 (80.6)
    AUC0-Tlast (ng · h/mL) 16.8 (74.0) 12.6 (85.7)
    t1/2 (h) 16.5 (23.8) 14.4 (42.2)
    λz (1/h) 0.0445 (26.6)  0.0623 (65.9) 
    [a] 6 milligram doxepin tablet, under fed or fasted conditions.
    [b] Indicates median (range) values.
  • Table 2 shows the estimates of clearance and volume of distribution for doxepin. Mean CL/F and Vd/F were 43% and 14% lower in the fed state compared to the fasted state, respectively. Mean CL/F values were lower in the fed (477 L/h) versus the fasted (837 L/h) states. Mean Vd/F remained almost unchanged for fed (10280 L) and fasted (11930 L) states.
  • TABLE 2
    Summary Statistics [arithmetic mean (CV %)] of Mean
    Clearance and Volume of Distribution for Doxepin
    Treatment [a]
    Parameter (unit) Fed (N = 16) Fasted (N = 15)
    CL/F (L/h)  477 (63.4)   837 (114.3)
    Vd/F (L) 10280 (43.3) 11930 (46.9)
    [a] 6 milligram doxepin tablet, under fed or fasted conditions.
  • The effect of a high-fat meal on the pharmacokinetics of the 6 milligram doxepin tablet was assessed and statistical comparisons of doxepin pharmacokinetic parameters between treatments are presented in Table 3.
  • The 90% confidence intervals for the ratio of population geometric least-square means between fed and fasted treatments was not completely contained within the equivalence limits of 80-125% for Cmax, and were outside the equivalence limits for AUC0-∞ and AUC0-Tlast, indicating that there was a food effect on exposure. Under fed conditions, AUC0-∞, AUC0-Tlast, and Cmax were higher by 41%, 46%, and 15%, respectively, compared to fasted conditions.
  • TABLE 3
    Statistical Comparison of Doxepin Pharmacokinetic Parameters Between Treatments
    Treatment Geometric Pairwise Comparisons
    Parameter (unit) [a] N LS Mean Pair Ratio (%) 90% CI
    AUC0-∞ (ng · h/mL) Fed 16 15.14
    Fasted 15 10.72 Fed/Fasted 141.3 (124.7, 160.1)
    AUC0-Tlast (ng · h/mL) Fed 16 13.39
    Fasted 15 9.194 Fed/Fasted 145.6 (127.0, 166.9)
    Cmax (ng/mL) Fed 16 0.822
    Fasted 15 0.717 Fed/Fasted 114.6 (101.8, 129.1)
    Note:
    Results are based on mixed effect analysis of variance with sequence, period and treatment as fixed effects and subject within sequence as a random effect.
    [a] 6 milligram doxepin tablet, under fed or fasted conditions.
  • CONCLUSIONS
  • Following a single 6 milligram dose of doxepin, a food effect was observed on the pharmacokinetics of doxepin. In the fed state, the maximum concentration (Cmax) and exposure (AUC0-∞) of doxepin were approximately 15% and 41% higher, respectively, compared to the fasted state, and median Tmax was delayed by approximately 3.0 hours. The increase in exposure was statistically significant and represents an increase in bioavailability in the fed state (Table 4).
  • TABLE 4
    Arithmetic Arithmetic Effect of Food
    Mean Mean (% of
    Parameter (Fed condition) (Fasted condition) fasted state)a
    AUC0-∞ (ng · h/mL) 18.6 14.1 141
    Cmax (ng/mL) 0.951 0.854 115
    Tmax (h)b 6 3 200
    aNote that percentages documenting food effect on Cmax and AUC are calculated from geometric LS means rather than the arithmetic means presented in this table
    bTmax is presented as a median value
  • Many modifications and variations of the embodiments described herein may be made without departing from the scope, as is apparent to those skilled in the art. The specific embodiments described herein are offered by way of example only.

Claims (20)

