US20130053448A1 - Therapeutic Regimens - Google Patents

Therapeutic Regimens Download PDF

Info

Publication number
US20130053448A1
US20130053448A1 US13/697,230 US201113697230A US2013053448A1 US 20130053448 A1 US20130053448 A1 US 20130053448A1 US 201113697230 A US201113697230 A US 201113697230A US 2013053448 A1 US2013053448 A1 US 2013053448A1
Authority
US
United States
Prior art keywords
dosage form
unit dosage
ethyl
compound
salt
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
US13/697,230
Inventor
Louis O'Dea
C. Richard Lyttle
Jonathan Guerriero
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.)
Radius Health Inc
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US13/697,230 priority Critical patent/US20130053448A1/en
Publication of US20130053448A1 publication Critical patent/US20130053448A1/en
Assigned to RADIUS HEALTH, INC. reassignment RADIUS HEALTH, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GUERRIERO, JONATHAN, LYTTLE, C. RICHARD, O'DEA, LOUIS
Assigned to RADIUS HEALTH, INC., RADIUS PHARMACEUTICALS, INC. reassignment RADIUS HEALTH, INC. RELEASE BY SECURED PARTY (SEE DOCUMENT FOR DETAILS). Assignors: MIDCAP FINANCIAL TRUST, MIDCAP FUNDING IV TRUST
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
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/485Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/12Drugs for genital or sexual disorders; Contraceptives for climacteric disorders
    • 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
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones

Definitions

  • This invention relates to clinically useful therapeutic dosing regimens for the compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) or its salts.
  • This invention also includes unit dosage forms containing the compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) or its salts.
  • this invention includes a unit dosage form suitable for administration to a human comprising 10 mg of the compound of formula I as a salt
  • the 10 mg of the compound of Formula 1 is present as its dihydrochloride salt.
  • the 10 mg of the compound of Formula 1 is present as an acid addition salt.
  • the 10 mg of the compound of Formula 1 is present as its hydrogen chloride salt.
  • this invention includes a unit dosage form suitable for administration to a human comprising 5 mg of the compound of formula I as a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • a salt e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt.
  • this invention includes a unit dosage form suitable for administration to a human comprising 2.5 mg of the compound of formula I as a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • a salt e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt.
  • this invention includes a unit dosage form suitable for administration to a human comprising 1 mg of the compound of formula I as a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • a salt e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt.
  • the unit dosage form comprising the compound of formula I contains 1 mg, 2.5 mg, 5 mg or 10 mg as a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • a salt e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt.
  • the unit dosage form comprising the compound of formula I contains 1 mg, 2.5 mg, 5 mg or 10 mg as a hydrochloride salt.
  • the unit dosage form comprising the compound of formula I contains 1 mg, 2.5 mg, 5 mg or 10 mg as a dihydrochloride salt.
  • the unit dosage form comprising the compound of formula I contains 1 mg, 2.5 mg, 5 mg or 10 mg as a salt, wherein said salt is a hydrogen chloride salt wherein said hydrogen chloride is present in a stoichiometry of at least one hydrogen chloride molecule but no more than 2.1 hydrogen chloride molecules for each molecule of formula I.
  • the unit dosage form is for once daily administration. In another aspect, the unit dosage form is for oral administration. In yet another aspect the unit dosage form is suitable for once daily, oral administration.
  • the unit dosage form further comprises one or more pharmaceutically acceptable excipients.
  • the dosage form unit is a tablet or capsule suitable for oral administration.
  • the dosage unit form after administration, is essentially dissolved in the stomach prior to being released into the small intestines.
  • the dosage form unit is not coated with an acid resistant coating.
  • the intended human is a woman.
  • This invention also includes methods of treating vasomotor disturbances in a peri- or post-menopausal woman in need thereof comprising administering to the woman a unit dosage form comprising 10 mg of the compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) as a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • a salt e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt.
  • This invention also includes methods of treating vasomotor disturbances in a peri- or post-menopausal woman in need thereof comprising administering to the woman a unit dosage form comprising 10 mg of the compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) as its dihydrochloride salt.
  • This invention also includes methods of treating vasomotor disturbances in a post-menopausal woman in need thereof comprising administering to the woman a unit dosage form containing 10 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • a salt e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt.
  • This invention also includes methods of treating vasomotor disturbances in a post-menopausal woman in need thereof comprising administering to the woman a unit dosage form containing 10 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as its dihydrochloride salt.
  • the methods of this invention comprise the once-daily, oral administration of 10 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • salt e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt.
  • the methods of this invention comprise the once-daily, oral administration of 10 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as its dihydrochloride salt.
  • This invention also includes methods of treating vasomotor disturbances in a peri- or post-menopausal woman in need thereof comprising administering to the woman a unit dosage form comprising 1 mg, 2.5 mg or 5 mg of the compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) as a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • a salt e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt.
  • This invention also includes methods of treating vasomotor disturbances in a peri- or post-menopausal woman in need thereof comprising administering to the woman a unit dosage form comprising 1 mg, 2.5 mg or 5 mg of the compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) as its dihydrochloride salt.
  • This invention also includes methods of treating vasomotor disturbances in a post-menopausal woman in need thereof comprising administering to the woman a unit dosage form containing 1 mg, 2.5 mg or 5 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8 -tetrahydronaphthalen-2-ol as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • a unit dosage form containing 1 mg, 2.5 mg or 5 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8 -tetrahydronaphthalen-2-ol as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • This invention also includes methods of treating vasomotor disturbances in a post-menopausal woman in need thereof comprising administering to the woman a unit dosage form containing 1 mg, 2.5 mg or 5 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as its dihydrochloride salt.
  • the methods of this invention comprise the once-daily, oral administration of 1 mg, 2.5 mg or 5 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • salt e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt.
  • the methods of this invention comprise the once-daily, oral administration of 1 mg, 2.5 mg or 5 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8 -tetrahydronaphthalen-2-ol as its dihydrochloride salt.
  • the dosage form unit is a tablet or capsule suitable for oral administration.
  • the methods of this invention administer a unit dosage form that, after administration, is essentially dissolved in the stomach prior to being released into the small intestines.
  • the methods of this invention administer a unit dosage form that is not coated with an acid resistant coating.
  • This invention also includes methods of treating vasomotor disturbances in a peri- or post-menopausal woman comprising administering to the woman a compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) or a salt thereof wherein said administration results in a steady state plasma C max of from 0.40 ng/mL to 1.44, such as from 0.40 ng/mL to 1.43 ng/mL.
  • said administration results in a steady state C max of from 0.22 ng/mL to 1.61 ng/mL.
  • the compound of formula I is in the form of a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • the compound of formula is orally administered, once-daily.
  • the compound that is administered orally, once-daily is in a unit dosage form.
  • the unit dosage form is a tablet or a capsule.
  • This invention also includes methods of treating vasomotor disturbances in a post-menopausal woman comprising administering to the woman a compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) wherein said administration results in a steady state plasma C max of from 0.40 ng/mL to 1.44 ng/mL, such as from 0.40 ng/mL to1.43 ng/mL. In certain related embodiments, said administration results in a steady state C max of from 0.22 ng/mL to 1.61 ng/mL.
  • the compound is administered once-daily. In another aspect, the compound is administered orally, once-daily.
  • (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol is administered as its dihydrochloride salt.
  • This invention also includes methods of treating vasomotor disturbances in a peri- or post-menopausal woman comprising the administering of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol wherein said daily administration results in a steady state trough level of from 0.11 ng/mL to 0.79 ng/mL or from 0.12 ng/mL to 0.86 ng/mL.
  • (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol is administered as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • its salt e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt.
  • this invention also includes methods of treating vasomotor disturbances in post-menopausal women comprising the daily administration of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol wherein said administration results in a steady state plasma C max of from 0.40 ng/mL to 1.43 ng/mL or a steady state plasma C max of from 0.22 ng/mL to 1.61 ng/mL and a steady state trough plasma level of from 0.11 ng/mL to 0.79 ng/mL or from 0.12 ng/mL to 0.86 ng/mL In some embodiments, (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8
  • the one or more delineated pharmacokinetic parameters are achieved by the administration of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as a once-daily, oral administration of a unit dosage form.
  • the unit dosage form is a capsule or tablet.
  • the capsule or tablet is essentially dissolved in the stomach prior to being released into the small intestines.
  • the dosage form unit is not coated with an acid resistant coating.
  • the one or more delineated pharmacokinetic parameters are achieved by the administration of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as a once-daily, oral administration of a dosage form unit comprising 10 mg of the compound as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • a dosage form unit comprising 10 mg of the compound as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • the dosage form unit is a capsule or tablet. In certain embodiments, the capsule or tablet is essentially dissolved in the stomach prior to being released into the small intestines. In some embodiments, the dosage form unit is not coated with an acid resistant coating.
  • the one or more delineated pharmacokinetic parameters are achieved by the administration of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as a once-daily, oral administration of a dosage form unit comprising 1 mg, 2.5 mg or 5mg of the compound as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • a dosage form unit comprising 1 mg, 2.5 mg or 5mg of the compound as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • the dosage form unit is a capsule or tablet. In certain embodiments, the capsule or tablet is essentially dissolved in the stomach prior to being released into the small intestines. In some embodiments, the dosage form unit is not coated with an acid resistant coating.
  • FIG. 1 is a graph showing the reduction in mean weekly frequency of moderate and severe hot flashes (Safety/Intent-to-treat population (ITT)).
  • FIG. 2 is a graph showing the reduction in mean weekly frequency of all hot flashes (ITT).
  • FIG. 3 is a graph showing reduction in mean severity of all hot flashes (ITT).
  • FIG. 4 is a graph showing the reduction in weekly composite scores.
  • CNS central nervous system
  • hormone levels ie estrogens and progestins
  • the side effects are variously described but most prominently include an uncomfortable heating of the skin, usually on the upper body and most often in the face and neck.
  • the sensations are transitory in nature, typically less than 30 minutes in duration.
  • the sensation of heat can be accompanied by sweating, accelerated heart rate and a significant amount of physical discomfort.
  • These sensations are typically referred to as hot flashes or hot flushes and may occur not only in the day but at night as well.
  • women might experience additional CNS disturbances. For example, some women experience depression, irritability, mood swings and other emotional or mood disorders as a consequence of the declining and/or fluctuating hormone levels associated with the menopause period. It has been known for some time that if women supplement their declining and/or fluctuating hormone levels with estrogens and/or progestins, these side effects can be largely, if not completely, eliminated. However, depending on the particular treatment modality being employed, the use of these hormones in postmenopausal women can be associated with certain side effects including the possibility of increased risk for breast cancer, uterine bleeding, uterine cancer, stroke, blood clots and even heart disease. Not surprisingly, researchers have for some time been trying to find therapies that will treat the hormone-associated CNS effects of the menopausal period while avoiding some, and preferably all of the side effects.
  • a dihydrochloride salt means that the base compound (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol has from 1.6 to 2 HCl's associated with the molecule.
  • a dihydrochloride salt refers to the base compound (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol wherein that base compound has from 1.8 to 2 HCl's associated with it.
  • the term dihydrochloride refers to the base compound with approximately 2HCl's associated with it.
  • a “unit dosage form” is a dosage form that is a discrete dosage formulation meaning that the entire drug content to be administered at a single time is contained in a single dosage form unit.
  • pills, tablets, lozenges, capsules are all dosage form units for purposes of this invention.
  • the drug or some component of the drug formulation is not a single dosage form but only that the dose to be administered as a whole is a discrete unit.
  • a capsule may contain granules of the drug within the capsule but it is a single capsule that is to be administered.
  • a lozenge may contain the drug substance dissolved in a matrix but the drug form is the lozenge itself and thus is a unit dosage formulation.
  • the “unit dosage form” can be administered to a patient or preferably self-administered by a patient in a form that is generally acceptable in the pharmaceutical venue.
  • the unit dosage form is pharmaceutically useful.
  • pharmaceutically useful dosage forms are suitable for oral delivery and include pills, tablets, buccal tablets, orally-disintegrating tablets, thin films, chewable tablets, lozenges, sublingual tablets or pills, liquid solutions, suspensions, syrups, powder or granules suitable for sprinkling in food or beverages capsules, food or candy with the drug contained within.
  • Excipients include those excipients that are generally regarded as safe for administration to humans.
  • Excipients especially suitable for formulations suitable for oral delivery include diluents (monosaccharides, disaccharides and polyhydric alcohols including starch, mannitol, dextrose, sucrose, microcrystalline cellulose, maltodextrin, sorbitol, xylitol, fructose and the like), binders (starch, gelatin, natural sugars, gums, waxes and the like), disintegrants (alginic acid, carboxymethylcellulose (calcium or sodium), cellulose, crocarmellose, crospovidone, microcrystalline cellulose, sodium starch glycolate, agar and the like), acidic or basic buffering agents (citrates, phoshphates, gluconates, acetates, carbonates, bicarbonates and the like), chelating agents (edetic acid, edetate calcium, edetate disodium and
  • the unit dosage formulations or unit dosage forms of this invention can be prepared in different forms including most commonly, tablets and capsules.
  • the tablets can be formulated by a wide variety of methods known to one of skill in the art including, for example, preparing a dry powder mixture of the drug substance in combination with one or more of the excipients granulating the mixture and pressing to together into a tablet and optionally coating the tablet with an enteric or non-enteric coating.
  • the final coat typically includes a light protective pigment such as titanium oxide and a shellac or wax to keep the tablet dry and stable. While not intending to be limited by theory or example, in some instances it might be preferred to prepare the tablets by wet granulating the drug with one or more of the excipients and then extruding the granulated material.
  • the unit dosage formulations or unit dosage forms of this invention also include capsules wherein the drug is enclosed inside the capsule either as a powder together with optional excipients or as granules containing usually including one or more excipients together with the drug and wherein the granule in turn can be optionally coated, for example, enterically or non-enterically.
  • the formulations of this invention may be solids and when present as solids, they maybe of defined particle size. It maybe sometimes preferable to administer the compound with a certain particle size—a particle size with a preferred range where the average mean particle size diameter is under 100 microns, or 75 microns, or 50 microns, or 35 microns, or 10 microns or 5 microns.
  • a dose description of 10 mg when referring to (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride means about 10 mg of the entire weight of the compound as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt) such as its dihydrochloride salt. Small variations about the 10 mg should be considered to fall within the 10 mg description of this invention. For example, differences in weighing, humidity, small impurities of the synthesis, etc means that a dose range from 9 mg to 11 mg will be considered equal to 10 mg for purpose of the description in this invention.
  • a dose description of 1 mg, 2.5 mg or 5 mg when referring to (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride means about 1 mg, 2.5 mg or 5 mg of the entire weight of the compound as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt), such as its dihydrochloride salt. Small variations about the 1 mg, 2.5 mg or 5 mg should be considered to fall within the weight description of this invention.
  • differences in weighing, humidity, small impurities of the synthesis, etc means that a dose range from 0.9 mg to 1.1 mg will be considered equal to 1 mg for purpose of the description in this invention or 2.2 mg to 2.8 mg will be considered equal to 2.5 mg for purpose of the description in this invention, or 4.4 mg to 5.6 mg will be considered equal to 5 mg for purpose of the description in this invention.
  • the compound (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride may vary to a certain degree with regard to the amount of the (S)-enantiomer that may be present in the drug product.
  • the phrase “essentially dissolved in the stomach” means that after oral administration of a dosage form of this invention or a method of this invention, the compound (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol is more than 50% dissolved in the stomach prior to being released into the duodenum.
  • the phrase “essentially dissolved in the stomach” means that after oral administration of a dosage form of this invention or a method of this invention, the compound (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol is more than 70% dissolved in the stomach prior to being released into the duodenum.
  • the phrase “essentially dissolved in the stomach” means that after oral administration of a dosage form of this invention or a method of this invention, the compound (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol is more than 90% dissolved in the stomach prior to being released into the duodenum.
  • the dissolution of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol in the stomach can be affected by different parameters, particularly those related to the excipients that the compound is co-formulated with and especially to whether the dosage formulation form is coated with a material that prevents dissolution of the coating material in the acid environment of the stomach.
  • the dosage form is not coated with an acid resistant coating.
  • an acid resistant coating refers to a coating on a tablet, granule, tablet, capsule or any other particle that contains (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol within it.
  • An acid resistant material means that the coating is resistant to pH's of up to about 5.5.
  • Acid resistant coating materials are well known by those of ordinary skill in the art and include materials such as polymeric acids such as methacrylic acid polymers, and the like. Acid resistant materials also include such materials as waxes, shellacs, fatty acids, polymeric acids, plant fibers and the like.
  • the drug in a form that avoids or largely avoids exposure of the drug substance in the person's stomach being treated.
  • a patient suffering from gastritis or some other form of irritation in the stomach may prefer to take a dosage formulation that largely or completely bypasses the direct exposure of the drug to the stomach lining.
  • a coating may be added to the drug dosage form such as an enteric coating that is stable in the acidic pH in the stomach but is more readily dissolved in the less acidic environment of the small intestine.
  • Acid resistant materials also include such materials as waxes, shellacs, fatty acids, polymeric acids, plant fibers and the like.
  • vasomotor disturbances refers to the constellation of central nervous system disturbances associated with the peri- and postmenopausal period.
  • vasomotor disturbances includes hot flashes and/or hot flushes that are severe, moderate or mild.
  • Vasomotor disturbances may also include inappropriate sweating and/or sweating at night.
  • Vasomotor disturbances is sometimes referred to in the art as “vasomotor symptoms” associated with the peri- or postmenopause periods.
  • a “perimenopausal woman” is a woman who is transitioning into menopause.
  • the perimenopausal period is not strictly defined but rather is understood by one of skill in the medical arts to include women whose estrogen levels have begun to fluctuate unevenly often leading to irregular menstrual cycles.
  • the perimenopausal period typically begins several years before true menopause and includes up to one year after the woman's final menstrual period. From a functional perspective with regard to treating a perimenopausal woman with the compositions and methods of this invention, the perimenopausal period is most easily identified by the symptoms associated with it.
  • postmenopausal woman refers to any woman who has is at least 40 years of age and is naturally amenorrheic for a period of at least one year or has elevated FSH levels (>25 mIU in some embodiments, >30 mIU in some embodiments and >50 mIU in other embodiments).
  • postmenopausal woman refers to women who are amenorrheic through surgical removal of the ovaries.
  • the term “postmenopausal woman” refers to women who are amenorrheic through treatment with an agent that causes suppression of ovarian function such as leutinizing hormone agonists (eg leuprolide), antagonists or gonadotropin hormone releasing hormone antagonists such (eg ganirelix).
  • an agent that causes suppression of ovarian function such as leutinizing hormone agonists (eg leuprolide), antagonists or gonadotropin hormone releasing hormone antagonists such (eg ganirelix).
  • Postmenopausal women from 40 to 75 years of age (inclusive) who meet the study entry criteria and have provided written informed consent will be eligible for the study.
  • the women will be established to be postmenopausal on the basis of menstrual history (one year of amenorrhea) and/or serum FSH levels. Women with documented surgical menopause who meet the study entry criteria will also be eligible 6 months after surgery.
  • the principal entry criterion is a documented history of moderate to severe hot flashes with an event frequency of 7 per day or 50 per week. All patients are to be in good general health as determined by medical history, physical examination (including pelvic examination and Pap test) and clinical laboratory testing. Pelvic ultrasound should also demonstrate no clinically significant abnormality.
  • BMI Body Mass Index
  • a total of 100 patients are planned to be enrolled in the study.
  • Study Diary entries of hot flash frequency and severity and associated symptoms will be reviewed at each study visit. Serum markers of estrogen effect will be measured at specific study visits while on treatment.
  • Safety evaluations include physical examinations, vital signs, 12-lead ECG, clinical laboratory tests, and adverse events. Study specific algorithms for management of vaginal bleeding and clinically significant breast tenderness have been included.
  • Safety laboratory assessments will be performed at specific study visits during the treatment period and the final visit. All patients will be monitored for cardiac safety (QTc interval assessment) at specific study visits using a standardized 12-lead ECG.
  • the safety data includes the incidence and severity of adverse events by dose and cumulative dose, and the pathological changes in hematology, chemistry and urinalysis data. Changes in physical examination, vital signs, ECG and clinical laboratory tests will be summarized using descriptive statistics. Shift frequencies will be summarized for clinical laboratory tests.
  • Treatment groups will be assessed for uniformity at baseline (baseline characteristics, medical history, physical examination, vital signs, and ECG).
  • Placebo contained the same excipients as active medication, but without (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride. The patients were instructed to take the capsules orally in the fasted state (morning).
  • the capsules containing (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride were prepared in dosage strengths of 0 mg, 10 mg, 25 mg and 50 mg.
  • the composition of the capsules is shown in Table 3.
  • Capsules containing 1, 2.5 and 5 mg of drug can be similarly prepared.
  • the maximum duration of study participation for an individual patient is approximately 10-weeks (72 days) from the initial Screening visit to the completion of final study evaluations. Patients will complete screening procedures during the Screening Period within 14 days prior to the first dose of study medication. After completion of the Screening Period, patients will be randomized if they meet the entrance criteria and will receive the first dose of study medication on Day 1 of the Treatment Period.
  • the Treatment Period will be 28 days in duration and will involve daily oral self-administration of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride or its Placebo and documentation of hot flashes/symptoms using a Study Diary.
  • patients will return to the clinic for the End-of-Treatment Visit (Day 29) and enter the Follow-up Period of 30 days.
  • the end of patient participation will be at the End-of-Study Visit, scheduled at the end of the Follow-up Period.
  • the treatment period duration was 4 weeks.
  • the maximum duration of study participation for an individual patient was approximately 10 weeks (72 days) from the initial Screening visit to the completion of final study evaluations.
  • Safety population (Safety/Intent-to-treat [ITT]), defined as all patients who received one or more doses of study drug
  • Per-Protocol Population defined as all patients of the ITT Population who met all entrance criteria or had a waiver of unmet criterion, had not used any prohibited concomitant medications, met study drug compliance of 90%, provided acceptably complete (25 days of full data) diary information, and met assessment and visit compliance at baseline and End-of-Treatment Visits. All efficacy endpoints were to be analyzed for the ITT and PP populations, with the exception of the drug levels (analyzed for the ITT population only). Safety endpoints were to be analyzed for the Safety/ITT population.
  • Continuous, quantitative variable summaries included the number of patients (N) (with non missing values), mean, standard deviation, median, minimum, maximum, and 95% confidence interval.
  • Categorical, qualitative variable summaries included the frequency and percentage of patients who were in the particular category. In general, the denominator for the percentage calculation was to be based upon the total number of patients in the study population for the treatment groups at the specific timepoint, unless otherwise specified. Descriptive summaries were to be presented for the number and percentage of patients in each study population (overall and by center), disposition of patients (including number of patients screened, screen failure patients, and patients randomized, completed and withdrawn), and study withdrawals by reason of withdrawal.
  • Summaries of demographics and baseline characteristics were to include the following: age, race, height, weight, BMI, medical history (time since last menses and number of months of hot flash history), hot flash history (moderate, severe, and moderate and severe), and concomitant medication at baseline, Baseline physical examination (abnormal and normal), vital signs (systolic and diastolic blood pressure, pulse rate, temperature, respiratory rate), and 12-lead ECG (abnormal and normal).
  • Summaries of AEs clinical laboratory investigations (chemistries, hematology, coagulation, serum estradiol, and urinalysis), vital signs (blood pressure, pulse rate, respiratory rate, and body temperature), physical examinations, ECG investigations, and pelvic ultrasound were to be provided. Adverse events of vaginal bleeding and breast tenderness (and related breast events) were to be tabulated separately.
  • Pair-wise comparisons of change from baseline were to be tested between the placebo group and each drug dose group for each timepoint; the mean difference together with a 95% confidence interval of the difference was to be presented for placebo compared with each active treatment group. Nominal p-values at Week 4 were to be presented. In addition, graphical presentations of mean ( ⁇ standard error of the mean) and percent group responses over time together with repeated measures ANOVA to assess potential trend in responses over time in each dose group wereto be presented. For the associated vasomotor symptoms (palpitations, insomnia, joint aches, and headaches collected in the patient diaries), patient and event frequencies were to be summarized by treatment week and treatment group.
  • the ranges were derived from the mean number listed in the table plus or minus one standard deviation (the number in parenthesis after each mean value). For example a steady state trough level ranging from 0.11 ng/mL to 0.79 ng/mL is derived from Table 4 by taking the trough level for 10 mg, 0.45 ng/mL, adding 0.34 to get the upper value of 0.79 and subtracting 0.34 to get the lover value of 0.11. Other ranges for steady state trough levels and ranges for Cmax can be similarly derived from the data in Tables 4 and 5.
  • the graphical representation of the data from table 6 is shown in FIG. 1 .
  • the 10 mg group demonstrated the greatest reduction in hot flashes at each weekly time point.
  • the graphical representation of the data from Table 7 is shown in FIG. 2 .
  • Hot flashes were also scored according to their severity with a mild hot flash scored a “1”, a moderate hot flash scored a “2” and a severe hot flash scored a “3”.
  • a mean hot flash severity can be derived by taking the sum of the total obtained by multiplying the total frequency of hot flashes in each group (mild, moderate or severe) by the severity assigned to that type of hot flash (1, 2 or 3, respectively) and then dividing the total by the frequency of all hot flashes. This gives the mean severity and is averaged over a week period and presented in Table 8. A graph of the data from Table 8 is shown in FIG. 3 . The 10 mg dose had the greatest reduction in severity of hot flashes at all time points.
  • FIG. 4 is a graphical representation of the data in Table 9.
  • the composite score indicates that the 10 mg group had the greatest composite score reduction at each weekly timepoint.

Abstract

This invention relates to clinically useful therapeutic regimens comprising the administration of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol.

Description

    RELATED APPLICATION
  • This application claims the benefit of U.S. Provisional Application No. 61/334,095, filed on May 12, 2010. The entire teachings of the above application is incorporated herein by reference.
  • BACKGROUND OF THE INVENTION
  • The compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)-benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) and it salts are described and claimed in U.S. Pat. No. 7,612,114 B2, the entire content of which is hereby incorporated by reference.
  • Figure US20130053448A1-20130228-C00001
  • Use of the compound formula I for the treatment of vasomotor symptoms is described in WO2008/002490 also incorporated herein by reference in its entirety. While the effectiveness of compound 1 and its salts in the treatment of vasomotor symptoms has been described, improved dosing regimens would benefit those suffering from vasomotor symptoms.
  • SUMMARY OF THE INVENTION
  • This invention relates to clinically useful therapeutic dosing regimens for the compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) or its salts.
  • This invention also includes unit dosage forms containing the compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) or its salts.
  • In certain embodiments, this invention includes a unit dosage form suitable for administration to a human comprising 10 mg of the compound of formula I as a salt
  • Figure US20130053448A1-20130228-C00002
  • In a particular embodiment, the 10 mg of the compound of Formula 1 is present as its dihydrochloride salt.
  • In another particular embodiment, the 10 mg of the compound of Formula 1 is present as an acid addition salt.
  • In yet another particular embodiment, the 10 mg of the compound of Formula 1 is present as its hydrogen chloride salt.
  • In certain embodiments, this invention includes a unit dosage form suitable for administration to a human comprising 5 mg of the compound of formula I as a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • In certain embodiments, this invention includes a unit dosage form suitable for administration to a human comprising 2.5 mg of the compound of formula I as a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • In certain embodiments, this invention includes a unit dosage form suitable for administration to a human comprising 1 mg of the compound of formula I as a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • In some embodiments, the unit dosage form comprising the compound of formula I contains 1 mg, 2.5 mg, 5 mg or 10 mg as a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • In some embodiments, the unit dosage form comprising the compound of formula I contains 1 mg, 2.5 mg, 5 mg or 10 mg as a hydrochloride salt.
  • In some embodiments, the unit dosage form comprising the compound of formula I contains 1 mg, 2.5 mg, 5 mg or 10 mg as a dihydrochloride salt.
  • In some embodiments, the unit dosage form comprising the compound of formula I contains 1 mg, 2.5 mg, 5 mg or 10 mg as a salt, wherein said salt is a hydrogen chloride salt wherein said hydrogen chloride is present in a stoichiometry of at least one hydrogen chloride molecule but no more than 2.1 hydrogen chloride molecules for each molecule of formula I.
  • In one aspect, the unit dosage form is for once daily administration. In another aspect, the unit dosage form is for oral administration. In yet another aspect the unit dosage form is suitable for once daily, oral administration.
  • In some instances, the unit dosage form further comprises one or more pharmaceutically acceptable excipients.
  • In certain embodiments, the dosage form unit is a tablet or capsule suitable for oral administration.
  • In certain embodiments, the dosage unit form, after administration, is essentially dissolved in the stomach prior to being released into the small intestines.
  • In some instances, the dosage form unit is not coated with an acid resistant coating.
  • In certain embodiments, the intended human is a woman.
  • This invention also includes methods of treating vasomotor disturbances in a peri- or post-menopausal woman in need thereof comprising administering to the woman a unit dosage form comprising 10 mg of the compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) as a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • This invention also includes methods of treating vasomotor disturbances in a peri- or post-menopausal woman in need thereof comprising administering to the woman a unit dosage form comprising 10 mg of the compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) as its dihydrochloride salt.
  • This invention also includes methods of treating vasomotor disturbances in a post-menopausal woman in need thereof comprising administering to the woman a unit dosage form containing 10 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • This invention also includes methods of treating vasomotor disturbances in a post-menopausal woman in need thereof comprising administering to the woman a unit dosage form containing 10 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as its dihydrochloride salt.
  • In some instances, the methods of this invention comprise the once-daily, oral administration of 10 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • In some instances, the methods of this invention comprise the once-daily, oral administration of 10 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as its dihydrochloride salt.
  • This invention also includes methods of treating vasomotor disturbances in a peri- or post-menopausal woman in need thereof comprising administering to the woman a unit dosage form comprising 1 mg, 2.5 mg or 5 mg of the compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) as a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • This invention also includes methods of treating vasomotor disturbances in a peri- or post-menopausal woman in need thereof comprising administering to the woman a unit dosage form comprising 1 mg, 2.5 mg or 5 mg of the compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) as its dihydrochloride salt.
  • This invention also includes methods of treating vasomotor disturbances in a post-menopausal woman in need thereof comprising administering to the woman a unit dosage form containing 1 mg, 2.5 mg or 5 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8 -tetrahydronaphthalen-2-ol as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • This invention also includes methods of treating vasomotor disturbances in a post-menopausal woman in need thereof comprising administering to the woman a unit dosage form containing 1 mg, 2.5 mg or 5 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as its dihydrochloride salt.
  • In some instances, the methods of this invention comprise the once-daily, oral administration of 1 mg, 2.5 mg or 5 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • In some instances, the methods of this invention comprise the once-daily, oral administration of 1 mg, 2.5 mg or 5 mg of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8 -tetrahydronaphthalen-2-ol as its dihydrochloride salt.
  • In some embodiments, the dosage form unit is a tablet or capsule suitable for oral administration.
  • In some embodiments, the methods of this invention administer a unit dosage form that, after administration, is essentially dissolved in the stomach prior to being released into the small intestines.
  • In some instances, the methods of this invention administer a unit dosage form that is not coated with an acid resistant coating.
  • This invention also includes methods of treating vasomotor disturbances in a peri- or post-menopausal woman comprising administering to the woman a compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) or a salt thereof wherein said administration results in a steady state plasma Cmax of from 0.40 ng/mL to 1.44, such as from 0.40 ng/mL to 1.43 ng/mL. In certain related embodiments, said administration results in a steady state Cmax of from 0.22 ng/mL to 1.61 ng/mL. In one aspect, the compound of formula I is in the form of a salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • In another aspect, the compound of formula is orally administered, once-daily. In yet another aspect, the compound that is administered orally, once-daily is in a unit dosage form. In a further aspect, the unit dosage form is a tablet or a capsule.
  • This invention also includes methods of treating vasomotor disturbances in a post-menopausal woman comprising administering to the woman a compound of formula I ((R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol) wherein said administration results in a steady state plasma Cmax of from 0.40 ng/mL to 1.44 ng/mL, such as from 0.40 ng/mL to1.43 ng/mL. In certain related embodiments, said administration results in a steady state Cmax of from 0.22 ng/mL to 1.61 ng/mL. In one aspect, the compound is administered once-daily. In another aspect, the compound is administered orally, once-daily.
  • In some embodiments, (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol is administered as its dihydrochloride salt.
  • This invention also includes methods of treating vasomotor disturbances in a peri- or post-menopausal woman comprising the administering of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol wherein said daily administration results in a steady state trough level of from 0.11 ng/mL to 0.79 ng/mL or from 0.12 ng/mL to 0.86 ng/mL. In some embodiments, (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol is administered as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • In some aspects, this invention also includes methods of treating vasomotor disturbances in post-menopausal women comprising the daily administration of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol wherein said administration results in a steady state plasma Cmax of from 0.40 ng/mL to 1.43 ng/mL or a steady state plasma Cmax of from 0.22 ng/mL to 1.61 ng/mL and a steady state trough plasma level of from 0.11 ng/mL to 0.79 ng/mL or from 0.12 ng/mL to 0.86 ng/mL In some embodiments, (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol is administered as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • In certain embodiments of this invention, the one or more delineated pharmacokinetic parameters are achieved by the administration of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as a once-daily, oral administration of a unit dosage form. In some instances, the unit dosage form is a capsule or tablet. In certain embodiments, the capsule or tablet is essentially dissolved in the stomach prior to being released into the small intestines. In some embodiments, the dosage form unit is not coated with an acid resistant coating.
  • In some embodiments of this invention, the one or more delineated pharmacokinetic parameters are achieved by the administration of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as a once-daily, oral administration of a dosage form unit comprising 10 mg of the compound as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • In some instances, the dosage form unit is a capsule or tablet. In certain embodiments, the capsule or tablet is essentially dissolved in the stomach prior to being released into the small intestines. In some embodiments, the dosage form unit is not coated with an acid resistant coating.
  • In some embodiments of this invention, the one or more delineated pharmacokinetic parameters are achieved by the administration of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol as a once-daily, oral administration of a dosage form unit comprising 1 mg, 2.5 mg or 5mg of the compound as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt).
  • In some instances, the dosage form unit is a capsule or tablet. In certain embodiments, the capsule or tablet is essentially dissolved in the stomach prior to being released into the small intestines. In some embodiments, the dosage form unit is not coated with an acid resistant coating.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a graph showing the reduction in mean weekly frequency of moderate and severe hot flashes (Safety/Intent-to-treat population (ITT)).
  • FIG. 2 is a graph showing the reduction in mean weekly frequency of all hot flashes (ITT).
  • FIG. 3 is a graph showing reduction in mean severity of all hot flashes (ITT).
  • FIG. 4 is a graph showing the reduction in weekly composite scores.
  • DETAILED DESCRIPTION OF THE INVENTION
  • During the period immediately leading up to the menopausal period and usually for 1 or even many more years after, many women experience various central nervous system (CNS) disturbances associated with decreasing and/or fluctuating hormone levels (ie estrogens and progestins) brought on by the shutdown of ovarian function. The side effects are variously described but most prominently include an uncomfortable heating of the skin, usually on the upper body and most often in the face and neck. The sensations are transitory in nature, typically less than 30 minutes in duration. The sensation of heat can be accompanied by sweating, accelerated heart rate and a significant amount of physical discomfort. These sensations are typically referred to as hot flashes or hot flushes and may occur not only in the day but at night as well. Beyond the hot flashes or hot flushes associated with menopause, women might experience additional CNS disturbances. For example, some women experience depression, irritability, mood swings and other emotional or mood disorders as a consequence of the declining and/or fluctuating hormone levels associated with the menopause period. It has been known for some time that if women supplement their declining and/or fluctuating hormone levels with estrogens and/or progestins, these side effects can be largely, if not completely, eliminated. However, depending on the particular treatment modality being employed, the use of these hormones in postmenopausal women can be associated with certain side effects including the possibility of increased risk for breast cancer, uterine bleeding, uterine cancer, stroke, blood clots and even heart disease. Not surprisingly, researchers have for some time been trying to find therapies that will treat the hormone-associated CNS effects of the menopausal period while avoiding some, and preferably all of the side effects.
  • The results of a phase 2a clinical study assessing the effects of the selective estrogen receptor modulator (SERM) (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol on women suffering from hot flushes are reported herein. The preclinical data for (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol indicate that it does not stimulate estrogen-dependent proliferation of mammary carcinoma cells (MCF-7) cells in the same way as protypical estrogen agonists (e.g. estradiol) and in fact, is a potent antagonist of estradiol stimulation on these cells, suggesting that this compound will not increase the risk of breast cancer in women taking the drug and possibly could even decrease that risk. Moreover, in preclinical rat models measuring estrogenic agonist effects on the endometrium, (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride demonstrates negligible estrogen agonist stimulation effects. This indicates that (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride is unlikely to cause unacceptable uterine stimulation in postmenopausal women.
  • For purposes of this invention, a dihydrochloride salt means that the base compound (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol has from 1.6 to 2 HCl's associated with the molecule. In some embodiments, a dihydrochloride salt refers to the base compound (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol wherein that base compound has from 1.8 to 2 HCl's associated with it. In certain embodiments, the term dihydrochloride refers to the base compound with approximately 2HCl's associated with it.
  • For purposes of this invention, a “unit dosage form” is a dosage form that is a discrete dosage formulation meaning that the entire drug content to be administered at a single time is contained in a single dosage form unit. For example, pills, tablets, lozenges, capsules are all dosage form units for purposes of this invention. Also contemplated in the scope of this definition is the possibility that the drug or some component of the drug formulation is not a single dosage form but only that the dose to be administered as a whole is a discrete unit. This means that a capsule may contain granules of the drug within the capsule but it is a single capsule that is to be administered. Likewise, a lozenge may contain the drug substance dissolved in a matrix but the drug form is the lozenge itself and thus is a unit dosage formulation.