1. A method of treating a sleep disorder, comprising:
selecting a patient with insomnia;
providing between about 0.5 mg and 7 mg doxepin to the patient; and
administering the doxepin to the patient without food.
2. The method of claim 1, wherein the administering without food comprises administering the doxepin at least about 30 minutes after consuming food.
3. The method of claim 1, wherein administering without food comprises administering the doxepin at about 1 hour prior to consuming food.
4. The method of claim 1, wherein the doxepin is provided in an amount of about 3 milligrams.
5. The method of claim 1 wherein the doxepin is provided in an amount of about 6 milligrams.
6. The method of claim 1 wherein the doxepin is provided as a tablet, capsule or liquid.
7. The method of claim 1, wherein the doxepin is provided as a unit dosage form.
8. The method of claim 1, wherein the doxepin is provided as an oral dosage form.
9. In a method of treating insomnia with doxepin, wherein an insomnia patient receives a dosage of between 0.5 and 7 mg of doxepin, the improvement altering the pharmacokinetics of doxepin by providing the patient with instructions to take the doxepin without food, and administering the doxepin without food, thereby decreasing the time to reach maximum plasma concentration (Tmax) in the patient, reducing Cmax, and reducing the area under the curve (AUC compared to administering the doxepin with food.
10. The method of claim 9, wherein decreasing the time to reach maximum plasma concentration (Tmax) in the patient, reducing Cmax, and/or reducing the area under the curve (AUC) shortens the time required to achieve sleep onset and/or reduces next-day sedation effects.
11. The method of claim 9, wherein the instructions indicate a dosage of 6 milligrams.
12. The method of claim 9, wherein the doxepin is provided as a tablet, capsule or liquid.
13. The method of claim 9, wherein the doxepin is provided as a unit dosage form.
14. The method of claim 9, wherein the doxepin is provided as an oral dosage form.
15. A method of treating a sleep disorder with doxepin while achieving a desired alteration in doxepin pharmacokinetics, comprising:
identifying a patient with insomnia;
providing between 0.5 mg and 7 mg doxepin to the patient;
instructing the patient to take the doxepin without food; and
administering the doxepin to the patient without food, thereby decreasing the time to reach a maximum plasma concentration (Tmax) in the patient, reducing Cmax, and reducing the area under the curve (AUC), compared to administering the doxepin with food.
16. The method of claim 15, decreasing the time to reach maximum plasma concentration (Tmax) in the patient, reducing Cmax, and/or reducing the area under the curve (AUC) shortens the time required to achieve sleep onset and/or reduces next-day sedation effects.
17. The method of claim 15, wherein the therapeutically effective amount is about 6 milligrams.
18. The method of claim 15, wherein the doxepin is provided as a tablet, capsule or liquid.
19. The method of claim 15, wherein the doxepin is provided as a unit dosage form.
20. The method of claim 15, wherein the doxepin is provided as an oral dosage form.
US13/007,334 2006-07-20 2011-01-14 Methods of improving the pharmacokinetics of doxepin Abandoned US20110166215A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
US13/007,334 US20110166215A1 (en) 2006-07-20 2011-01-14 Methods of improving the pharmacokinetics of doxepin
US13/653,213 US9572814B2 (en) 2006-07-20 2012-10-16 Methods of improving the pharmacokinetics of doxepin
US15/436,293 US10653660B2 (en) 2006-07-20 2017-02-17 Methods of improving the pharmacokinetics of doxepin
US16/876,492 US11110074B2 (en) 2006-07-20 2020-05-18 Methods of improving the pharmacokinetics of doxepin
US17/467,360 US20210393567A1 (en) 2006-07-20 2021-09-06 Methods Of Improving The Pharmacokinetics Of Doxepin

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US83272706P 2006-07-20 2006-07-20
US83361706P 2006-07-24 2006-07-24
US11/781,165 US7915307B2 (en) 2006-07-20 2007-07-20 Methods of improving the pharmacokinetics of doxepin
US13/007,334 US20110166215A1 (en) 2006-07-20 2011-01-14 Methods of improving the pharmacokinetics of doxepin

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/781,165 Continuation US7915307B2 (en) 2006-07-20 2007-07-20 Methods of improving the pharmacokinetics of doxepin

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/653,213 Continuation US9572814B2 (en) 2006-07-20 2012-10-16 Methods of improving the pharmacokinetics of doxepin

Publications (1)

Publication Number Publication Date
US20110166215A1 true US20110166215A1 (en) 2011-07-07

Family

ID=38658703

Family Applications (6)

Application Number Title Priority Date Filing Date
US11/781,165 Active 2027-08-24 US7915307B2 (en) 2006-07-20 2007-07-20 Methods of improving the pharmacokinetics of doxepin
US13/007,334 Abandoned US20110166215A1 (en) 2006-07-20 2011-01-14 Methods of improving the pharmacokinetics of doxepin
US13/653,213 Active US9572814B2 (en) 2006-07-20 2012-10-16 Methods of improving the pharmacokinetics of doxepin
US15/436,293 Active US10653660B2 (en) 2006-07-20 2017-02-17 Methods of improving the pharmacokinetics of doxepin
US16/876,492 Active US11110074B2 (en) 2006-07-20 2020-05-18 Methods of improving the pharmacokinetics of doxepin
US17/467,360 Pending US20210393567A1 (en) 2006-07-20 2021-09-06 Methods Of Improving The Pharmacokinetics Of Doxepin

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/781,165 Active 2027-08-24 US7915307B2 (en) 2006-07-20 2007-07-20 Methods of improving the pharmacokinetics of doxepin

Family Applications After (4)

Application Number Title Priority Date Filing Date
US13/653,213 Active US9572814B2 (en) 2006-07-20 2012-10-16 Methods of improving the pharmacokinetics of doxepin
US15/436,293 Active US10653660B2 (en) 2006-07-20 2017-02-17 Methods of improving the pharmacokinetics of doxepin
US16/876,492 Active US11110074B2 (en) 2006-07-20 2020-05-18 Methods of improving the pharmacokinetics of doxepin
US17/467,360 Pending US20210393567A1 (en) 2006-07-20 2021-09-06 Methods Of Improving The Pharmacokinetics Of Doxepin

Country Status (3)

Country Link
US (6) US7915307B2 (en)
CA (1) CA2693992C (en)
WO (1) WO2008011150A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070281990A1 (en) * 2006-05-19 2007-12-06 Rogowski Roberta L Methods of using low-dose doxepin for the improvement of sleep
US20080182890A1 (en) * 2006-10-04 2008-07-31 Somaxon Pharmaceuticals, Inc. Methods of using low-dose doxepin for the improvement of sleep
US20090074862A1 (en) * 2007-04-13 2009-03-19 Luigi Schioppi Low-dose doxepin formulations and methods of making and using the same
US20100105614A1 (en) * 2006-10-25 2010-04-29 Somaxon Pharmaceuticals, Inc. Ultra low dose doxepin and methods of using the same to treat sleep disorders
US20100179215A1 (en) * 2006-05-19 2010-07-15 Somaxon Pharmaceuticals, Inc. Doxepin isomers and isomeric mixtures and methods of using the same to treat sleep disorders
US20100179214A1 (en) * 2006-05-19 2010-07-15 Somaxon Pharmaceuticals, Inc. Doxepin trans isomers and isomeric mixtures and methods of using the same to treat sleep disorders
US20100227916A1 (en) * 2006-05-19 2010-09-09 Somaxon Pharmaceuticals, Inc N-desmethyl-doxepin and methods of using the same to treat sleep disorders
US20110077200A1 (en) * 2006-12-06 2011-03-31 Somaxon Pharmaceuticals, Inc. Combination therapy using low-dose doxepin for the improvement of sleep
US9572814B2 (en) 2006-07-20 2017-02-21 Pernix Sleep, Inc. Methods of improving the pharmacokinetics of doxepin
US11234954B2 (en) 2006-05-19 2022-02-01 Currax Pharmaceuticals Llc Low-dose doxepin for treatment of sleep disorders in elderly patients

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2760464T3 (en) 2003-04-29 2020-05-14 Nalpropion Pharmaceuticals Inc Compositions to affect weight loss
WO2007067341A2 (en) * 2005-11-22 2007-06-14 Orexigen Therapeutics, Inc. Compositions and methods for increasing insulin sensitivity
WO2007089318A2 (en) * 2005-11-23 2007-08-09 Orexigen Therapeutics, Inc. Compositions and methods for reducing food cravings
US8916195B2 (en) * 2006-06-05 2014-12-23 Orexigen Therapeutics, Inc. Sustained release formulation of naltrexone
US10095836B2 (en) 2006-09-29 2018-10-09 Gearbox Llc Computational systems for biomedical data
US10546652B2 (en) 2006-09-29 2020-01-28 Gearbox Llc Computational systems for biomedical data
US10503872B2 (en) 2006-09-29 2019-12-10 Gearbox Llc Computational systems for biomedical data
US20080082359A1 (en) * 2006-09-29 2008-04-03 Searete Llc, A Limited Liability Corporation Of State Of Delaware Computational systems for biomedical data
US10068303B2 (en) 2006-09-29 2018-09-04 Gearbox Llc Computational systems for biomedical data
KR20160072276A (en) * 2006-11-09 2016-06-22 오렉시젠 세러퓨틱스 인크. Unit dosage packages
EP2089005B1 (en) 2006-11-09 2010-03-17 Orexigen Therapeutics, Inc. Layered pharmaceutical formulations comprising an intermediate rapidly dissolving layer
EP2303025A4 (en) * 2008-05-30 2012-07-04 Orexigen Therapeutics Inc Methods for treating visceral fat conditions
KR20120124423A (en) * 2010-01-11 2012-11-13 오렉시젠 세러퓨틱스 인크. Methods of providing weight loss therapy in patients with major depression
RS62149B1 (en) * 2010-12-03 2021-08-31 Nalpropion Pharmaceuticals Llc Increasing drug bioavailability in naltrexone therapy
PT2806863T (en) 2012-01-26 2017-11-09 Vanda Pharmaceuticals Inc Treatment of circadian rhythm disorders
US11918557B2 (en) 2012-01-26 2024-03-05 Vanda Pharmaceuticals Inc. Treatment of circadian rhythm disorders
IL300868A (en) 2012-06-06 2023-04-01 Orexigen Therapeutics Inc Methods of treating overweight and obesity
US11090285B2 (en) 2013-11-12 2021-08-17 Vanda Pharmaceuticals Inc Treatment of circadian rhythm disorders
US10376487B2 (en) 2013-11-12 2019-08-13 Vanda Pharmaceuticals Inc. Method of treatment
US8969371B1 (en) 2013-12-06 2015-03-03 Orexigen Therapeutics, Inc. Compositions and methods for weight loss in at risk patient populations
CN108096363A (en) * 2018-01-31 2018-06-01 常州康普药业有限公司 A kind of doxepin hydrochloride piece and preparation method thereof