  • The “unit dosage form” can be administered to a patient or preferably self-administered by a patient in a form that is generally acceptable in the pharmaceutical venue. In other word, the unit dosage form is pharmaceutically useful. By way of non-limiting example, pharmaceutically useful dosage forms are suitable for oral delivery and include pills, tablets, buccal tablets, orally-disintegrating tablets, thin films, chewable tablets, lozenges, sublingual tablets or pills, liquid solutions, suspensions, syrups, powder or granules suitable for sprinkling in food or beverages capsules, food or candy with the drug contained within.
  • Pharmaceutically acceptable excipients include those excipients that are generally regarded as safe for administration to humans. Excipients especially suitable for formulations suitable for oral delivery include diluents (monosaccharides, disaccharides and polyhydric alcohols including starch, mannitol, dextrose, sucrose, microcrystalline cellulose, maltodextrin, sorbitol, xylitol, fructose and the like), binders (starch, gelatin, natural sugars, gums, waxes and the like), disintegrants (alginic acid, carboxymethylcellulose (calcium or sodium), cellulose, crocarmellose, crospovidone, microcrystalline cellulose, sodium starch glycolate, agar and the like), acidic or basic buffering agents (citrates, phoshphates, gluconates, acetates, carbonates, bicarbonates and the like), chelating agents (edetic acid, edetate calcium, edetate disodium and the like), preservatives (benzoic acid, chlorhexidine gluconate, potassium benzoate, potassium sorbate, sorbic acid, sodium benzoate and the like), glidants and lubricants (calcium stearate, oils, magnesium stearate, magnesium trisilicate, sodium fumarate, colloidal silica, zinc stearate, sodium oleate, stearic acid, and the like), antioxidants and/or preservatives (tocopherols, ascorabtes, phenols, and the like) and acidifying agents (citric acid, fumaric acid, malic acid, tartaric acid and the like) as well as coloring agents, coating agents, flavoring agents, suspending agents, dessicants, humectants and other excipients known to those of skill in the art.
  • The unit dosage formulations or unit dosage forms of this invention can be prepared in different forms including most commonly, tablets and capsules. The tablets can be formulated by a wide variety of methods known to one of skill in the art including, for example, preparing a dry powder mixture of the drug substance in combination with one or more of the excipients granulating the mixture and pressing to together into a tablet and optionally coating the tablet with an enteric or non-enteric coating. The final coat typically includes a light protective pigment such as titanium oxide and a shellac or wax to keep the tablet dry and stable. While not intending to be limited by theory or example, in some instances it might be preferred to prepare the tablets by wet granulating the drug with one or more of the excipients and then extruding the granulated material.
  • The unit dosage formulations or unit dosage forms of this invention also include capsules wherein the drug is enclosed inside the capsule either as a powder together with optional excipients or as granules containing usually including one or more excipients together with the drug and wherein the granule in turn can be optionally coated, for example, enterically or non-enterically.
  • The formulations of this invention may be solids and when present as solids, they maybe of defined particle size. It maybe sometimes preferable to administer the compound with a certain particle size—a particle size with a preferred range where the average mean particle size diameter is under 100 microns, or 75 microns, or 50 microns, or 35 microns, or 10 microns or 5 microns.
  • For purposes of this invention, a dose description of 10 mg when referring to (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride means about 10 mg of the entire weight of the compound as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt) such as its dihydrochloride salt. Small variations about the 10 mg should be considered to fall within the 10 mg description of this invention. For example, differences in weighing, humidity, small impurities of the synthesis, etc means that a dose range from 9 mg to 11 mg will be considered equal to 10 mg for purpose of the description in this invention.
  • For purposes of this invention, a dose description of 1 mg, 2.5 mg or 5 mg when referring to (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride means about 1 mg, 2.5 mg or 5 mg of the entire weight of the compound as its salt (e.g., an acid addition salt, dihydrochloride salt or hydrogen chloride salt), such as its dihydrochloride salt. Small variations about the 1 mg, 2.5 mg or 5 mg should be considered to fall within the weight description of this invention. For example, differences in weighing, humidity, small impurities of the synthesis, etc means that a dose range from 0.9 mg to 1.1 mg will be considered equal to 1 mg for purpose of the description in this invention or 2.2 mg to 2.8 mg will be considered equal to 2.5 mg for purpose of the description in this invention, or 4.4 mg to 5.6 mg will be considered equal to 5 mg for purpose of the description in this invention. For purposes of this invention, the compound (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride may vary to a certain degree with regard to the amount of the (S)-enantiomer that may be present in the drug product. For example, when (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride is referred to, it should be understood that it generally will contain at least 95% of the R-enantiomer and up to 5% of the S-enantiomer. For purposes of calculating the desired dosaging amount, the S-enantiomer will be included in the total weight of the drug provided it does not exceed the 5% preferred limit set in this definition. However, if the drug substance is dosed having >5% of the (S)-enantiomer, then the (S)-enantiomer will be subtracted from the total weight of the drug substance for purposes of dosaging calculations.
  • For purposes of this invention, the phrase “essentially dissolved in the stomach” means that after oral administration of a dosage form of this invention or a method of this invention, the compound (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol is more than 50% dissolved in the stomach prior to being released into the duodenum. In certain embodiments of this invention, the phrase “essentially dissolved in the stomach” means that after oral administration of a dosage form of this invention or a method of this invention, the compound (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol is more than 70% dissolved in the stomach prior to being released into the duodenum. In certain embodiments of this invention, the phrase “essentially dissolved in the stomach” means that after oral administration of a dosage form of this invention or a method of this invention, the compound (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol is more than 90% dissolved in the stomach prior to being released into the duodenum. The dissolution of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol in the stomach can be affected by different parameters, particularly those related to the excipients that the compound is co-formulated with and especially to whether the dosage formulation form is coated with a material that prevents dissolution of the coating material in the acid environment of the stomach. In some embodiments of this invention, it should be noted that the dosage form is not coated with an acid resistant coating. For purposes of this invention, an acid resistant coating refers to a coating on a tablet, granule, tablet, capsule or any other particle that contains (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol within it. An acid resistant material means that the coating is resistant to pH's of up to about 5.5. Acid resistant coating materials are well known by those of ordinary skill in the art and include materials such as polymeric acids such as methacrylic acid polymers, and the like. Acid resistant materials also include such materials as waxes, shellacs, fatty acids, polymeric acids, plant fibers and the like.
  • In some embodiments of this invention, it may be preferable to provide the drug in a form that avoids or largely avoids exposure of the drug substance in the person's stomach being treated. For example, a patient suffering from gastritis or some other form of irritation in the stomach may prefer to take a dosage formulation that largely or completely bypasses the direct exposure of the drug to the stomach lining. In this regard, a coating may be added to the drug dosage form such as an enteric coating that is stable in the acidic pH in the stomach but is more readily dissolved in the less acidic environment of the small intestine. Acid resistant materials also include such materials as waxes, shellacs, fatty acids, polymeric acids, plant fibers and the like.
  • For purposes of this invention, the term “vasomotor disturbances” refers to the constellation of central nervous system disturbances associated with the peri- and postmenopausal period. In particular,vasomotor disturbances includes hot flashes and/or hot flushes that are severe, moderate or mild. Vasomotor disturbances may also include inappropriate sweating and/or sweating at night. Vasomotor disturbances is sometimes referred to in the art as “vasomotor symptoms” associated with the peri- or postmenopause periods.
  • For purposes of this invention, a “perimenopausal woman” is a woman who is transitioning into menopause. The perimenopausal period is not strictly defined but rather is understood by one of skill in the medical arts to include women whose estrogen levels have begun to fluctuate unevenly often leading to irregular menstrual cycles. The perimenopausal period typically begins several years before true menopause and includes up to one year after the woman's final menstrual period. From a functional perspective with regard to treating a perimenopausal woman with the compositions and methods of this invention, the perimenopausal period is most easily identified by the symptoms associated with it. If a woman is still having her menstrual periods or has within one year had a menstrual period but yet she is at least 30 years of age and more likely at least 40 years of age and is experiencing hot flashes/flushes, night sweats, unusual anxiety, depression and/or mood swings then she can be considered as a perimenopausal woman for who this invention can be of value.
  • For purposes of this invention description, the term “postmenopausal woman” refers to any woman who has is at least 40 years of age and is naturally amenorrheic for a period of at least one year or has elevated FSH levels (>25 mIU in some embodiments, >30 mIU in some embodiments and >50 mIU in other embodiments). In some embodiments of this invention, the term “postmenopausal woman” refers to women who are amenorrheic through surgical removal of the ovaries. In yet other aspects of this invention, the term “postmenopausal woman” refers to women who are amenorrheic through treatment with an agent that causes suppression of ovarian function such as leutinizing hormone agonists (eg leuprolide), antagonists or gonadotropin hormone releasing hormone antagonists such (eg ganirelix).
  • EXAMPLE
  • A phase 2a study was undertaken to assess the effect of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride (referred to as “drug” in tables below) to alleviate hot flashes in postmenopausal women. The study design and objectives are detailed below.
  • Study Objectives:
  • The overall objectives of the study were to assess the clinical safety and efficacy of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride in postmenopausal women with frequent, moderate to severe vasomotor symptoms. Changes in frequency and severity of symptoms were assessed. Data was evaluated to analyze the effect of increasing doses of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride on overall efficacy.
  • The specific objectives of this study are to:
      • Monitor the effects of 28-days of treatment with (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride on frequency and severity of hot flashes in postmenopausal women with frequent, moderate to severe vasomotor symptoms.
      • Monitor the safety and tolerability of 28-days of dosing with (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride in postmenopausal women with frequent, moderate to severe vasomotor symptoms.
    Study Population: Planned Inclusion Criteria:
  • Postmenopausal women from 40 to 75 years of age (inclusive) who meet the study entry criteria and have provided written informed consent will be eligible for the study. The women will be established to be postmenopausal on the basis of menstrual history (one year of amenorrhea) and/or serum FSH levels. Women with documented surgical menopause who meet the study entry criteria will also be eligible 6 months after surgery. The principal entry criterion is a documented history of moderate to severe hot flashes with an event frequency of 7 per day or 50 per week. All patients are to be in good general health as determined by medical history, physical examination (including pelvic examination and Pap test) and clinical laboratory testing. Pelvic ultrasound should also demonstrate no clinically significant abnormality. Breast health should be confirmed by mammogram in the past 6 months or at screening. Body Mass Index (BMI) of 18-30 kg/m2 (inclusive). The resting 12-lead ECG obtained during screening should have no clinically significant abnormality and a QTc (Bazett's correction) of ≦450 msec.
  • Planned Exclusion Criteria:
  • Patients reporting recent use (within 6 months of enrollment) of estrogens, SERMs, or other estrogen preparations might be excluded, as will those with recent (within 3 months of enrollment) use of any medication known to reduce or enhance vasomotor symptoms. Women with a history of thrombosis or blood clotting disorders, chronic medical or psychiatric illness, and breast, uterine or other cancer might be excluded. Patients with known substance abuse problems will also be excluded and patients who have received an investigational drug product in the past 12 months might be excluded.
  • Patients must have understood and agreed to comply with all study requirements and had signed the written Informed Consent Form (ICF).
  • Study Design and Methodology: Number of Patients:
  • A total of 100 patients are planned to be enrolled in the study.
  • Design:
  • This is a randomized, placebo-controlled, double-blind, parallel-group, Phase 2, multi-center, dose-finding study to evaluate the effects of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride in the treatment of vasomotor symptoms in postmenopausal women.
  • A total of 100 eligible patients will be randomized to receive one of 5 blinded treatment regimens. (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride at doses 0, 10, 25, 50 or 100 mg will be dosed for 28-days. The dosages of study medications and the number of patients randomized to each treatment regimen are shown in Table 1.
  • TABLE 1
    Treatment Study Daily Number
    Regimen Medication Dose (PO) of Patients
    1 drug 10 mg 20
    2 drug 25 mg 20
    3 drug 50 mg 20
    4 drag 100 mg 20
    5 Placebo 20
    Total 100
  • Study Visits:
  • The study periods and number of clinic visits are summarized in Table 2.
  • TABLE 2
    Study Duration of Scheduled
    Period Study Period Visits (#)
    Screening ≦2 weeks 2
    Treatment 4 weeks 4
    Follow-up 1 month 1
    Total ~10 Weeks 7
  • Planned Procedures and Assessments: Efficacy:
  • Patients will document in real time hot flash frequency and severity according to a predetermined scale, in the provided electronic or paper Study Diary. Study Diary entries of hot flash frequency and severity and associated symptoms will be reviewed at each study visit. Serum markers of estrogen effect will be measured at specific study visits while on treatment.
  • Safety:
  • Safety evaluations include physical examinations, vital signs, 12-lead ECG, clinical laboratory tests, and adverse events. Study specific algorithms for management of vaginal bleeding and clinically significant breast tenderness have been included.
  • Safety laboratory assessments will be performed at specific study visits during the treatment period and the final visit. All patients will be monitored for cardiac safety (QTc interval assessment) at specific study visits using a standardized 12-lead ECG.
  • Endpoints and Data Analysis:
  • The efficacy endpoints assessed are:
      • Change in frequency of hot flashes over time
      • Change in severity of hot flashes over time;
      • Change in composite score of hot flashes over time (frequency x severity)
  • The safety data includes the incidence and severity of adverse events by dose and cumulative dose, and the pathological changes in hematology, chemistry and urinalysis data. Changes in physical examination, vital signs, ECG and clinical laboratory tests will be summarized using descriptive statistics. Shift frequencies will be summarized for clinical laboratory tests.
  • Treatment groups will be assessed for uniformity at baseline (baseline characteristics, medical history, physical examination, vital signs, and ECG).
  • Treatments Administered:
  • (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride was provided by sponsor in capsule form for oral administration. All doses (0, 10, 25, 50 and 100 mg) were provided as identical presentations. Each patient was provided with 2 containers of 28-capsules each at the start of treatment and took one capsule from each container every day for 28-days. Placebo contained the same excipients as active medication, but without (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride. The patients were instructed to take the capsules orally in the fasted state (morning).
  • Capsule Formulations
  • The capsules containing (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride were prepared in dosage strengths of 0 mg, 10 mg, 25 mg and 50 mg. The composition of the capsules is shown in Table 3.
  • TABLE 3
    Strength of
    capsule** Content (drug) Prosolv ®* Total weight
     0 mg   0 mg 69.1 mg 69.1 mg
    10 mg 10.3 mg 58.8 mg 69.1 mg
    25 mg 25.8 mg 43.3 mg 69.1 mg
    50 mg 51.5 mg 17.6 mg 69.1 mg
    *Silicified microcrystalline cellulose, as a combination of 98% microcrystalline cellulose (MCC) and 2% colloidal silicon dioxide (CSD). SMCC 50 (JRS Pharma; Holzmühle 1 D-73494 Rosenberg (Germany); 2981 Route 22, Suite 1 Patterson, NY 12563-2359 (USA).
    **Capsules used were Capsugel (100 Route 206 North/Pfizer Way, Peapack, NJ 07977 USA) Coni-Snap ® gelatin capsules, size 4, white opaque.
  • A slight excess of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride was used in order to compensate for residual impurities and moisture in the drug product. Each patient self-administered two capsules to which they were blinded with regard to the capsule contents. The placebo group received 2- 0 mg capsules; the 10 mg group received 1- 10 mg capsule and 1-0 mg capsule; the 25 mg group received 1-25 mg capsule and 1-0 mg capsule; the 50 mg group received 1- 50 mg capsule and 1-0 mg capsule; and the 100 mg group received 2-50 mg capsules.
  • Capsules containing 1, 2.5 and 5 mg of drug can be similarly prepared.
  • Planned Duration of Patient Participation:
  • The maximum duration of study participation for an individual patient is approximately 10-weeks (72 days) from the initial Screening visit to the completion of final study evaluations. Patients will complete screening procedures during the Screening Period within 14 days prior to the first dose of study medication. After completion of the Screening Period, patients will be randomized if they meet the entrance criteria and will receive the first dose of study medication on Day 1 of the Treatment Period. The Treatment Period will be 28 days in duration and will involve daily oral self-administration of (R)-6-(2-(ethyl(4-(2-(ethylamino)ethyl)benzyl)amino)-4-methoxyphenyl)-5,6,7,8-tetrahydronaphthalen-2-ol dihydrochloride or its Placebo and documentation of hot flashes/symptoms using a Study Diary. After completion of 28 days of dosing, patients will return to the clinic for the End-of-Treatment Visit (Day 29) and enter the Follow-up Period of 30 days. The end of patient participation will be at the End-of-Study Visit, scheduled at the end of the Follow-up Period.
  • Study Results
  • Analyzed: A total of 136 patients were screened of which 36 (26.5%) were screen failures; therefore, 100 patients were enrolled and randomized in the study. Of the 100 patients enrolled, 100 were included in the Safety/Intent-to-Treat (ITT) population and 81 were included in the Per-Protocol Population (PP).
  • Duration of Treatment:
  • The treatment period duration was 4 weeks. The maximum duration of study participation for an individual patient was approximately 10 weeks (72 days) from the initial Screening visit to the completion of final study evaluations.
  • Efficacy:
  • Efficacy was assessed by evaluating change including baseline and percent change from baseline for the following primary endpoints:
      • Frequency of hot flashes
      • Severity of hot flashes
      • Composite score of hot flashes over time (frequency x severity)
      • drug levels
    Statistical Methods:
  • The following analysis populations were analyzed in this study: Safety population (Safety/Intent-to-treat [ITT]), defined as all patients who received one or more doses of study drug, and Per-Protocol Population (PP) defined as all patients of the ITT Population who met all entrance criteria or had a waiver of unmet criterion, had not used any prohibited concomitant medications, met study drug compliance of 90%, provided acceptably complete (25 days of full data) diary information, and met assessment and visit compliance at baseline and End-of-Treatment Visits. All efficacy endpoints were to be analyzed for the ITT and PP populations, with the exception of the drug levels (analyzed for the ITT population only). Safety endpoints were to be analyzed for the Safety/ITT population.
  • Continuous, quantitative variable summaries included the number of patients (N) (with non missing values), mean, standard deviation, median, minimum, maximum, and 95% confidence interval. Categorical, qualitative variable summaries included the frequency and percentage of patients who were in the particular category. In general, the denominator for the percentage calculation was to be based upon the total number of patients in the study population for the treatment groups at the specific timepoint, unless otherwise specified. Descriptive summaries were to be presented for the number and percentage of patients in each study population (overall and by center), disposition of patients (including number of patients screened, screen failure patients, and patients randomized, completed and withdrawn), and study withdrawals by reason of withdrawal. Summaries of demographics and baseline characteristics were to include the following: age, race, height, weight, BMI, medical history (time since last menses and number of months of hot flash history), hot flash history (moderate, severe, and moderate and severe), and concomitant medication at baseline, Baseline physical examination (abnormal and normal), vital signs (systolic and diastolic blood pressure, pulse rate, temperature, respiratory rate), and 12-lead ECG (abnormal and normal). Summaries of AEs, clinical laboratory investigations (chemistries, hematology, coagulation, serum estradiol, and urinalysis), vital signs (blood pressure, pulse rate, respiratory rate, and body temperature), physical examinations, ECG investigations, and pelvic ultrasound were to be provided. Adverse events of vaginal bleeding and breast tenderness (and related breast events) were to be tabulated separately.
  • Changes in the efficacy endpoints of frequency, severity, and composite score of hot flashes over time; associated hot flash symptoms (palpitations, insomnia, joint aches, and headaches), PD markers of estrogen effect (FSH, LH, and lipid profile), drug levels, and MRS were to be summarized using descriptive statistics. Frequency, severity, and composite score of hot flashes over time were to be analyzed using repeated measures analysis of variance (ANOVA) with factors for treatment (treatment groups), time (study day) and their interaction. A linear trend test for treatment groups (placebo, and 10, 25, 50, 100 mg doses of drug) and for study day as well as any potential interaction between the two was to be included as part of the analyses by including both dose and time as continuous variables in the ANOVA model. Pair-wise comparisons of change from baseline were to be tested between the placebo group and each drug dose group for each timepoint; the mean difference together with a 95% confidence interval of the difference was to be presented for placebo compared with each active treatment group. Nominal p-values at Week 4 were to be presented. In addition, graphical presentations of mean (±standard error of the mean) and percent group responses over time together with repeated measures ANOVA to assess potential trend in responses over time in each dose group wereto be presented. For the associated vasomotor symptoms (palpitations, insomnia, joint aches, and headaches collected in the patient diaries), patient and event frequencies were to be summarized by treatment week and treatment group.
  • Additional exploratory, post-hoc analyses were to be performed as appropriate to complement the overall understanding of study results.
  • Descriptive summaries of actual (absolute) values and changes from baseline values by study visit were to be presented for hematology, chemistry, coagulation, serum estradiol, and urinalysis by treatment for the safety population. Additionally, for each laboratory parameter, shifts in assessments from baseline to End-of Treatment Visit and End-of-Study Visit were to be presented (shift tables).
  • Results: Pharmacokinetic Evaluation
  • TABLE 4
    Mean Trough Level of Drug (ng/mL) (ITT)
    Pla-
    Visit cebo 10 mg (SD) 25 mg (SD) 50 mg (SD) 100 mg (SD)
    Day 8 0.00 0.45 (0.34) 1.33 (0.52) 3.11 (1.66) 6.91 (4.39)
    Day 15 0.00 0.49 (0.37) 1.49 (0.71) 3.39 (2.01) 6.66 (4.60)
    Day 29 0.00 0.34 (0.26) 1.18 (0.73) 3.23 (2.41) 6.03 (4.46)
    SD = standard deviation in ng/mL
  • TABLE 5
    Peak Level of drug (ng/mL) (ITT)
    Pla-
    Visit cebo 10 mg (SD) 25 mg (SD) 50 mg (SD) 100 mg (SD)
    Day 1 0.00 0.56 (0.34) 1.41 (0.63) 2.83 (1.72)  8.39 (4.04)
    Day 8 0.00 0.92 (0.52) 2.53 (0.74) 6.36 (2.58) 15.47 (7.14)
    Day 15 0.00 0.92 (0.69) 2.97 (1.09) 6.22 (3.46) 12.76 (5.28)
    SD = standard deviation in ng/mL
  • Values for ranges of steady state trough levels and steady state Cmax can be derived from Tables 4 and 5. Steady state values are the values listed for days 8 and 15.
  • The ranges were derived from the mean number listed in the table plus or minus one standard deviation (the number in parenthesis after each mean value). For example a steady state trough level ranging from 0.11 ng/mL to 0.79 ng/mL is derived from Table 4 by taking the trough level for 10 mg, 0.45 ng/mL, adding 0.34 to get the upper value of 0.79 and subtracting 0.34 to get the lover value of 0.11. Other ranges for steady state trough levels and ranges for Cmax can be similarly derived from the data in Tables 4 and 5.
  • Hot Flash Frequency Moderate and Severe Hot Flash Frequency
  • The total number of hot flashes rated as moderate and severe were reported as a weekly mean for each treatment group. As can be seen in Table 6, the 10 mg treatment group demonstrated the best treatment effect.
  • TABLE 6
    Weekly Mean Frequency of Moderate and Severe Hot Flash Numbers (ITT)
    Treatment Group
    Placebo
    10 mg 25 mg 50 mg 100 mg Total
    Timepoint Statistic N = 19 N = 22 N = 20 N = 21 N = 18 N = 100
    Baseline N 18 22 19 21 17 97
    Mean (SD) 61.3 (12.84) 63.1 (14.23) 65.9 (44.30) 71.9 (36.92) 58.5 (12.67) 64.4 (27.81)
    Week 1 N 18 22 17 20 17 94
    Mean (SD) 41.2 (24.39) 30.4 (18.92) 57.7 (50.49) 41.7 (23.79) 41.9 (24.23) 41.9 (30.37)
    Week 2 N 17 20 17 18 17 89
    Mean (SD) 36.6 (25.48) 18.5 (13.34) 42.3 (49.73) 32.8 (25.52) 35.6 (28.06) 32.7 (30.72)
    Week 3 N 17 20 17 18 16 88
    Mean (SD) 33.6 (24.80) 14.5 (11.92) 43.3 (57.15) 29.6 (25.09) 24.5 (24.06) 28.7 (32.62)
    Week 4 N 16 20 17 18 15 86
    Mean (SD) 29.5 (24.53) 13.4 (11.54) 37.4 (54.76) 29.4 (26.53) 18.6 (19.16) 25.4 (31.29)
    Abbreviations:
    (SD) = standard deviation
  • The graphical representation of the data from table 6 is shown in FIG. 1. The 10 mg group demonstrated the greatest reduction in hot flashes at each weekly time point.
  • Total Hot Flash Frequency
  • The total number of hot flashes rated as mild, moderate and severe (all hot flashes) were reported as a weekly mean for each treatment group. As can be seen in Table 7, the 10 mg treatment group demonstrated the greatest reduction in hot flashes at each weekly time point and once again demonstrates the best treatment effect.
  • TABLE 7
    Weekly Mean Frequency of Mild, Moderate and Severe Hot Flash Numbers (ITT)
    Treatment Group
    Placebo
    10 mg 25 mg 50 mg 100 mg Total
    Timepoint Statistic N = 19 N = 22 N = 20 N = 21 N = 18 N = 100
    Baseline N 18 22 19 21 17 97
    Mean (SD) 65.2 (13.29) 66.0 (13.89) 75.8 (46.46) 76.0 (36.96) 63.6 (13.23) 69.5 (28.66)
    Week 1 N 18 22 17 20 17 94
    Mean (SD) 46.7 (23.56) 36.0 (18.97) 64.4 (48.88) 54.0 (27.15) 48.6 (23.32) 49.3 (30.45)
    Week 2 N 17 20 17 18 17 89
    Mean (SD) 40.1 (23.88) 27.2 (15.16) 48.0 (48.80) 45.8 (25.16) 41.6 (27.37) 40.1 (30.03)
    Week 3 N 17 20 17 18 16 88
    Mean (SD) 38.7 (25.25) 21.2 (12.99) 46.6 (56.27) 40.5 (22.37) 29.4 (23.91) 34.9 (31.91)
    Week 4 N 16 20 17 18 15 86
    Mean (SD) 34.9 (30.84) 19.1 (12.95) 42.4 (53.18) 37.9 (23.45) 23.1 (19.45) 31.3 (31.52)
    Abbreviations:
    (SD) = standard deviation
  • The graphical representation of the data from Table 7 is shown in FIG. 2.
  • Hot Flash Severity
  • Hot flashes were also scored according to their severity with a mild hot flash scored a “1”, a moderate hot flash scored a “2” and a severe hot flash scored a “3”. A mean hot flash severity can be derived by taking the sum of the total obtained by multiplying the total frequency of hot flashes in each group (mild, moderate or severe) by the severity assigned to that type of hot flash (1, 2 or 3, respectively) and then dividing the total by the frequency of all hot flashes. This gives the mean severity and is averaged over a week period and presented in Table 8. A graph of the data from Table 8 is shown in FIG. 3. The 10 mg dose had the greatest reduction in severity of hot flashes at all time points.
  • TABLE 8
    Mean Severity Scores by Week (ITT)
    Treatment Group
    Placebo
    10 mg 25 mg 50 mg 100 mg Total
    Timepoint Statistic N = 19 N = 22 N = 20 N = 21 N = 18 N = 100
    Baseline N 18 22 19 21 17 97
    Mean (SD) 2.36 (0.430) 2.37 (0.347) 2.26 (0.348) 2.14 (0.313) 2.21 (0.330) 2.27 (0.359)
    Week 1 N 18 22 17 20 17 94
    Mean (SD) 2.12 (0.397) 1.99 (0.460) 2.12 (0.392) 1.92 (0.402) 2.04 (0.334) 2.03 (0.401)
    Week 2 N 17 20 17 17 17 88
    Mean (SD) 2.11 (0.446) 1.80 (0.514) 2.09 (0.447) 1.88 (0.535) 2.01 (0.406) 1.97 (0.478)
    Week 3 N 16 19 16 18 16 85
    Mean (SD) 2.00 (0.482) 1.84 (0.570) 2.14 (0.408) 1.86 (0.544) 1.88 (0.473) 1.94 (0.503)
    Week 4 N 14 18 16 18 15 81
    Mean (SD) 1.94 (0.504) 1.86 (0.562) 2.06 (0.404) 1.83 (0.568) 1.89 (0.484) 1.91 (0.505)
    Abbreviations:
    (SD) = standard deviation
  • Hot Flash Composite Score
  • A composite score that takes into account both the total frequency and the mean severity was calculated by multiplying together that patient's mean weekly severity with that patient's total hot flash frequency to give the composite score. The composite index is a helpful measure of the overall alleviation of hot flash burden. Table 9 lists the composite scores for the treatment groups. FIG. 4 is a graphical representation of the data in Table 9.
  • The composite score indicates that the 10 mg group had the greatest composite score reduction at each weekly timepoint.
  • TABLE 9
    Composite Scores by Week (ITT)
    Treatment Group
    Placebo
    10 mg 25 mg 50 mg 100 mg Total
    Timepoint Statistic N = 19 N = 22 N = 20 N = 21 N = 18 N = 100
    Baseline N 18 22 19 21 17 97
    Mean (SD) 155.17 156.45 165.47 162.19 140.35 156.40
    (47.09) (38.91) (119.10) (76.44) (36.82) (69.97)
    Week 1 N 18 22 17 20 17 94
    Mean (SD) 98.44 73.68 142.71 104.35 104.35 102.98
    (54.65) (41.67) (125.55) (55.52) (68.35) (74.70)
    Week 2 N 17 20 17 18 17 89
    Mean (SD) 86.24 47.75 102.82 87.78 87.18 81.25
    (57.05) (24.95) (118.57) (54.61) (74.31) (72.30)
    Week 3 N 17 20 17 18 16 88
    Mean (SD) 80.29 37.15 102.59 77.67 59.75 70.52
    (55.48) (21.96) (139.75) (51.87) (62.00) (77.19)
    Week 4 N 16 20 17 18 15 86
    Mean (SD) 69.31 34.35 89.59 75.06 43.67 61.92
    (57.98) (23.48) (134.32) (55.82) (40.32) (73.76)
    Abbreviations:
    (SD) = standard deviation
  • Taken in sum, the data presented herein demonstrate the unexpected and surprisingly consistent reduction on hot flash incidence and severity of the lowest dose tested (10 mg) compared to placebo and three higher doses. This discovery is not only unexpected and surprising but potentially beneficial as well. In general, the ability to use a lower dose suggests a lower cost of goods as well as decreased likelihood of side effects or off target effects that tend to increase as dosage of any drug is increased.
  • When the effect of the compound of formula I at 10 mg is analyzed by concentration of drug in the plasma and compared to its efficacy, it was observed that the lower concentration cohort experienced similar efficacy as the higher concentration cohort. For example, in one analysis, a cohort with a 2 hour postdose plasma concentration range of between 0.43 ng/mL to 0.53 ng/mL of the compound of formula I had a similar efficacy in hot flush frequency reduction (−75%) as the higher concentration cohort having a range of 0.66 ng/mL to 1.34 ng/mL (−77%). This indicates that lower dosages of the compound of formula I such as 5 mg, 2.5 mg or 1 mg can be effective.
  • The teachings of all patents, published applications and references cited herein are incorporated by reference in their entirety.
  • While this invention has been particularly shown and described with references to example embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims.