Citations (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3420851A (en) * 1962-12-19 1969-01-07 Pfizer & Co C Novel dibenzoxepines
US3438981A (en) * 1963-07-09 1969-04-15 Boehringer & Soehne Gmbh Dibenzooxepine and dibenzothiepine derivatives
US3509175A (en) * 1963-10-14 1970-04-28 Pfizer & Co C Recovery of pure cis 11-(3-dimethylaminopropylidene) - 6,11 - dihydrodibenz(b,e)oxepine from admixture with its trans isomer
US4110438A (en) * 1977-03-21 1978-08-29 American Home Products Corporation Method of treating depression
US4434171A (en) * 1980-11-28 1984-02-28 Sandoz Ltd. Dibenzazepine derivatives, pharmaceutical compositions containing them, and pharmaceutical methods using them
US5030632A (en) * 1986-09-23 1991-07-09 Sandoz Pharm. Corp. Low dose temazepam
US5116852A (en) * 1990-12-03 1992-05-26 Bristol-Myers Squibb Co. Treatment of sleep disorders
US5332661A (en) * 1991-07-31 1994-07-26 Abbott Laboratories Reagents and methods for the quantification of total doxepins in biological fluids
US5502047A (en) * 1993-03-22 1996-03-26 Kavey; Neil B. Treatment for insomnia
US5585115A (en) * 1995-01-09 1996-12-17 Edward H. Mendell Co., Inc. Pharmaceutical excipient having improved compressability
US5733578A (en) * 1995-11-15 1998-03-31 Edward Mendell Co., Inc. Directly compressible high load acetaminophen formulations
US5741524A (en) * 1995-01-09 1998-04-21 Edward Mendell Co., Inc. Sustained-release formulations utilizing pharmaceutical excipient having improved compressibility
US5948438A (en) * 1995-01-09 1999-09-07 Edward Mendell Co., Inc. Pharmaceutical formulations having improved disintegration and/or absorptivity
US6106865A (en) * 1995-01-09 2000-08-22 Edward Mendell Co., Inc. Pharmaceutical excipient having improved compressibility
US6211229B1 (en) * 2000-02-17 2001-04-03 Neil B. Kavey Treatment of transient and short term insomnia
US6219674B1 (en) * 1999-11-24 2001-04-17 Classen Immunotherapies, Inc. System for creating and managing proprietary product data
US6344487B1 (en) * 2000-06-05 2002-02-05 Neil B. Kavey Treatment of insomnia
US20020037828A1 (en) * 1997-10-28 2002-03-28 Wilson Leland F. Administration of phosphodiesterase inhibitors for the treatment of premature ejaculation
US6391337B2 (en) * 1995-11-15 2002-05-21 Edward Mendell Co., Inc. Directly compressible high load acetaminophen formulations
US6395303B1 (en) * 1996-06-10 2002-05-28 Edward Mendell Co., Inc. Process for preparing a directly compressible solid dosage form containing microcrystalline cellulose
US6407128B1 (en) * 2001-12-03 2002-06-18 Elan Pharmaceuticals, Inc. Method for increasing the bioavailability of metaxalone
US6471994B1 (en) * 1995-01-09 2002-10-29 Edward Mendell Co., Inc. Pharmaceutical excipient having improved compressibility
US20020197235A1 (en) * 2000-11-03 2002-12-26 Moran Stanford Mark Method for short-term and long-term drug dosimetry
US20030206978A1 (en) * 2001-11-29 2003-11-06 Bob Sherwood Agglomerated particles including an active agent coprocessed with silicified microcrystalline cellulose
US20030235617A1 (en) * 2002-02-07 2003-12-25 Martino Alice C. Pharmaceutical dosage form for mucosal delivery
US20040063721A1 (en) * 2002-08-15 2004-04-01 Wyeth Agonism of the 5HT2A receptor for treatment of thermoregulatory dysfunction
US20040115142A1 (en) * 2002-09-05 2004-06-17 Jrs Pharma Lp Compositions for industrial applications
US6852336B2 (en) * 1995-11-15 2005-02-08 J. Rettenmaier & Soehne Gmbh + Co. Kg Directly compressible high load acetaminophen formulations
US20050118261A1 (en) * 2003-06-12 2005-06-02 Oien Hal J. Compositions and methods of administering doxepin to mucosal tissue
US20050123609A1 (en) * 2001-11-30 2005-06-09 Collegium Pharmaceutical, Inc. Pharmaceutical composition for compressed annular tablet with molded triturate tablet for both intraoral and oral administration
US20050171160A1 (en) * 2003-12-10 2005-08-04 Edgar Dale M. Doxepin analogs and methods of use thereof
US6936277B2 (en) * 1995-01-09 2005-08-30 J. Rettenmaier & Soehne Gmbh & Co. Kg Pharmaceutical excipient having improved compressibility
US20050196439A1 (en) * 2003-10-24 2005-09-08 J. Rettenmaier & Soehne Gmbh + Co. Kg Process for co-spray drying agents with dry silicified MCC
US20050214365A1 (en) * 2004-03-24 2005-09-29 Gulf Pharmaceutical Industries [Instant dissolving tablet composition for loratidine and desloratidine]
US20050239838A1 (en) * 2004-04-23 2005-10-27 Dale Edgar Methods of treating sleep disorders
US20050256165A1 (en) * 2003-12-10 2005-11-17 Edgar Dale M Doxepin analogs and methods of use thereof
US20060228487A1 (en) * 2005-04-11 2006-10-12 J. Rettenmaier & Söehne GmbH + Co. KG Methods of combining active agents with augmented microcrystalline cellulose
US7135196B2 (en) * 2000-09-19 2006-11-14 Vitra Pharmaceuticals Limited Iron compositions
US7179488B2 (en) * 2001-11-29 2007-02-20 Bob Sherwood Process for co-spray drying liquid herbal extracts with dry silicified MCC
US7276536B2 (en) * 2003-03-17 2007-10-02 Japan Tobacco Inc. Method for increasing the bioavailability of the active form of S-[2-([[1-(2-ethylbutyl)cyclohexyl]carbonyl]amino) phenyl] 2-methylpropanethioate
US20070281990A1 (en) * 2006-05-19 2007-12-06 Rogowski Roberta L Methods of using low-dose doxepin for the improvement of sleep
US20080058407A1 (en) * 2006-07-20 2008-03-06 Cara Baron Methods of improving the pharmacokinetics of doxepin
US20080054408A1 (en) * 2006-08-31 2008-03-06 Kimberly-Clark Worldwide, Inc. Conduction through a flexible substrate in an article
US20080182890A1 (en) * 2006-10-04 2008-07-31 Somaxon Pharmaceuticals, Inc. Methods of using low-dose doxepin for the improvement of sleep
US7452872B2 (en) * 2005-08-24 2008-11-18 Salix Pharmaceuticals, Inc. Formulations and uses of 2-hydroxy-5-phenylazobenzoic acid derivatives
US20090042972A1 (en) * 2006-05-19 2009-02-12 Somaxon Pharmaceuticals, Inc. Methods of using low-dose doxepin for the improvement of sleep
US20090042971A1 (en) * 2006-05-19 2009-02-12 Somaxon Pharmaceuticals, Inc. Method of using low-dose doxepin for the improvement of sleep
US20090074862A1 (en) * 2007-04-13 2009-03-19 Luigi Schioppi Low-dose doxepin formulations and methods of making and using the same
US20100105614A1 (en) * 2006-10-25 2010-04-29 Somaxon Pharmaceuticals, Inc. Ultra low dose doxepin and methods of using the same to treat sleep disorders
US20100179214A1 (en) * 2006-05-19 2010-07-15 Somaxon Pharmaceuticals, Inc. Doxepin trans isomers and isomeric mixtures and methods of using the same to treat sleep disorders
US20100179215A1 (en) * 2006-05-19 2010-07-15 Somaxon Pharmaceuticals, Inc. Doxepin isomers and isomeric mixtures and methods of using the same to treat sleep disorders
US20100227916A1 (en) * 2006-05-19 2010-09-09 Somaxon Pharmaceuticals, Inc N-desmethyl-doxepin and methods of using the same to treat sleep disorders
US20110077200A1 (en) * 2006-12-06 2011-03-31 Somaxon Pharmaceuticals, Inc. Combination therapy using low-dose doxepin for the improvement of sleep
US20110318412A1 (en) * 2006-05-19 2011-12-29 Somaxon Pharmaceuticals, Inc. Low dose doxepin formulations, including buccal, sublingual and fastmelt formulations, and uses of the same to treat insomnia