Claims (26)

1. A unit dosage form comprising 10 mg of the compound of formula I as its dihydrochloride salt
Figure US20130053448A1-20130228-C00003
2. The unit dosage form according to claim 1, further comprising one or more pharmaceutically acceptable excipients.
3. The unit dosage form according to claim 2, wherein the unit dosage form is a tablet or capsule suitable for oral administration.
4. The unit dosage form according to claim 3, wherein following oral administration the unit dosage form, is more than 50% dissolved in the stomach prior to being released into the small intestines.
5. The unit dosage form according to claim 4, wherein the unit dosage form is not coated with an acid resistant coating.
6. A method of treating vasomotor disturbances in a peri-menopausal or postmenopausal woman in need thereof comprising administering to the woman the unit dosage form of claim 1.
7-8. (canceled)
9. The method according to claim 6, wherein the unit dosage form is more than 50% dissolved in the stomach prior to being released into the small intestines.
10. The method according to claim 9, wherein the unit dosage form is not coated with an acid resistant coating.
11. A method of treating vasomotor disturbances in a peri-menopausal or postmenopausal woman in need thereof comprising administering to the woman a compound of formula I or a salt thereof
Figure US20130053448A1-20130228-C00004
wherein the administration results in a steady state Cmax of from 0.40 ng/mL to 1.43 ng/mL.
12. The method of claim 11, wherein a steady state trough level of the compound of formula I ranging from 0.11 ng/mL to 0.79 ng/mL or ranging from 0.12 ng/mL to 0.86 ng/mL is achieved.
13. The method of claim 11, wherein the compound of formula I is in the form of a dihydrochloride salt.
14. (canceled)
15. The method of claim 13, wherein the compound is orally administered once daily in a unit dosage form.
16. (canceled)
17. The method of claim 15, wherein the unit dosage form is a capsule or tablet and is more than 50% dissolved in the stomach prior to being released into the small intestines.
18. The method according to claim 17, wherein the unit dosage form is not coated with an acid resistant coating.
19. A method of treating vasomotor disturbances in a peri-menopausal or postmenopausal woman in need thereof comprising administering to the woman a compound of formula I or a salt thereof
Figure US20130053448A1-20130228-C00005
wherein the administration results in a steady state Cmax of from 0.22 ng/mL to 1.61 ng/mL
20. The method of claim 19, wherein the compound of formula 1 is in its dihydrochloride salt form.
21. The method of claim 19, wherein a steady state trough level of the compound of formula I ranging from 0.11 ng/mL to 0.79 ng/mL or ranging from 0.12 ng/mL to 0.86 ng/mL is achieved.
22. The method of claim 19, wherein the compound of formula I is in the form of a dihydrochloride salt.
23. (canceled)
24. The method of claim 22, wherein the compound is orally administered once daily in a unit dosage form.
25. (canceled)
26. The method of claim 24, wherein the unit dosage form is a capsule or tablet and is more than 50% dissolved in the stomach prior to being released into the small intestines.
27. The method according to claim 26, wherein the unit dosage form is not coated with an acid resistant coating.
US13/697,230 2010-05-12 2011-05-12 Therapeutic Regimens Abandoned US20130053448A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/697,230 US20130053448A1 (en) 2010-05-12 2011-05-12 Therapeutic Regimens

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US33409510P 2010-05-12 2010-05-12
US13/697,230 US20130053448A1 (en) 2010-05-12 2011-05-12 Therapeutic Regimens
PCT/US2011/036311 WO2011143469A1 (en) 2010-05-12 2011-05-12 Therapeutic regimens

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/036311 A-371-Of-International WO2011143469A1 (en) 2010-05-12 2011-05-12 Therapeutic regimens

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/281,475 Continuation US9555014B2 (en) 2010-05-12 2014-05-19 Therapeutic regimens

Publications (1)

Publication Number Publication Date
US20130053448A1 true US20130053448A1 (en) 2013-02-28

Family

ID=44914714

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/697,230 Abandoned US20130053448A1 (en) 2010-05-12 2011-05-12 Therapeutic Regimens
US14/281,475 Active US9555014B2 (en) 2010-05-12 2014-05-19 Therapeutic regimens

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/281,475 Active US9555014B2 (en) 2010-05-12 2014-05-19 Therapeutic regimens

Country Status (17)

Country Link
US (2) US20130053448A1 (en)
EP (2) EP3106159A1 (en)
CA (1) CA2799183A1 (en)
CY (1) CY1117893T1 (en)
DK (1) DK2568806T3 (en)
ES (1) ES2585884T3 (en)
HR (1) HRP20160929T1 (en)
HU (1) HUE030072T2 (en)
LT (1) LT2568806T (en)
ME (1) ME02474B (en)
MX (1) MX342898B (en)
PL (1) PL2568806T3 (en)
PT (1) PT2568806T (en)
RS (1) RS54993B1 (en)
SI (1) SI2568806T1 (en)
SM (1) SMT201600263B (en)
WO (1) WO2011143469A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100105733A1 (en) * 2006-06-23 2010-04-29 Lyttle C Richard Treatment of vasomotor symptoms with selective estrogen receptor modulators
US9555014B2 (en) 2010-05-12 2017-01-31 Radius Health, Inc. Therapeutic regimens
US10071066B2 (en) 2014-03-28 2018-09-11 Duke University Method of treating cancer using selective estrogen receptor modulators
US10385008B2 (en) 2017-01-05 2019-08-20 Radius Pharmaceuticals, Inc. Polymorphic forms of RAD1901-2HCL
US10420734B2 (en) 2014-03-28 2019-09-24 Duke University Method of treating cancer using selective estrogen receptor modulators

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012047617A1 (en) 2010-09-28 2012-04-12 Radius Health, Inc. Selective androgen receptor modulators
WO2017223115A1 (en) 2016-06-22 2017-12-28 Radius Health, Inc. Ar+ breast cancer treatment methods
SG11202013177WA (en) 2018-07-04 2021-01-28 Radius Pharmaceuticals Inc Polymorphic forms of rad 1901-2hcl

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008002490A2 (en) * 2006-06-23 2008-01-03 Radius Health, Inc. Treatment of vasomotor symptoms with selective estrogen receptor modulators
US8399520B2 (en) * 2002-12-26 2013-03-19 Eisai R&D Management Co., Ltd. Selective estrogen receptor modulator

Family Cites Families (150)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1547758A (en) 1975-07-29 1979-06-27 Shell Int Research Herbicidal composition
JPH01261381A (en) 1988-04-12 1989-10-18 Nippon Soda Co Ltd Oxa(thia)diazole derivative, its production and miticide
US5411981A (en) 1991-01-09 1995-05-02 Roussel Uclaf Phenylimidazolidines having antiandrogenic activity
FR2693461B1 (en) 1992-07-08 1994-09-02 Roussel Uclaf New substituted phenylimidazolidines, process for their preparation, their use as medicaments and the pharmaceutical compositions containing them.
US5589452A (en) 1992-07-14 1996-12-31 Syntex (U.S.A.) Inc. Analogs of parathyroid hormone and parathyroid hormone related peptide: synthesis and use for the treatment of osteoporosis
GB9310635D0 (en) 1993-05-21 1993-07-07 Glaxo Group Ltd Chemical compounds
DE19517430A1 (en) 1995-05-12 1996-11-14 Boehringer Mannheim Gmbh Pharmaceutical dosage form of parathyroid hormone with a two to six hour drug release period
UA51652C2 (en) 1995-06-08 2002-12-16 Новартіс Аг A method of hydrogenation of imines
US6544949B1 (en) 1995-07-13 2003-04-08 Societe De Conseils De Recherches Et D'applications Scientifiques, S.A.S. Analogs of parathyroid hormone
US5969095A (en) 1995-07-13 1999-10-19 Biomeasure, Inc. Analogs of parathyroid hormone
US5723577A (en) 1995-07-13 1998-03-03 Biomeasure Inc. Analogs of parathyroid hormone
US5955574A (en) 1995-07-13 1999-09-21 Societe De Conseils De Recherches Et D'applications Scientifiques, S.A. Analogs of parathyroid hormone
US7410948B2 (en) 1995-07-13 2008-08-12 Societe De Conseils De Recherches Et D'applications Scientifiques, Sas Analogs of parathyroid hormone
EP0918774B9 (en) 1996-06-27 2002-04-10 Ligand Pharmaceuticals Incorporated Androgen receptor modulator compounds and methods
US20090264534A1 (en) 1996-11-27 2009-10-22 Dalton James T Selective androgen receptor modulators
FR2770842B1 (en) 1997-11-13 1999-12-17 Oreal NOVEL COMPOUNDS DERIVED FROM N-ARYL 2-HYDROXY ALKYLAMIDES
EP0922467A3 (en) 1997-12-12 2000-05-24 Takeda Chemical Industries, Ltd. Iontophoretic drug delivery
JP4154017B2 (en) 1997-12-30 2008-09-24 久光製薬株式会社 Iontophoresis device and drug unit
US6159959A (en) 1999-05-06 2000-12-12 American Home Products Corporation Combined estrogen and antiestrogen therapy
WO2001036039A2 (en) 1999-11-17 2001-05-25 Novartis Ag Iontophoretic transdermal delivery of peptides
US6544553B1 (en) * 1999-12-28 2003-04-08 Watson Pharmaceuticals, Inc. Dosage forms and methods for oral delivery of progesterone
KR20020075388A (en) 1999-12-30 2002-10-04 시그널 파머슈티컬스 인크 Compounds and methods for modulation of estrogen receptors
KR20020073566A (en) 2000-01-28 2002-09-27 앙도르쉐르슈 인코포레이티드 Selective estrogen receptor modulators in combination with estrogens
EP1299094A2 (en) 2000-06-28 2003-04-09 Bristol-Myers Squibb Company Selective androgen receptor modulators and methods for their identification, design and use
AU2001282064B2 (en) 2000-08-03 2007-02-01 Antares Pharma Ipl Ag Novel composition for transdermal and/or transmucosal administration of active compounds that ensures adequate therapeutic levels
EP1401801B1 (en) 2000-08-24 2006-11-02 The University Of Tennessee Research Corporation Selective androgen receptor modulators and methods of use thereof
US7186683B2 (en) 2000-09-18 2007-03-06 Sanos Bioscience A/S Use of GLP for the treatment, prevention, diagnosis, and prognosis of bone-related and nutrition-related disorders
US7371721B2 (en) 2000-09-18 2008-05-13 Sanos Bioscience A/S Use of GLP-2 and related compounds for the treatment, prevention, diagnosis, and prognosis of bone-related disorders and calcium homeostasis related syndromes
MXPA03003815A (en) 2000-10-26 2004-08-12 Johnson & Johnson Transdermal drug delivery devices having coated microprotrusions.
WO2003011824A1 (en) 2001-07-31 2003-02-13 Bristol-Myers Squibb Company Bicyclic modulators of androgen receptor function
CA2456150A1 (en) 2001-08-13 2003-02-27 Merck & Co., Inc. Selective estrogen receptor modulators
US8853266B2 (en) 2001-12-06 2014-10-07 University Of Tennessee Research Foundation Selective androgen receptor modulators for treating diabetes
EP1465619A1 (en) 2002-01-14 2004-10-13 Nordic Bioscience A/S Suppression of cartilage degradation via the estrogen receptor
ATE345125T1 (en) 2002-02-15 2006-12-15 Endorech Inc BIPHENYL DERIVATIVES AND THEIR USE AS ANTIANDROGENS
US7772433B2 (en) 2002-02-28 2010-08-10 University Of Tennessee Research Foundation SARMS and method of use thereof
ES2350446T3 (en) 2002-04-24 2011-01-24 MERCK SHARP & DOHME CORP. MODULATORS OF ESTROGEN RECEIVERS.
TW200407324A (en) 2002-05-17 2004-05-16 Bristol Myers Squibb Co Bicyclic modulators of androgen receptor function
TW200307553A (en) 2002-05-24 2003-12-16 Akzo Nobel Nv Treatment of post-menopausal complaints in breast cancer patients
AU2003251527A1 (en) 2002-06-13 2003-12-31 Beth Israel Deaconess Medical Center, Inc. Analogs of parathyroid hormone and pth-related protein as bone anabolic agents
SI1557411T1 (en) 2002-07-12 2013-01-31 Astellas Pharma Inc
CA2495383A1 (en) 2002-08-12 2004-02-26 Takeda Pharmaceutical Company Limited Fused benzene derivative and use
IL152574A (en) 2002-10-31 2009-09-22 Transpharma Medical Ltd Transdermal delivery system for dried particulate or lyophilized medications
US7662404B2 (en) 2002-10-31 2010-02-16 Transpharma Medical Ltd. Transdermal delivery system for dried particulate or lyophilized peptides and polypeptides
US7383084B2 (en) 2002-10-31 2008-06-03 Transpharma Medical Ltd. Transdermal delivery system for dried particulate or lyophilized medications
US8133505B2 (en) 2002-10-31 2012-03-13 Transpharma Medical Ltd. Transdermal delivery system for dried particulate or lyophilized medications
AU2003286757B2 (en) 2002-11-01 2009-06-04 Merck Sharp & Dohme Corp. Carbonylamino-benzimidazole derivatives as androgen receptor modulators
UA79504C2 (en) 2002-11-07 2007-06-25 Organon Nv Indols for treating diseases associated with androgen receptors
EP1567487A4 (en) 2002-11-15 2005-11-16 Bristol Myers Squibb Co Open chain prolyl urea-related modulators of androgen receptor function
WO2004080377A2 (en) 2003-03-11 2004-09-23 Neurosearch A/S Kcnq channel modulating compounds and their pharmaceutical use
JP2007500245A (en) 2003-06-10 2007-01-11 スミスクライン ビーチャム コーポレーション Compound
EP1636167A2 (en) 2003-06-10 2006-03-22 SmithKline Beecham Corporation 1-aminonaphthalenes as modulators of androgen, glucocorticoid, mineralocorticoid and progesterone receptors
FI20030958A0 (en) 2003-06-27 2003-06-27 Orion Corp New compounds
WO2005000309A2 (en) 2003-06-27 2005-01-06 Ionix Pharmaceuticals Limited Chemical compounds
BRPI0412029A (en) 2003-06-30 2006-09-05 Alza Corp formulations for coated microprojections containing nonvolatile counterions
CA2531136A1 (en) * 2003-07-04 2005-01-13 Nycomed Danmark Aps Parathyroid hormone (pth) containing pharmaceutical compositions for oral use
CN1849120A (en) 2003-09-10 2006-10-18 默克公司 17-heterocyclic-4-azasteroid derivatives as androgen receptor modulators
US20050124625A1 (en) 2003-10-21 2005-06-09 Salvati Mark E. Piperazine derivatives and their use as modulators of nuclear hormone receptor function
GB0324551D0 (en) 2003-10-21 2003-11-26 Karobio Ab Novel compounds
EP1680154B1 (en) 2003-10-31 2012-01-04 ALZA Corporation Self-actuating applicator for microprojection array
EP1682012A4 (en) 2003-11-13 2008-09-24 Alza Corp Composition and apparatus for transdermal delivery
US7256208B2 (en) 2003-11-13 2007-08-14 Bristol-Myers Squibb Company Monocyclic N-Aryl hydantoin modulators of androgen receptor function
CA2545048A1 (en) 2003-11-20 2005-06-02 Warner-Lambert Company Llc Androgen receptor modulators
IL159273A0 (en) 2003-12-09 2004-06-01 Transpharma Medical Ltd Transdermal delivery system for sustained release of polypeptides
US20070196395A1 (en) 2003-12-12 2007-08-23 Mackerell Alexander Immunomodulatory compounds that target and inhibit the py'binding site of tyrosene kinase p56 lck sh2 domain
WO2005066194A1 (en) 2004-01-07 2005-07-21 Endorecherche, Inc. Helix 12 directed steroidal pharmaceutical products
EP1709021B1 (en) 2004-01-22 2010-08-04 Eli Lilly And Company Selective estrogen receptor modulators for the treatment of vasomotor symptoms
IL160033A0 (en) 2004-01-25 2004-06-20 Transpharma Medical Ltd Transdermal delivery system for polynucleotides
US20050182105A1 (en) 2004-02-04 2005-08-18 Nirschl Alexandra A. Method of using 3-cyano-4-arylpyridine derivatives as modulators of androgen receptor function
US7820702B2 (en) 2004-02-04 2010-10-26 Bristol-Myers Squibb Company Sulfonylpyrrolidine modulators of androgen receptor function and method
EP1725522B1 (en) 2004-03-03 2014-09-10 GlaxoSmithKline LLC Aniline derivatives as selective androgen receptor modulators
US7388027B2 (en) 2004-03-04 2008-06-17 Bristol-Myers Squibb Company Bicyclic compounds as modulators of androgen receptor function and method
US7696241B2 (en) 2004-03-04 2010-04-13 Bristol-Myers Squibb Company Bicyclic compounds as modulators of androgen receptor function and method
US7625923B2 (en) 2004-03-04 2009-12-01 Bristol-Myers Squibb Company Bicyclic modulators of androgen receptor function
GB0405033D0 (en) 2004-03-05 2004-04-07 Karobio Ab Novel pharmaceutical compositions
WO2005090282A1 (en) 2004-03-12 2005-09-29 Ligand Pharmaceuticals Incorporated Androgen receptor modulator compounds and methods
JP2007223901A (en) 2004-03-24 2007-09-06 Takeda Chem Ind Ltd Heterocyclic compound and use thereof
US20080125399A1 (en) 2004-04-08 2008-05-29 Jiabing Wang 17 Beta-Acetamide-4-Azasteroids As Androgen Receptor Modulators
TW200602317A (en) 2004-04-23 2006-01-16 Akzo Nobel Nv Novel androgens
AU2005243240B2 (en) 2004-05-03 2012-03-15 Janssen Pharmaceutica N.V. Novel indole derivatives as selective androgen receptor modulators (SARMS)
BRPI0510808A (en) 2004-05-11 2007-11-06 Pfizer Prod Inc benzonitrile derivatives to treat musculoskeletal fragility
WO2005112984A2 (en) 2004-05-13 2005-12-01 Alza Corporation Apparatus and method for transdermal delivery of parathyroid hormone agents
EP1756101A2 (en) 2004-05-17 2007-02-28 Acadia Pharmaceuticals Inc. Androgen receptor modulators and method of treating disease using the same
JP2008503447A (en) 2004-05-29 2008-02-07 7ティーエム ファーマ エイ/エス Substituted thiazole acetic acid as CRTH2 ligand
GEP20094851B (en) 2004-06-07 2009-12-10 The Univ Of Tennessee Research Foundation Selective androgen receptor modulators and methods of use thereof
CN101056862B (en) 2004-09-10 2013-03-13 詹森药业有限公司 Novel imidazolidin-2-one derivatives as selective androgen receptor modulators (SARMS)
CA2580777A1 (en) * 2004-09-20 2006-03-30 Janssen Pharmaceutica N.V. Novel tetracyclic heteroatom containing derivatives useful as sex steroid hormone receptor modulators
CA2582356A1 (en) 2004-09-30 2006-04-13 Janssen Pharmaceutica N.V. Novel benzimidazole derivatives useful as selective androgen receptor modulators (sarms)
US8143425B2 (en) 2004-10-12 2012-03-27 Bristol-Myers Squibb Company Heterocyclic aromatic compounds useful as growth hormone secretagogues
JP2008515998A (en) 2004-10-13 2008-05-15 スミスクライン ビーチャム コーポレーション Compound
AU2005310238A1 (en) 2004-10-29 2006-06-08 Merck Sharp & Dohme Corp. N-(pyridin-3-yl)-2-phenylbutanamides as androgen receptor modulators
CN104803916B (en) 2004-11-16 2017-08-11 詹森药业有限公司 Hete rocyclic derivatives as SARM (SARMS)
JP2008520331A (en) 2004-11-18 2008-06-19 トランスファーマ メディカル リミテッド Combined microchannel formation and iontophoresis for transdermal delivery of drugs
CA2588607A1 (en) 2004-11-23 2006-06-01 Ptc Therapeutics, Inc. Carbazole, carboline and indole derivatives useful in the inhibition of vegf production
KR20070112775A (en) 2005-01-10 2007-11-27 아카디아 파마슈티칼스 인코포레이티드 Aminophenyl derivatives as selective androgen receptor modulators
EP1871379A4 (en) 2005-04-15 2010-06-02 Glaxosmithkline Llc Cyanoarylamines
SI1891038T1 (en) 2005-05-13 2009-04-30 Lilly Co Eli Substituted n-arylpyrrolidines as selective androgen receptor modulators
US20090170907A1 (en) 2005-06-06 2009-07-02 Smithkline Beecham Corporation Chemical Compounds
US7829589B2 (en) 2005-06-10 2010-11-09 Elixir Pharmaceuticals, Inc. Sulfonamide compounds and uses thereof
US7709516B2 (en) 2005-06-17 2010-05-04 Endorecherche, Inc. Helix 12 directed non-steroidal antiandrogens
CN101203491A (en) 2005-06-24 2008-06-18 伊莱利利公司 Tetrahydrocarbazole derivatives used as androgen receptor modifier
AR057656A1 (en) 2005-07-01 2007-12-12 Ligand Pharm Inc MODULATING COMPOUNDS OF ANDROGEN RECEPTORS AND RELATED METHODS
US8592452B2 (en) 2005-08-01 2013-11-26 Takeda Pharmaceutical Company Limited Cyclic amine compound
JP2008303145A (en) 2005-09-22 2008-12-18 Takeda Chem Ind Ltd Cardiotonic comprising grk inhibitor
US7776859B2 (en) 2005-10-14 2010-08-17 Bristol-Myers Squibb Company Hexahydroimidazopyrazin-3-one compounds useful as modulators of androgen receptor function
WO2007061781A1 (en) 2005-11-18 2007-05-31 3M Innovative Properties Company Coatable compositions, coatings derived therefrom and microarrays having such coatings
TW200730505A (en) 2005-12-07 2007-08-16 Merck & Co Inc Polymorphs of an androgen receptor modulator
AU2006336187A1 (en) 2005-12-28 2007-07-26 Alza Corporation Stable therapeutic formulations
BRPI0706726B8 (en) 2006-01-24 2021-05-25 Janssen Pharmaceutica Nv 2-substituted benzimidazoles as selective androgen receptor modulators (sarms), pharmaceutical composition comprising them and process for preparing
EP1991523B1 (en) 2006-03-03 2012-08-15 Orion Corporation Selective androgen receptor modulators
US20080039775A1 (en) 2006-03-15 2008-02-14 Alza Corporation Apparatus and Method for Transdermal Delivery of Parathyroid Hormone Agents to Prevent or Treat Osteopenia
WO2007124411A1 (en) 2006-04-20 2007-11-01 3M Innovative Properties Company Device for applying a microneedle array
MX2009000385A (en) 2006-07-12 2009-04-06 Univ Tennessee Res Foundation Substituted acylanilides and methods of use thereof.
AU2007275737A1 (en) 2006-07-19 2008-01-24 Osurf Selective androgen receptor modulators, analogs and derivatives thereof and uses thereof
EA016853B1 (en) 2006-08-24 2012-08-30 Юниверсити Оф Теннесси Рисерч Фаундейшн Substituted acylanilides and use thereof
EP2079466B1 (en) 2006-09-29 2014-01-15 GlaxoSmithKline LLC Substituted indole compounds
BRPI0719821B8 (en) 2006-10-03 2021-05-25 Ipsen Pharma Sas storage stable composition suitable for administration to patients
US7803770B2 (en) 2006-10-03 2010-09-28 Radius Health, Inc. Method of treating osteoporosis comprising administration of PTHrP analog
WO2008044033A1 (en) 2006-10-11 2008-04-17 Astrazeneca Ab Amide derivatives
UA98777C2 (en) 2006-11-20 2012-06-25 Эли Лилли Энд Компани Tetrahydrocyclopenta[b]indole compounds as androgen receptor modulators
WO2008121602A1 (en) 2007-03-29 2008-10-09 Smithkline Beecham Corporation Chemical compounds
WO2008124000A2 (en) 2007-04-02 2008-10-16 Ligand Pharmaceuticals Incorporated Thiazole derivatives as androgen receptor modulator compounds
US9284345B2 (en) 2007-04-12 2016-03-15 Endorecherche, Inc. 17alpha-substituted steroids as systemic antiandrogens and selective androgen receptor modulators
US7696227B2 (en) 2007-04-13 2010-04-13 Regents Of The University Of California Small-molecule inhibitors of the androgen receptor
EP2146689B1 (en) 2007-04-16 2020-08-12 Corium, Inc. Solvent-cast microneedle arrays containing active
US8420694B2 (en) 2007-08-07 2013-04-16 Takeda Pharmaceutical Company Limited Pyrrolidin-2-one derivatives as androgen receptor modulator
US20090117158A1 (en) 2007-10-23 2009-05-07 Mahmoud Ameri Transdermal sustained release drug delivery
WO2009065600A2 (en) 2007-11-21 2009-05-28 Technische Universität Dresden Means for treating myosin-related diseases
AU2008339572B2 (en) 2007-12-21 2012-05-10 Astrazeneca Ab Bicyclic derivatives for use in the treatment of androgen receptor associated conditions
ES2488990T3 (en) 2008-02-22 2014-09-01 Radius Health, Inc. Selective androgen receptor modulators
US8268872B2 (en) 2008-02-22 2012-09-18 Radius Health, Inc. Selective androgen receptor modulators
WO2009133861A1 (en) 2008-04-28 2009-11-05 武田薬品工業株式会社 Cyclic amine compound
WO2009137104A1 (en) * 2008-05-09 2009-11-12 Radius Health, Inc. Combination therapy for breastcancer comprising an antiestrogenic agent
KR101278788B1 (en) 2008-05-16 2013-06-25 일라이 릴리 앤드 캄파니 Tetrahydrocyclopenta[b]indole androgen receptor modulators
WO2010022176A1 (en) 2008-08-19 2010-02-25 Ferring International Center S.A. Methods of treatment for skeletal conditons
EP2366401B1 (en) 2008-11-04 2015-07-15 ASKA Pharmaceutical Co., Ltd. Aqueous composition containing follicle-stimulating hormone
CN102264429B (en) 2008-12-26 2014-01-08 久光制药株式会社 Microneedle device
US20100226966A1 (en) 2009-03-03 2010-09-09 Daddona Peter E Method for transdermal controlled release drug delivery
WO2010118287A1 (en) 2009-04-10 2010-10-14 Radius Health, Inc. Selective androgen receptor modulators
KR102462145B1 (en) 2009-10-16 2022-11-03 가부시키가이샤 한도오따이 에네루기 켄큐쇼 Liquid crystal display device and electronic apparatus having the same
US20110172609A1 (en) 2010-01-08 2011-07-14 Ratio, Inc. Microneedle component assembly for drug delivery device
EP2531029B1 (en) 2010-02-04 2016-10-19 Radius Health, Inc. Selective androgen receptor modulators
IE20100174A1 (en) 2010-03-25 2012-02-29 Trinity College Dublin Transdermal administration of peptides
JP6327852B2 (en) 2010-05-04 2018-05-23 コリウム インターナショナル, インコーポレイテッド Methods and devices for transdermal delivery of parathyroid hormone using microprojection arrays
WO2011143469A1 (en) 2010-05-12 2011-11-17 Radius Health,Inc Therapeutic regimens
EP3225247B1 (en) 2010-05-28 2020-09-02 3M Innovative Properties Company Aqueous formulations for coating microneedle arrays
US8642632B2 (en) 2010-07-02 2014-02-04 Radius Health, Inc. Selective androgen receptor modulators
WO2012047617A1 (en) 2010-09-28 2012-04-12 Radius Health, Inc. Selective androgen receptor modulators
WO2012075375A1 (en) 2010-12-02 2012-06-07 Lanco Biosciences, Inc. Delivery of parathyroid hormones by microinjection systems
RU2013151314A (en) 2011-04-22 2015-05-27 Радиус Хэлс, Инк. METHOD FOR DELIVERY OF MEDICINES FOR PTH, PTH-P AND RELATED PEPTIDES
CA2857501C (en) 2011-11-30 2020-06-23 3M Innovative Properties Company Microneedle device having a peptide therapeutic agent and an amino acid, methods of making and using the same

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8399520B2 (en) * 2002-12-26 2013-03-19 Eisai R&D Management Co., Ltd. Selective estrogen receptor modulator
WO2008002490A2 (en) * 2006-06-23 2008-01-03 Radius Health, Inc. Treatment of vasomotor symptoms with selective estrogen receptor modulators

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100105733A1 (en) * 2006-06-23 2010-04-29 Lyttle C Richard Treatment of vasomotor symptoms with selective estrogen receptor modulators
US8933130B2 (en) 2006-06-23 2015-01-13 Radius Health, Inc. Treatment of vasomotor symptoms with selective estrogen receptor modulators
US9555014B2 (en) 2010-05-12 2017-01-31 Radius Health, Inc. Therapeutic regimens
US10071066B2 (en) 2014-03-28 2018-09-11 Duke University Method of treating cancer using selective estrogen receptor modulators
US10420734B2 (en) 2014-03-28 2019-09-24 Duke University Method of treating cancer using selective estrogen receptor modulators
US11779552B2 (en) 2014-03-28 2023-10-10 Duke University Method of treating cancer using selective estrogen receptor modulators
US11951080B2 (en) 2014-03-28 2024-04-09 Duke University Method of treating cancer using selective estrogen receptor modulators
US10385008B2 (en) 2017-01-05 2019-08-20 Radius Pharmaceuticals, Inc. Polymorphic forms of RAD1901-2HCL

Also Published As

Publication number Publication date
MX2012013014A (en) 2013-02-27
US9555014B2 (en) 2017-01-31
WO2011143469A8 (en) 2012-01-19
RS54993B1 (en) 2016-11-30
US20160022608A1 (en) 2016-01-28
EP2568806B1 (en) 2016-05-11
DK2568806T3 (en) 2016-08-15
HUE030072T2 (en) 2017-04-28
ES2585884T3 (en) 2016-10-10
MX342898B (en) 2016-10-18
HRP20160929T1 (en) 2016-10-07
EP2568806A4 (en) 2013-09-11
PT2568806T (en) 2016-08-05
PL2568806T3 (en) 2017-03-31
EP3106159A1 (en) 2016-12-21
EP2568806A1 (en) 2013-03-20
ME02474B (en) 2017-02-20
WO2011143469A1 (en) 2011-11-17
SI2568806T1 (en) 2016-09-30
LT2568806T (en) 2016-09-26
CY1117893T1 (en) 2017-05-17
SMT201600263B (en) 2016-08-31
CA2799183A1 (en) 2011-11-17

Similar Documents

Publication Publication Date Title
US9555014B2 (en) Therapeutic regimens
RU2522212C2 (en) Oxicodon and naloxon-containing pharmaceutical compositions with instant release
JP2015110604A (en) Hrt formulation
TW202228682A (en) Treatment of bipolar disorders and psychosis using dexmedetomidine hydrochloride
EP2716290B1 (en) Dry-coated tablet containing tegafur, gimeracil and oteracil potassium
TW202038954A (en) Montelukast for the treatment of erosive hand osteoarthritis
US20190224208A1 (en) Pharmaceutical composition for treating premature ejaculation and method for treating premature ejaculation
TW202317101A (en) Methods for treating depressive states
US20110027324A1 (en) O-desmethyl-venlafaxine for treating major depressive disorder
CA3137633A1 (en) Composition comprising pridopidine and analog thereof for treating huntington disease and symptoms thereof
CA2973540A1 (en) Treatment of autoimmune disease in a patient receiving additionally a beta-blocker
US20230225994A1 (en) Sublingual epinephrine formulations and methods of use
WO2021130779A1 (en) Pharmaceutical composition of casr modulators and methods and uses thereof
US20200323780A1 (en) Bilayer combination tablet for oral administration containing tramadol and celecoxib
TW202228698A (en) Tablet for use in treating huntington's disease and method of making the same
Agent PrRAN™-PIOGLITAZONE
Tablets et al. RAN-Pioglitazone
Hydrochloride et al. PrMYLAN-PIOGLITAZONE
OA16802A (en) Pharmaceutical composition for treating premature ejaculation and method for treating premature ejaculation.

Legal Events

Date Code Title Description
AS Assignment

Owner name: RADIUS HEALTH, INC., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:O'DEA, LOUIS;LYTTLE, C. RICHARD;GUERRIERO, JONATHAN;REEL/FRAME:029943/0585

Effective date: 20110531

STCB Information on status: application discontinuation

Free format text: EXPRESSLY ABANDONED -- DURING PUBLICATION PROCESS

AS Assignment

Owner name: RADIUS HEALTH, INC., MASSACHUSETTS

Free format text: RELEASE BY SECURED PARTY;ASSIGNORS:MIDCAP FINANCIAL TRUST;MIDCAP FUNDING IV TRUST;REEL/FRAME:053315/0231

Effective date: 20200723

Owner name: RADIUS PHARMACEUTICALS, INC., MASSACHUSETTS

Free format text: RELEASE BY SECURED PARTY;ASSIGNORS:MIDCAP FINANCIAL TRUST;MIDCAP FUNDING IV TRUST;REEL/FRAME:053315/0231

Effective date: 20200723