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4833154A (en) 1987-12-14 1989-05-23 Jean Louis Louis J Natural sleeping pill to prevent and alleviate insomnia
SE9601438D0 (en) 1996-04-16 1996-04-16 Tetra Laval Holdings & Finance plate heat exchangers
AU2406299A (en) 1998-02-12 1999-08-30 Centrapharm Inc. Sublingual drug formulations having combined rapid onset of action and long lasting therapeutic effect
CA2341400A1 (en) 1998-08-25 2000-03-02 Sepracor, Inc. Methods and compositions employing optically pure s(+) vigabatrin
GB9904163D0 (en) 1999-02-23 1999-04-14 Bioglan Lab Ltd Pharmaceutical compositions
WO2003004009A1 (en) 2001-07-02 2003-01-16 Geneva Pharmaceuticals, Inc. Pharmaceutical composition
WO2004082615A2 (en) 2003-03-14 2004-09-30 Nirmal Mulye A process for preparing sustained release tablets
PT1691811E (en) 2003-12-11 2014-10-30 Sunovion Pharmaceuticals Inc Combination of a sedative and a neurotransmitter modulator, and methods for improving sleep quality and treating depression
US7280750B2 (en) 2005-10-17 2007-10-09 Watlow Electric Manufacturing Company Hot runner nozzle heater and methods of manufacture thereof
US20080058408A1 (en) 2006-05-19 2008-03-06 Rogowski Roberta L Low-dose doxepin for treatment of sleep disorders in elderly patients

Patent Citations (74)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3420851A (en) * 1962-12-19 1969-01-07 Pfizer & Co C Novel dibenzoxepines
US3438981A (en) * 1963-07-09 1969-04-15 Boehringer & Soehne Gmbh Dibenzooxepine and dibenzothiepine derivatives
US3509175A (en) * 1963-10-14 1970-04-28 Pfizer & Co C Recovery of pure cis 11-(3-dimethylaminopropylidene) - 6,11 - dihydrodibenz(b,e)oxepine from admixture with its trans isomer
US4110438A (en) * 1977-03-21 1978-08-29 American Home Products Corporation Method of treating depression
US4434171A (en) * 1980-11-28 1984-02-28 Sandoz Ltd. Dibenzazepine derivatives, pharmaceutical compositions containing them, and pharmaceutical methods using them
US5030632A (en) * 1986-09-23 1991-07-09 Sandoz Pharm. Corp. Low dose temazepam
US5116852A (en) * 1990-12-03 1992-05-26 Bristol-Myers Squibb Co. Treatment of sleep disorders
US5332661A (en) * 1991-07-31 1994-07-26 Abbott Laboratories Reagents and methods for the quantification of total doxepins in biological fluids
US5643897A (en) * 1993-03-22 1997-07-01 Kavey; Neil B. Treatment for insomnia
US5502047A (en) * 1993-03-22 1996-03-26 Kavey; Neil B. Treatment for insomnia
US6521261B2 (en) * 1995-01-09 2003-02-18 Edward Mendell Co., Inc. Pharmaceutical excipient having improved compressibility
US20060008522A1 (en) * 1995-01-09 2006-01-12 J. Rettenmaier & Soehne Gmbh + Co. Kg Pharmaceutical excipient having improved compressibility
US5725883A (en) * 1995-01-09 1998-03-10 Edward Mendell Co., Inc. Pharmaceutical excipient having improved compressibility
US6858231B2 (en) * 1995-01-09 2005-02-22 J. Rettenmaier & Soehne Gmbh + Co. Kg Pharmaceutical excipient having improved compressibility
US5741524A (en) * 1995-01-09 1998-04-21 Edward Mendell Co., Inc. Sustained-release formulations utilizing pharmaceutical excipient having improved compressibility
US5858412A (en) * 1995-01-09 1999-01-12 Edward Mendell Co., Inc. Sustained release formulations utilizing pharmaceutical excipient having improved compressibility with modified microcrystalline
US5866166A (en) * 1995-01-09 1999-02-02 Edward Mendell Co., Inc. Pharmaceutical excipient having improved compressibility
US5948438A (en) * 1995-01-09 1999-09-07 Edward Mendell Co., Inc. Pharmaceutical formulations having improved disintegration and/or absorptivity
US20050013861A1 (en) * 1995-01-09 2005-01-20 J. Rettenmaier & Soehne Gmbh + Co. Kg Pharmaceutical excipient having improved compressibility
US6103219A (en) * 1995-01-09 2000-08-15 Edward Mendell Co., Inc. Pharmaceutical excipient having improved compressibility
US6106865A (en) * 1995-01-09 2000-08-22 Edward Mendell Co., Inc. Pharmaceutical excipient having improved compressibility
US20040265374A1 (en) * 1995-01-09 2004-12-30 J. Rettenmaier & Soehne Gmbh + Co. Kg Pharmaceutical exipient having improved compressibility
US6936277B2 (en) * 1995-01-09 2005-08-30 J. Rettenmaier & Soehne Gmbh & Co. Kg Pharmaceutical excipient having improved compressibility
US6746693B2 (en) * 1995-01-09 2004-06-08 J. Rettenmaier & Soehne Gmbh + Co. Kg Pharmaceutical excipient having improved compressibility
US6217909B1 (en) * 1995-01-09 2001-04-17 Edward Mendell Co., Inc. Pharmaceutical excipient having improved compressibility
US5725884A (en) * 1995-01-09 1998-03-10 Edward Mendell Co., Inc. Pharmaceutical excipient having improved compressibility
US6358533B2 (en) * 1995-01-09 2002-03-19 Edward Mendell, Co., Inc. Pharmaceutical excipient having improved compressibility
US5585115A (en) * 1995-01-09 1996-12-17 Edward H. Mendell Co., Inc. Pharmaceutical excipient having improved compressability
US6471994B1 (en) * 1995-01-09 2002-10-29 Edward Mendell Co., Inc. Pharmaceutical excipient having improved compressibility
US6217907B1 (en) * 1995-11-15 2001-04-17 Edward Mendell Co., Inc. Directly compressible high load acetaminophen formulations
US6391337B2 (en) * 1995-11-15 2002-05-21 Edward Mendell Co., Inc. Directly compressible high load acetaminophen formulations
US5733578A (en) * 1995-11-15 1998-03-31 Edward Mendell Co., Inc. Directly compressible high load acetaminophen formulations
US6852336B2 (en) * 1995-11-15 2005-02-08 J. Rettenmaier & Soehne Gmbh + Co. Kg Directly compressible high load acetaminophen formulations
US5965166A (en) * 1995-11-15 1999-10-12 Edward Mendell Co., Inc. Directly compressible high load acetaminophen formulations
US20050147673A1 (en) * 1996-06-10 2005-07-07 J. Rettenmaier & Soehne Gmbh + Co. Kg Directly compressible sustained release formulation containing microcrystalline cellulose
US6866867B2 (en) * 1996-06-10 2005-03-15 J. Rettenmaier & Soehne Gmbh + Co. Kg Process for preparing a directly compressible solid dosage form containing microcrystalline cellulose
US6395303B1 (en) * 1996-06-10 2002-05-28 Edward Mendell Co., Inc. Process for preparing a directly compressible solid dosage form containing microcrystalline cellulose
US20020037828A1 (en) * 1997-10-28 2002-03-28 Wilson Leland F. Administration of phosphodiesterase inhibitors for the treatment of premature ejaculation
US6584472B2 (en) * 1999-11-24 2003-06-24 Classen Immunotherapies, Inc. Method, system and article for creating and managing proprietary product data
US6219674B1 (en) * 1999-11-24 2001-04-17 Classen Immunotherapies, Inc. System for creating and managing proprietary product data
US6211229B1 (en) * 2000-02-17 2001-04-03 Neil B. Kavey Treatment of transient and short term insomnia
US6344487B1 (en) * 2000-06-05 2002-02-05 Neil B. Kavey Treatment of insomnia
US7135196B2 (en) * 2000-09-19 2006-11-14 Vitra Pharmaceuticals Limited Iron compositions
US20020197235A1 (en) * 2000-11-03 2002-12-26 Moran Stanford Mark Method for short-term and long-term drug dosimetry
US20030206978A1 (en) * 2001-11-29 2003-11-06 Bob Sherwood Agglomerated particles including an active agent coprocessed with silicified microcrystalline cellulose
US7179488B2 (en) * 2001-11-29 2007-02-20 Bob Sherwood Process for co-spray drying liquid herbal extracts with dry silicified MCC
US20050123609A1 (en) * 2001-11-30 2005-06-09 Collegium Pharmaceutical, Inc. Pharmaceutical composition for compressed annular tablet with molded triturate tablet for both intraoral and oral administration
US6683102B2 (en) * 2001-12-03 2004-01-27 Elan Pharmaceuticals, Inc. Methods of using metaxalone in the treatment of musculoskeletal conditions
US6407128B1 (en) * 2001-12-03 2002-06-18 Elan Pharmaceuticals, Inc. Method for increasing the bioavailability of metaxalone
US20030235617A1 (en) * 2002-02-07 2003-12-25 Martino Alice C. Pharmaceutical dosage form for mucosal delivery
US20040063721A1 (en) * 2002-08-15 2004-04-01 Wyeth Agonism of the 5HT2A receptor for treatment of thermoregulatory dysfunction
US20040115142A1 (en) * 2002-09-05 2004-06-17 Jrs Pharma Lp Compositions for industrial applications
US7276536B2 (en) * 2003-03-17 2007-10-02 Japan Tobacco Inc. Method for increasing the bioavailability of the active form of S-[2-([[1-(2-ethylbutyl)cyclohexyl]carbonyl]amino) phenyl] 2-methylpropanethioate
US20050118261A1 (en) * 2003-06-12 2005-06-02 Oien Hal J. Compositions and methods of administering doxepin to mucosal tissue
US20050196439A1 (en) * 2003-10-24 2005-09-08 J. Rettenmaier & Soehne Gmbh + Co. Kg Process for co-spray drying agents with dry silicified MCC
US20050256165A1 (en) * 2003-12-10 2005-11-17 Edgar Dale M Doxepin analogs and methods of use thereof
US20050171160A1 (en) * 2003-12-10 2005-08-04 Edgar Dale M. Doxepin analogs and methods of use thereof
US20050214365A1 (en) * 2004-03-24 2005-09-29 Gulf Pharmaceutical Industries [Instant dissolving tablet composition for loratidine and desloratidine]
US20050239838A1 (en) * 2004-04-23 2005-10-27 Dale Edgar Methods of treating sleep disorders
US20060228487A1 (en) * 2005-04-11 2006-10-12 J. Rettenmaier & Söehne GmbH + Co. KG Methods of combining active agents with augmented microcrystalline cellulose
US7452872B2 (en) * 2005-08-24 2008-11-18 Salix Pharmaceuticals, Inc. Formulations and uses of 2-hydroxy-5-phenylazobenzoic acid derivatives
US20070281990A1 (en) * 2006-05-19 2007-12-06 Rogowski Roberta L Methods of using low-dose doxepin for the improvement of sleep
US20090042972A1 (en) * 2006-05-19 2009-02-12 Somaxon Pharmaceuticals, Inc. Methods of using low-dose doxepin for the improvement of sleep
US20090042971A1 (en) * 2006-05-19 2009-02-12 Somaxon Pharmaceuticals, Inc. Method of using low-dose doxepin for the improvement of sleep
US20100179214A1 (en) * 2006-05-19 2010-07-15 Somaxon Pharmaceuticals, Inc. Doxepin trans isomers and isomeric mixtures and methods of using the same to treat sleep disorders
US20100179215A1 (en) * 2006-05-19 2010-07-15 Somaxon Pharmaceuticals, Inc. Doxepin isomers and isomeric mixtures and methods of using the same to treat sleep disorders
US20100227916A1 (en) * 2006-05-19 2010-09-09 Somaxon Pharmaceuticals, Inc N-desmethyl-doxepin and methods of using the same to treat sleep disorders
US20110318412A1 (en) * 2006-05-19 2011-12-29 Somaxon Pharmaceuticals, Inc. Low dose doxepin formulations, including buccal, sublingual and fastmelt formulations, and uses of the same to treat insomnia
US20080058407A1 (en) * 2006-07-20 2008-03-06 Cara Baron Methods of improving the pharmacokinetics of doxepin
US20080054408A1 (en) * 2006-08-31 2008-03-06 Kimberly-Clark Worldwide, Inc. Conduction through a flexible substrate in an article
US20080182890A1 (en) * 2006-10-04 2008-07-31 Somaxon Pharmaceuticals, Inc. Methods of using low-dose doxepin for the improvement of sleep
US20100105614A1 (en) * 2006-10-25 2010-04-29 Somaxon Pharmaceuticals, Inc. Ultra low dose doxepin and methods of using the same to treat sleep disorders
US20110077200A1 (en) * 2006-12-06 2011-03-31 Somaxon Pharmaceuticals, Inc. Combination therapy using low-dose doxepin for the improvement of sleep
US20090074862A1 (en) * 2007-04-13 2009-03-19 Luigi Schioppi Low-dose doxepin formulations and methods of making and using the same

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
Adapin, 2005, 3 pages *
Insomnia, 2000, 4 pages *
Prescription Drugs, 1999, 3 pages *

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9498462B2 (en) 2006-05-19 2016-11-22 Pernix Sleep, Inc. Doxepin trans isomers and isomeric mixtures and methods of using the same to treat sleep disorders
US9107898B2 (en) 2006-05-19 2015-08-18 Pernix Sleep, Inc. Methods of using low-dose doxepin for the improvement of sleep
US20070281990A1 (en) * 2006-05-19 2007-12-06 Rogowski Roberta L Methods of using low-dose doxepin for the improvement of sleep
US10238620B2 (en) 2006-05-19 2019-03-26 Pernix Sleep, Inc. Methods of using low-dose doxepin for the improvement of sleep
US20100179215A1 (en) * 2006-05-19 2010-07-15 Somaxon Pharmaceuticals, Inc. Doxepin isomers and isomeric mixtures and methods of using the same to treat sleep disorders
US20100179214A1 (en) * 2006-05-19 2010-07-15 Somaxon Pharmaceuticals, Inc. Doxepin trans isomers and isomeric mixtures and methods of using the same to treat sleep disorders
US20100227916A1 (en) * 2006-05-19 2010-09-09 Somaxon Pharmaceuticals, Inc N-desmethyl-doxepin and methods of using the same to treat sleep disorders
US10143676B2 (en) 2006-05-19 2018-12-04 Pernix Sleep, Inc. Doxepin trans isomers and isomeric mixtures and methods of using the same to treat sleep disorders
US8513299B2 (en) 2006-05-19 2013-08-20 Pernix Sleep, Inc. Methods of using low-dose doxepin for the improvement of sleep
US10251859B2 (en) 2006-05-19 2019-04-09 Pernix Sleep, Inc. Doxepin isomers and isomeric mixtures and methods of using the same to treat sleep disorders
US9463181B2 (en) 2006-05-19 2016-10-11 Pemix Sleep, Inc. Doxepin isomers and isomeric mixtures and methods of using the same to treat sleep disorders
US9486437B2 (en) 2006-05-19 2016-11-08 Pernix Sleep, Inc. Methods of using low-dose doxepin for the improvement of sleep
US11234954B2 (en) 2006-05-19 2022-02-01 Currax Pharmaceuticals Llc Low-dose doxepin for treatment of sleep disorders in elderly patients
US9861607B2 (en) 2006-05-19 2018-01-09 Procom One, Inc. Methods of using low-dose doxepin for the improvement of sleep
US10653660B2 (en) 2006-07-20 2020-05-19 Currax Pharmaceuticals Llc Methods of improving the pharmacokinetics of doxepin
US9572814B2 (en) 2006-07-20 2017-02-21 Pernix Sleep, Inc. Methods of improving the pharmacokinetics of doxepin
US20080182890A1 (en) * 2006-10-04 2008-07-31 Somaxon Pharmaceuticals, Inc. Methods of using low-dose doxepin for the improvement of sleep
US20100105614A1 (en) * 2006-10-25 2010-04-29 Somaxon Pharmaceuticals, Inc. Ultra low dose doxepin and methods of using the same to treat sleep disorders
US9907779B2 (en) 2006-10-25 2018-03-06 Pernix Sleep, Inc. Ultra low dose doxepin and methods of using the same to treat sleep disorders
US10507193B2 (en) 2006-10-25 2019-12-17 Currax Pharmaceuticals Llc Ultra low dose doxepin and methods of using the same to treat sleep disorders
US20110077200A1 (en) * 2006-12-06 2011-03-31 Somaxon Pharmaceuticals, Inc. Combination therapy using low-dose doxepin for the improvement of sleep
US11013712B2 (en) 2006-12-06 2021-05-25 Currax Pharmaceuticals Llc Methods of treating insomnia using a combination therapy of low-dose doxepin and zolpidem
US9907780B2 (en) 2007-04-13 2018-03-06 Pernix Sleep, Inc. Low-dose doxepin formulations and methods of making and using the same
US20090074862A1 (en) * 2007-04-13 2009-03-19 Luigi Schioppi Low-dose doxepin formulations and methods of making and using the same
US11096920B2 (en) 2007-04-13 2021-08-24 Currax Pharmaceuticals Llc Low-dose doxepin formulations and methods of making and using the same
US9532971B2 (en) 2007-04-13 2017-01-03 Pernix Sleep, Inc. Low-dose doxepin formulations and methods of making and using the same

Also Published As

Publication number Publication date
CA2693992A1 (en) 2008-01-24
US20170157140A1 (en) 2017-06-08
US9572814B2 (en) 2017-02-21
US10653660B2 (en) 2020-05-19
WO2008011150A1 (en) 2008-01-24
US20200276151A1 (en) 2020-09-03
CA2693992C (en) 2017-01-31
US20080058407A1 (en) 2008-03-06
US20210393567A1 (en) 2021-12-23
US11110074B2 (en) 2021-09-07
US7915307B2 (en) 2011-03-29
US20130041021A1 (en) 2013-02-14

Similar Documents

Publication Publication Date Title
US11110074B2 (en) Methods of improving the pharmacokinetics of doxepin
US9655859B2 (en) Accordion pill comprising levodopa for an improved treatment of Parkinson's disease symptoms
EP2646031B9 (en) Increasing drug bioavailability in naltrexone therapy
US20190388370A1 (en) Treatment of symptoms associated with female gastroparesis
US20040034102A1 (en) Oral pediatric trimethobenzamide formulations and methods
US20220088007A1 (en) Increasing Drug Bioavailability In Naltrexone Therapy
CN116322682A (en) Lometapie for use in a method of treating hyperlipidemia and hypercholesterolemia in pediatric patients
RU2788450C2 (en) Methods for weight loss therapy in patients with dominant depression

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION