US20180153904A1 - High-strength testosterone undecanoate compositions - Google Patents

High-strength testosterone undecanoate compositions Download PDF

Info

Publication number
US20180153904A1
US20180153904A1 US15/803,691 US201715803691A US2018153904A1 US 20180153904 A1 US20180153904 A1 US 20180153904A1 US 201715803691 A US201715803691 A US 201715803691A US 2018153904 A1 US2018153904 A1 US 2018153904A1
Authority
US
United States
Prior art keywords
testosterone
capsule
dose
testosterone undecanoate
less
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
US15/803,691
Inventor
Chandrashekar Giliyar
Basawaraj Chickmath
Nachiappan Chidambaram
Mahesh V. Patel
Srinivansan Venkateshwaran
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.)
Lipocine Inc
Original Assignee
Lipocine Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US12/957,206 external-priority patent/US20120135074A1/en
Priority claimed from US13/485,807 external-priority patent/US9034858B2/en
Application filed by Lipocine Inc filed Critical Lipocine Inc
Priority to US15/803,691 priority Critical patent/US20180153904A1/en
Publication of US20180153904A1 publication Critical patent/US20180153904A1/en
Priority to US16/412,360 priority patent/US11433083B2/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/565Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol
    • A61K31/568Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids not substituted in position 17 beta by a carbon atom, e.g. estrane, estradiol substituted in positions 10 and 13 by a chain having at least one carbon atom, e.g. androstanes, e.g. testosterone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/22Heterocyclic compounds, e.g. ascorbic acid, tocopherol or pyrrolidones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/44Oils, fats or waxes according to two or more groups of A61K47/02-A61K47/42; Natural or modified natural oils, fats or waxes, e.g. castor oil, polyethoxylated castor oil, montan wax, lignite, shellac, rosin, beeswax or lanolin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4858Organic 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
    • A61K9/4875Compounds of unknown constitution, e.g. material from plants or animals
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • G01N33/743Steroid hormones
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/52Predicting or monitoring the response to treatment, e.g. for selection of therapy based on assay results in personalised medicine; Prognosis

Definitions

  • the present invention relates to testosterone undecanoate containing pharmaceutical compositions and oral dosage capsules as well as associated methods. Accordingly, this invention involves the fields of chemistry, pharmaceutical sciences, medicine and other health sciences.
  • hypogonadism is a serious condition affecting mostly aging men.
  • the common reasons for hypogonadism in men could be physiological abnormality involving among other factors, improper functioning or growth of the gonads and/or the pituitary-hypothalamus regulatory systems, and aging.
  • Many of the abnormalities that are identified to be commonly associated with the low or decreased testosterone levels include impaired sexual function and/or libido, metabolic syndrome which may be a combination of abdominal obesity, high blood pressure, insulin resistance, lipid disorders; high risk of cardiovascular diseases; reduced bone mass/mineral density and muscle weakness and or degeneration affecting the musculoskeletal system.
  • Other effects of low testosterone levels include negative changes in body composition, depression and other psychological disorders.
  • the average human male produces 4-7 mg of testosterone per day in a circadian pattern, with maximal plasma levels attained in early morning and minimal levels in the evening. It is generally recognized that in a normal adult man of age 17 to 54 years, the serum total testosterone (T) is between about 300 ng/dL to about 1100 ng/dL and this range is referred to as the eugonadal range. Restoration of testosterone levels to the eugonadal range typically corrects many of the cited clinical abnormalities associated with hypogonadism or low testosterone levels.
  • testosterone undecanoate a prodrug which gets converted to testosterone in vivo.
  • Testosterone undecanoate containing products are available in some countries as liquid filled soft-gelatin capsule containing 40 mg of fully solubilized testosterone undecanoate.
  • Testosterone undecanoate is extremely lipophilic (calculated log P of ⁇ 6.5) with a water solubility of ⁇ 0.3 ng/ml and a melting point around 62° C. It is generally believed that in order to promote lymphatic absorption for better safety profile and to facilitate effective oral delivery of testosterone undecanoate, the testosterone undecanoate must be presented in a bioacceptable solubilizer. Accordingly, research continues into the development of testosterone oral delivery products that can have high drug load and provide for practical unit oral dosage forms.
  • a pharmaceutical capsule for oral delivery includes a capsule shell and a capsule fill.
  • the capsule fill can include a solubilizer and about 14 wt % to about 35 wt % testosterone undecanoate based on the total weight of the capsule fill.
  • the oral dosage capsule is such that upon a single oral administration to a male subject of one or more capsules with a total testosterone undecanoate daily dose of about 350 mg to about 650 mg it provides a ratio of serum testosterone C max to serum testosterone C ave of about 2.7 or less.
  • a method for providing a serum concentration of testosterone within a target serum testosterone concentration C ave range for a male subject can include the step of orally administering to the male subject a daily dose of a testosterone undecanoate-containing composition.
  • the testosterone undecanoate can comprise about 14 wt % to about 35 wt % of the testosterone undecanoate-containing composition and the daily dose provided can be about 350 mg to about 420 mg of testosterone undecanoate to the male subject.
  • a method for providing a serum concentration of testosterone within a target serum testosterone concentration C ave range for a male subject can include the step of orally administering to the male subject an initial regimen including a daily dose of a testosterone undecanoate-containing composition.
  • the testosterone undecanoate can comprise about 14 wt % to about 35 wt % of the testosterone undecanoate-containing composition and the daily dose provided can be about 350 mg to about 650 mg of testosterone undecanoate to the male subject.
  • the method can include a step of determining the serum testosterone concentration for the male subject on at least one titration node day within the initial regimen.
  • the method further can include the step of orally administering to the male subject a maintenance regimen including a daily dose of testosterone undecanoate-containing composition that comprises about 14 wt % to about 35 wt % of the testosterone undecanoate.
  • the maintenance regimen can provide a daily dose of testosterone undecanoate to the subject based on the serum testosterone concentration determined by a titration metric on the at least one titration node day of the initial regimen and the said maintenance daily dose can be sufficient to provide a serum testosterone plasma concentration within the target range.
  • the method can provide a steady state ratio of serum testosterone C max to C ave of 2.7 or less.
  • capsule fill compositions with a lower TU loading of less than 14% w/w are unsuitable for optimal activity due to inadequate bioavailability which also leads to larger dosage forms/doses for therapy in treatment of symptoms related to male hypogonadism.
  • higher loading dosage forms i.e. those with ⁇ 14% w/w of TU based on total capsule fill, resulted in a superior C ave per mg testosterone undecanoate administered, thus providing improved performance of oral testosterone undecanoate as a testosterone therapy.
  • an oral capsule with TU loading in the range of about 14 wt % to about 18% is provided in which no more than 80 wt % of the TU in the capsule fill is dissolved. This capsule can result in dose normalized C max suggesting superior bioavailability yet manageable C max for effective therapeutics.
  • treatment when used in conjunction with the administration of pharmaceutical compositions and oral dosage capsules containing testosterone undecanoate, refers to the administration of the oral dosage capsules and pharmaceutically acceptable composition to subjects who are either asymptomatic or symptomatic.
  • “treatment” can both be to reduce or eliminate symptoms associated with a condition present in a subject, or it can be prophylactic treatment, i.e. to prevent the occurrence of the symptoms in a subject.
  • prophylactic treatment can also be referred to as prevention of the condition.
  • formulation and “composition” are used interchangeably and refer to a mixture of two or more compounds, elements, or molecules. In some aspects the terms “formulation” and “composition” may be used to refer to a mixture of one or more active agents with a carrier or other excipients.
  • dosage form can include one or more formulation(s) or composition(s) provided in a format for administration to a subject.
  • oral such terms refer to compositions, formulations, or dosage forms formulated and intended for oral administration to subjects.
  • fatty acid refers to unionized carboxylic acids with a long aliphatic tail (chain), either saturated or unsaturated, conjugated or non-conjugated.
  • C 8 to C 22 fatty acid glycerides refers to a mixture of mono-, di-, and/or tri-glycerol esters of medium to long chain (C 8 to C 22 ) fatty acids.
  • the term “dispersant” refers to any pharmaceutically acceptable additive that enables the contents of the compositions and/or oral dosage capsules to disperse in an aqueous medium.
  • the extent of dispersion in an aqueous medium can be determined spectrophotometrically from the absorbance exhibited by the dispersion at a wavelength of about 400 nm.
  • the dispersion of the composition (with or without the testosterone undecanoate) of the current invention in about 0.2 mM sodium lauryl sulphate solution in water has an absorbance of about 0.6 or less at about 400 nm wavelength, when the ratio of the composition to the sodium lauryl sulphate solution is about 1:2000.
  • the dispersion of the composition (with or without the testosterone undecanoate) of the current invention in about 0.2 mM sodium lauryl sulphate solution in water has an absorbance of about 0.3 or less at about 400 nm wavelength, when the ratio of the composition to the sodium lauryl sulphate solution is about 1:5000.
  • the composition can produce a fine dispersion upon dilution in an aqueous medium without the need of a hydrophilic surfactant.
  • the dispersant of the current invention is at least one selected from the group of hydrophilic surfactant or lipophilic surfactant.
  • the dispersant includes a hydrophilic surfactant.
  • solidifying agent or “solidifying additive” are used interchangeably and refer to a pharmaceutically acceptable additive that is in a solid physical state at 20° C.
  • solid lipophilic additive refers to a lipophilic compound or component that is in a solid physical state at 20° C. and/or renders the composition or dosage form non-liquid, such as solid or semi-solid.
  • the terms “solubilized” and “solubility,” when used to describe the state of testosterone undecanoate with respect to a composition and/or capsule fill, refer to the absence of testosterone undecanoate crystals in the composition or oral dosage form when observed under hot-stage microscope over a temperature of about 25° C. to about 65° C., or the absence of crystalline testosterone undecanoate melting related peak (about 62 to about 65° C.) when the composition or oral dosage form is subjected to differential scanning calorimetry.
  • solubility of testosterone undecanoate in a particular compound e.g.
  • compositions having crystalline forms of testosterone undecanoate at about room temperature would be considered to have an unsolubilized fraction and a solubilized fraction of testosterone undecanoate which fraction includes testosterone undecanoate in a solid state that is not crystalline such as amorphous and solid solution which are solubilized but undissolved.
  • undissolved or “non-dissolved” can be used interchangeably and when one is used to describe the state of testosterone undecanoate with respect to a composition and/or capsule fill refers to the testosterone undecanoate in a non-liquid testosterone undecanoate-containing composition that is solubilized (such as non-crystalline) and non-solubilized such as crystalline TU.
  • the solubility of TU in the composition can be estimated based on the individual solubility in the composition components.
  • TU solubility in the composition at room temperature (RT) may be estimated based on the observed values at higher temperature such as at 37° C.
  • dissolved when used to describe the state of testosterone undecanoate (TU) with respect to a composition or capsule fill refers to a testosterone undecanoate-containing liquid solution having no undissolved testosterone undecanoate.
  • lipophilic refers to compounds that are not freely soluble in water; and the term “lipophilic surfactant” refers to surfactants that have HLB values of about 10 or less. Conversely, the term “hydrophilic” refers to compounds that are soluble in water; and term “hydrophilic surfactant” refers to surfactants that have HLB values of more than about 10.
  • ionizable fatty acid refers to a fatty acid compound that changes its HLB as a function of pH.
  • oleic acid is predominantly lipophilic at lower pH values (such as those found in the stomach) but becomes a predominantly hydrophilic at higher pH values (such as those found in the intestine).
  • subject refers to a mammal that may benefit from the administration of a drug composition or method of this invention.
  • subjects include humans.
  • the subject can be a human male.
  • the subject can be a hypogonadal male.
  • the testosterone deficiency or hypogonadism in a male human subject refers to a condition wherein the average baseline plasma testosterone concentration (T-C avg-B ) is about 300 ng/dL or less.
  • testosterone deficiency or hypogonadism in a male human subject refers to a condition wherein the average baseline plasma testosterone concentration is about 400 ng/dL or less.
  • a “responder” is a subject who responds to exogenous oral testosterone undecanoate therapy.
  • “Responder analysis” is the assessment of the effectiveness of testosterone undecanoate therapy in a group of subjects deemed to get benefits of oral TU therapy.
  • group refers to a collection of at least 24 human male subjects who receive and respond to exogenous oral administration of the compositions disclosed herein, namely testosterone undecanoate-containing compositions.
  • the group can include at least 100 or at least 300 male subjects.
  • the group can include at least 1000 male subjects.
  • the subjects can be hypogonadal subjects.
  • oral administration represents any method of administration in which an active agent can be administered by swallowing, chewing, or sucking of the dosage form.
  • the composition of the current inventions can be admixed with food or drink prior to being orally consumed.
  • a “dosing regimen” or “regimen” such as an “initial dosing regimen” or a “maintenance dosing regimen” refers to how, when, how much, and for how long a dose of the compositions of the present invention can be administered to a subject.
  • an initial dosing regimen for a hypogonadal male subject may provide for a total daily dose of 600 mg administered in two divided doses at least 12 hours apart (e.g. once with breakfast and once with dinner) with meals having about 25-55 g of fat content repeated daily for 30 days.
  • “daily dose” refers to the amount of active agent (e.g. testosterone undecanoate) administered to a subject over a 24 hour period of time.
  • the daily dose can be administered two or more administrations during the 24 hour period. In one embodiment, the daily dose provides for two administrations in a 24 hour period.
  • an “initial dose” or initial daily dose” refers to a dose administered during the initial regimen or period of a dosing regimen.
  • An initial dose includes both the very first dose during the initial regimen as well as the subsequent doses during the same initial regimen.
  • a “maintenance dose” or “maintenance daily dose” refers to a dose administered during a maintenance regimen of a dosing regimen.
  • the maintenance dose follows a dose titration based on the serum testosterone determination on a titration node day, however the maintenance dose does not need to be of a different quantity as the initial dose or the previous maintenance dose (in the case of multiple titrations).
  • non-liquid when used to refer to the state of a composition disclosed herein refers to the physical state of the composition as being a semi-solid or solid.
  • solid and “semi-solid” refers to the physical state of a composition that supports its own weight at standard temperature and pressure, and has adequate viscosity or structure to not freely flow. Semi-solid materials may conform to the shape of a container under applied pressure.
  • titration or “dose titration” or “dose adjustment” are used interchangeably and refer to an increase or decrease of the total daily dose of testosterone undecanoate administered to a subject, typically based on the response of the subject to the exogenous administered testosterone undecanoate.
  • the dose can be increased or decreased based on the measurement of serum testosterone concentration after a steady state has been achieved.
  • steady state refers to the achievement of a stable response in serum total testosterone levels to exogenously administered testosterone undecanoate, typically achieved after at least 15 days following the start of a dosing regimen.
  • the titration can also include the adjustment of the way the total daily dose is administered such as whether it is administered as two or three doses within a 24 hour period, whether it is administered with a meal, with a meal with a particular fat content, or at a particular hour of the day.
  • initial daily dose is a dose of testosterone undecanoate administered daily to a subject in need of testosterone therapy.
  • the initial daily dose may be administered in two or more intervals over a 24 hour period, e.g. twice-a-day.
  • maintenance daily dose or “daily dose of the maintenance regiment” is a dose of testosterone undecanoate administered daily to a subject in need of testosterone therapy as determined based on measurement of the titration node day titration metric and is the daily dose going forward within a few days of measurement unless a dose change is needed based on a another titration node day measurements.
  • During a maintenance regime there may be two or more daily doses administered which at some point during the regime would be considered to be the maintenance daily dose.
  • titration node or “titration node day” are used interchangeably and refer to a day on which a serum sample is drawn from a subject for measurement of the serum testosterone concentrations in order to determine whether a testosterone undecanoate dose titration is necessary and what the titration type might need to be.
  • the measured serum testosterone levels may also be used to determine dose a titration metric to be utilized in deciding dose titration needs for an individual subject.
  • dosing regimens can include one or more titration node day the term may refer to a first titration node during a dosing regimen (e.g. between the initial dosing regimen and the maintenance dosing regimen) or it can refer to a subsequent titration node day between a maintenance dosing regimen and a subsequent maintenance dosing regimen.
  • titration day refers to the day when administration of a newly titrated (adjusted) dose is initiated. It should be noted that one or more titrations can be conducted to arrive at a maintenance daily dose in a maintenance regimen. Thus, the maintenance daily dose and regimen is considered to be the dose based on the last or most recent titration.
  • titration metric is a pharmacokinetic (PK) parameter determined from a serum sample on a titration node day.
  • the PK parameter used as the titration metric can include the serum testosterone C max , C avg , C min , C pre-dose or C t (serum concentration at a particular time of day).
  • the titration metric can be used to aid in the determination of whether a dose titration is necessary, its magnitude, and other factors possibly included in the titration adjustment.
  • release and “release rate” are used interchangeably to refer to the discharge or liberation of a substance, including without limitation a drug, from the dosage form into a surrounding environment such as an aqueous medium either in vitro or in vivo.
  • an “effective amount” or a “therapeutically effective amount” of a drug refers to a non-toxic, but sufficient amount of the drug, to achieve therapeutic results in treating a condition for which the drug is known to be effective. It is understood that various biological factors may affect the ability of a substance to perform its intended task. Therefore, an “effective amount” or a “therapeutically effective amount” may be dependent in some instances on such biological factors. Further, while the achievement of therapeutic effects may be measured by a physician or other qualified medical personnel using evaluations known in the art, it is recognized that individual variation and response to treatments may make the achievement of therapeutic effects a somewhat subjective decision. The determination of an effective amount is well within the ordinary skill in the art of pharmaceutical sciences and medicine. See, for example, Meiner and Tonascia, “Clinical Trials: Design, Conduct, and Analysis,” Monographs in Epidemiology and Biostatistics , Vol. 8 (1986), incorporated herein by reference.
  • the term “delayed release” refers to the release into an aqueous solution of the testosterone undecanoate from the composition or oral dosage form in a time delayed manner attributed either to the inherent nature of the composition or to a coating which may surround the composition or the oral dosage form.
  • a traditional gelatin or non-gelatin non-enteric capsule shell does not alone constitute a delayed release mechanism.
  • the delayed release is such that about 20% or less of the testosterone undecanoate is released within the first 15 minutes after the composition is contacted by the aqueous solution.
  • plasma testosterone concentration “testosterone concentration in the blood,” and “serum testosterone concentration” are used interchangeably and refer to the “total” testosterone concentration which is the sum of the bioavailable testosterone including free and protein-bound testosterone concentrations.
  • total testosterone concentration which is the sum of the bioavailable testosterone including free and protein-bound testosterone concentrations.
  • testosterone concentration refers to serum total testosterone concentration.
  • the average baseline plasma testosterone concentration of a human male is the arithmetic mean of the total plasma testosterone concentrations determined on at least two consecutive time points that are reasonably spaced from each other, for example from about 1 hour to about 168 hours apart.
  • the plasma testosterone concentration can be determined on at least two consecutive times that are about 12 hours to about 48 hours apart.
  • the plasma testosterone concentration of the human male can be determined at a time between about 5 o'clock and about 11 o'clock in the morning.
  • the plasma testosterone concentration can be the determined by standard analytical procedures and methods available in the art, such as for example, automated or manual immunoassay methods, liquid chromatography or liquid chromatography-tandem mass spectrometry (LC-MSMS) etc.
  • AUC 0-t is the area under the curve of a plasma-versus-time graph determined for the analyte from the time 0 to time “t”.
  • C avg As used herein, the term “C avg ,” “C ave ,” or “C-average” are used interchangeably, and is determined as the AUC 0-t or the mean AUC divided by the time period (t).
  • C avg-8 h is the average plasma concentration over a period of 8 hours post-dosing determined by dividing the AUC 0-8 value by 8.
  • C avg-12 h is the average plasma concentration over a period of 12 hours post-dosing determined by dividing the AUC 0-12 value by 12
  • C avg-24 h is the average plasma concentration over a period of 24 hours post-dosing determined by dividing the AUC 0-24 h , value by 24, and so on.
  • all C ave values are considered to be C ave-24 h .
  • C t refers to the serum concentration of testosterone at time “t” prior to or after administration of the dosage of the current invention.
  • the time “t” is generally in hours, unless otherwise specified.
  • a C t of “C ( ⁇ 2 to 0) refers to serum testosterone concentration measured in sample collected between the time of about 2 hours before and just immediately prior to dosage administration to the subject tested.
  • C t of “C (2 to 4) ” refers to serum testosterone concentration measured in sample collected between the time of about 2 hours and 4 hours after administration of a dosage to the subject tested.
  • SIF or “simulated intestinal fluid” refers to “intestinal fluid, simulated TS” in accordance with the USP.
  • the SIF does not contain pancreatic enzyme.
  • SIF may be a fed or fasted simulated intestinal aqueous solution comprising phosphatidyl choline and from about 2 mM to 20 mM bile salts.
  • SGF or “simulated gastric fluid” refers to “Gastric fluid, Simulated TS” in accordance with the USP.
  • the SGF does not contain the enzyme pepsin.
  • the SGF may also be a simple 0.1 N HCl solution in water.
  • free of or substantially free of of a particular compound or compositions refers to the absence of any separately added portion of the referenced compound or composition. Free of or substantially free of can include the presence of 1 wt % or less (based on total composition weight) of the referenced compound which may be present as a component or impurity of one or more of the ingredients.
  • the term “about” is used to provide flexibility to a numerical range endpoint by providing that a given value may be “a little above” or “a little below” the endpoint.
  • a plurality of items, structural elements, compositional elements, and/or materials may be presented in a common list for convenience. However, these lists should be construed as though each member of the list is individually identified as a separate and unique member. Thus, no individual member of such list should be construed as a de facto equivalent of any other member of the same list solely based on their presentation in a common group without indications to the contrary.
  • a pharmaceutical capsule for oral delivery includes a capsule shell and a capsule fill.
  • the capsule fill can include a solubilizer and about 14 wt % to about 35 wt % testosterone undecanoate based on the total weight of capsule fill.
  • the oral dosage capsule is such that when a single oral administration to a male subject of one or more capsules with a total testosterone undecanoate daily dose of about 350 mg to about 650 mg it provides a ratio of serum testosterone C max to serum testosterone C ave of about 2.7 or less.
  • compositions and oral dosage capsules of the present invention can be used to treat subjects, particularly human males, or even more particularly males who suffer from testosterone deficiency or hypogonadism. Accordingly, in one embodiment of the present invention, a method for providing a serum concentration of testosterone within a target serum testosterone concentration C ave range for a male subject is provided. The method includes the step of orally administering to the male subject a daily dose of a testosterone undecanoate-containing composition.
  • the testosterone undecanoate comprises about 14 wt % to about 35 wt % of the testosterone undecanoate-containing composition and the daily dose provides about 350 mg to about 420 mg of testosterone undecanoate to the male subject.
  • the testosterone undecanoate comprises about 14 wt % to about 18 wt % of the testosterone undecanoate-containing composition.
  • Testosterone deficiency is typically associated with a particular condition that is the source or causes the deficiency.
  • the compositions and oral dosage capsules of the present invention can be used to treat any condition associated with testosterone deficiency, including complete absence, of endogenous testosterone.
  • conditions associated with testosterone deficiency include, but are not limited to congenital or acquired primary hypogonadism, hypogonadotropic hypogonadism, cryptorchidism, bilateral torsion, orchitis, vanishing testis syndrome, orchidectomy, Klinefelter's syndrome, post castration, eunuchoidism, hypopituitarism, endocrine impotence, infertility due to spermatogenic disorders, impotence, male sexual dysfunction (MSD) including conditions such as premature ejaculation, erectile dysfunction, decreased libido, and the like, micropenis and constitutional delay, penile enlargement, appetite stimulation, testosterone deficiency associated with chemotherapy, testosterone deficiency associated with toxic damage from alcohol, testosterone deficiency associated with toxic damage from heavy metal, osteoporosis associated with androgen deficiency, and combinations thereof.
  • MSD male sexual dysfunction
  • compositions and oral dosage forms disclosed herein include idiopathic gonadotropin, LHRH deficiency, or pituitary hypothalamic injury from tumors, trauma, or radiation. Typically, these subjects have low serum testosterone levels but have gonadotropins in the normal or low range.
  • the compositions or oral dosage forms may be used to stimulate puberty in carefully selected males with clearly delayed puberty not secondary to pathological disorder.
  • the compositions and oral dosage forms may be used in female-to-male transsexuals in order to maintain or restore male physical and sexual characteristics including body muscle mass, muscle tone, bone density, body mass index (BMI), enhanced energy, motivation and endurance, restoring psychosexual activity etc.
  • the testosterone undecanoate compositions and oral dosage capsules may be useful in providing hormonal male contraception.
  • testosterone therapy can also be used to improve the quality of life of subjects suffering from conditions such as decreased libido, diminishing memory, anemia due to marrow failure, renal failure, chronic respiratory or cardiac failure, steroid-dependent autoimmune disease, muscle wasting associated with various diseases such as AIDS, preventing attacks of hereditary angioedema or urticaria; andropause, and palliating terminal breast cancer.
  • certain biomarkers such as for example, increased SHBG levels, can be used to diagnose a subject who may be in need of testosterone therapy.
  • biomarkers can be associated with conditions/disease states such as anorexia nervosa, hyperthyroidism, hypogonadism, androgen insensitivity/deficiency, alcoholic hepatic cirrhosis, primary biliary cirrhosis, and the like.
  • Subjects that can be treated by the testosterone undecanoate compositions and oral dosage capsule of the present disclosure can be any human male in need thereof.
  • the human male may be at least 14 years of age.
  • the human male is an adult of at least age 30.
  • the subject can be an adult male of at least age 50.
  • the subject can be an adult male of at least age 60.
  • the compositions and oral dosage capsules disclosed herein can be used to treat testosterone deficiency in human males.
  • the human male being treated can have an average baseline plasma testosterone concentration of about 400 ng/dL or less.
  • the human male being treated can have an average baseline plasma testosterone concentration of about 350 ng/dL or less.
  • the human male being treated can have an average baseline plasma testosterone concentration of about 300 ng/dL or less.
  • the human male being treated can have an average baseline plasma testosterone concentration of about 250 ng/dL or less.
  • the human male being treated can have an average baseline plasma testosterone concentration of about of about 190 ng/dL or less.
  • the human male has an average baseline plasma testosterone concentration of about 400 ng/dL or less, along with a co-morbid condition of insulin resistance.
  • the human male being treated can have a low density lipoproteins (LDL) level in greater than about 130 mg/dL of blood.
  • the human male being treated can have a high density lipoproteins (HDL) level less than about 40 mg/dL of blood.
  • the human male being treated can have a total cholesterol level greater than about 220 mg/dL of blood.
  • the human male being treated can have an average TG (triglycerides) levels greater than 250 mg/dL of blood.
  • the testosterone undecanoate dosage forms of the current invention can be administered to human male whose bioavailable or free or un-bound plasma estradiol levels are about 20 pg/mL or less. In another embodiment, dosage forms of the current invention can be administered to human male who has a ratio of the bioavailable or free or unbound plasma testosterone level to the bioavailable or free or un-bound plasma estradiol level at about 100 or less.
  • the testosterone undecanoate compositions and oral dosage capsules of the current invention can be administered orally to a human male who has an average body mass index (BMI) of about 28 kg/m 2 or more.
  • BMI body mass index
  • the human male has an average BMI of about 30 kg/m 2 or more.
  • the human male has an average BMI of about 37 kg/m 2 or more.
  • the subject male being treated can have a serum sex hormone binding globulin (SHBG) levels of about 40 nmol/L or more.
  • the human male being treated can have a serum SHBG levels of about 60 nmol/L or more.
  • the pharmaceutical compositions and oral dosage capsules of the present invention have the ability to provide for increased stability of the testosterone undecanoate present in the formulation.
  • the pharmaceutical compositions and oral dosage capsules of the present invention can provide for superior stability with respect to the degradation of the testosterone undecanoate that can occur during storage as compared to other formulation containing lower testosterone undecanoate concentration.
  • the pharmaceutical compositions and oral dosage capsules of the present invention can have increased stability such that, when stored for a period of at least three months there is at least 20% less degradation of the testosterone undecanoate as compared to testosterone undecanoate containing compositions having less than 14 wt % testosterone undecanoate.
  • the pharmaceutical compositions and oral dosage capsules of the present invention can have increased stability such that, when stored for a period of at least three months there is at least 20% less degradation of the testosterone undecanoate as compared to testosterone undecanoate containing compositions having less than 16 wt % testosterone undecanoate.
  • the pharmaceutical compositions and oral dosage capsules disclosed herein can provide therapeutically effective treatment without the need to include oils, triglycerides, and/or hydrophilic surfactants. Accordingly, in one embodiment, the pharmaceutical compositions and oral dosage capsules can be free of oil. In another embodiment, the pharmaceutical composition and oral dosage capsules can be free of triglycerides. In one embodiment, the composition or capsule fill can comprise 25 wt % or less of total triglycerides. In one embodiment, the composition or capsule fill can be free of ionizable fatty acids. In another embodiment, the composition or capsule fill can be free of oleic acid.
  • testosterone undecanoate-containing compositions or capsule fill that comprise greater than 25 wt % triglycerides have a higher dependence on digestion upon oral administration than do those in which the triglycerides comprise 25 wt % or less of the total composition or capsule fill.
  • the capsule fill can comprise 15 wt % or less of triglycerides.
  • the capsule fill can comprise 10 wt % or less of triglycerides.
  • the capsule fill can comprise 5 wt % or less of triglycerides.
  • the pharmaceutical compositions and oral dosage capsules can be free of hydrophilic surfactants.
  • the composition can include a hydrophilic surfactant as a dispersant and the hydrophilic surfactant can be present in an amount such that it does not appreciably solubilize the testosterone undecanoate in the composition.
  • a hydrophilic surfactant is said to “not appreciably solubilize” testosterone undecanoate when it solubilizes 5 wt % or less of the testosterone undecanoate in the composition or the dosage form.
  • a hydrophilic surfactant is deemed to “not appreciably solubilize” testosterone undecanoate when it solubilizes 2 wt % or less of the testosterone undecanoate in the composition or oral dosage capsule.
  • the pharmaceutical compositions and oral dosage capsules can still be capable of providing the necessary dispersion and pharmacokinetics parameters to effectively treat testosterone deficiency.
  • the testosterone undecanoate can be present in the pharmaceutical compositions and oral dosage capsules in amounts sufficient to comprise 14 wt % to about 35 wt % of the composition or capsule fill.
  • the testosterone undecanoate can make up about 15 wt % to about 30 wt % of the composition or oral dosage capsule.
  • the oral dosage capsule can comprise about 18 wt % to about 25 wt % of the composition or oral dosage capsule.
  • the compositions and/or capsule fill material can be such that the testosterone undecanoate comprises about 14 wt % to about 18 wt % of the total composition or capsule fill.
  • At least 35 wt % of the testosterone undecanoate in the composition or capsule fill can be in dissolved form. In yet another embodiment, at least about 66 wt % of the testosterone undecanoate in the capsule fill or composition can be present in dissolved form. In another embodiment, at least about 5 wt % of the testosterone undecanoate in the capsule fill or composition can be present in undissolved form.
  • the oral dosage capsules of the present application can include dosages of testosterone undecanoate of at least 50 mg.
  • the oral dosage capsules of the present application can include dosages of testosterone undecanoate of about 80 mg to about 400 mg.
  • the oral dosage capsule can include about 80 mg to about 140 mg testosterone undecanoate.
  • the oral dosage capsule can include about 120 mg to about 300 mg testosterone undecanoate.
  • the oral dosage capsule can include about 150 mg to about 250 mg of testosterone undecanoate.
  • the compositions and oral dosage capsule can be used as part of dosing regimens to provide daily doses of about 250 mg to about 650 mg per day, preferably, daily doses of about 350 mg to about 650 mg per day
  • the solubilizers used in the pharmaceutical compositions and oral dosage capsules of the present invention play role in the ability of the formulation to provide the desired therapeutic characteristics.
  • Solubilizers that can be used can be selected from a variety of compounds and mixtures of compounds that have the ability to facilitate loading of testosterone undecanoate.
  • the solubilizer can comprise about 50 wt % to about 86 wt % of the composition or capsule fill. In one embodiment, the solubilizer can comprise about 55 wt % to about 82 wt % of the pharmaceutical composition or oral dosage capsule. In another embodiment, the solubilizer can comprise about 60 wt % to about 80 wt % of the pharmaceutical composition or oral dosage capsule.
  • the solubilizer can be such that the testosterone undecanoate can have solubility in the solubilizer, at about 37° C., of about 250 mg/g to about 750 mg/g (mg testosterone undecanoate/gram of solubilizer and testosterone undecanoate).
  • solubilizers that can be used include C 8 to C 22 fatty acid glycerides, omega fatty acids, and mixtures thereof.
  • the C 8 to C 22 fatty acid glycerides can include C 8 to C 22 medium and/or long chain monoglycerides, medium and/or long chain diglycerides, or mixtures of a mixture of medium and/or long chain monoglycerides and medium and/or long chain diglycerides.
  • the solubilizer can consist essentially of medium and/or long chain monoglycerides and/or diglycerides.
  • Medium to long chain monoglycerides and diglycerides refers to compounds having chain lengths of C 8 to C 22 .
  • the mixture of monoglycerides and diglycerides can have chain lengths of C 8 to about C 13 . In another embodiment, the mixture of monoglycerides and diglycerides can have chain lengths of about C 14 to about C 22 .
  • monoglycerides can comprise at least about 40 wt % of the C 8 to C 22 fatty acid glycerides (such as commercially available Maisine® 35-1, Capmul® MCM, Peceol,® and the like). In another embodiment, the monoglycerides can comprise at least about 60 wt % of the C 8 to C 22 fatty acid glycerides. In yet a further embodiment, the monoglycerides can comprise at least about 80 wt % of the C 8 to C 22 fatty acid glycerides.
  • Non-limiting examples of C 8 to C 22 fatty acid glycerides that can be used as solubilizers in pharmaceutical compositions and oral dosage capsules of the present invention include monoglycerides and/or diglycerides derived from sources such as maize oil, poppy seed oil, safflower oil, sunflower oil, borage seed oil, coconut oil, palm kernel oil, castor oil, and mixtures thereof.
  • the solubilizer can also include a triglyceride.
  • the triglyceride can be a medium and/or long chain triglyceride, or mixture thereof, and can be present alone or with other solubilizers.
  • the triglycerides can be selected from a variety of well-known pharmaceutically acceptable triglycerides including, but not limited to vegetable oils such as peanut oil, safflower oil, sunflower oil, olive oil, castor oil, corn oil, maize oil, flax seed oil, wheat-germ oil and the like, or their hydrogenated derivatives and their mixtures thereof.
  • Additional triglyceride sources can include animal derived oils such as fish oil, seal oil, whale oil, and the like, triglycerides of C 8 -C 22 fatty acids or their mixtures; triglycerides of C 8 -C 13 fatty acids; triglycerides of C 14 -C 22 fatty acids.
  • the composition can include a fatty acid triglyceride and the testosterone undecanoate can comprise at least about 25 wt % of the composition.
  • the triglyceride can be castor oil.
  • the castor oil can comprise about 45 wt % or less of the total composition.
  • the castor oil can comprise about 40 wt % or less of the solubilizer.
  • the composition can be free of castor oil.
  • the solubilizer can include a glyceryl palmitostearate, a glyceryl stearate, a glyceryl distearate, glyceryl monostearate, or a combination thereof.
  • the solubilizer can include a C 8 to C 22 fatty acid glycerides that is monoglycerides and/or diglycerides of capric acid, caprylic acid, or mixtures thereof. In another embodiment, the solubilizer can include a C 8 to C 22 fatty acid glycerides that is a monoglycerides and/or diglycerides of linoleic acid, oleic acid, or mixtures thereof.
  • C 8 to C 22 fatty acids that can be used include capric acid, pelargonic acid, caprylic acid, undecanoic acid, lauric acid, myristic acid, palmitic acid, stearic acid, oleic acid, linoleic acid, linolenic acid, arachodonic acid, eicosapentaenoic acid, docosahexanoic acid, and mixtures thereof.
  • the C 8 to C 22 fatty acid can be capric acid, caprylic acid, undecanoic acid, lauric acid, myristic acid, palmitic acid, stearic acid, oleic acid, linoleic acid, linolenic acid or mixtures thereof.
  • the C 8 to C 22 fatty acid can be selected from the group consisting of capric acid, caprylic acid, oleic acid, linoleic acid, and mixtures thereof.
  • the composition or capsule fill can be free of ionizable fatty acids. In another embodiment, the composition or capsule fill can be free of oleic acid.
  • the solubilizer can include an alcohol.
  • alcohols that can be used as solubilizers include tocopherol, ethyl alcohol, isopropanol, butanol, benzyl alcohol, ethylene glycol, propylene glycol, butanediol, glycerol, pentaerythritol, transcutol, dimethyl isosorbide, polyethylene glycol and mixtures thereof.
  • the solubilizer can be ethyl alcohol, benzyl alcohol, tocopherol, and mixtures thereof.
  • compositions and oral dosage capsules can also include a dispersant.
  • the dispersant can be a hydrophilic surfactant having an HLB value of greater than 10, a lipophilic surfactant having an HLB value of 10 or less, or combinations thereof.
  • the compositions and oral dosage forms can include at least one hydrophilic surfactant.
  • the capsule fill includes at least one hydrophilic surfactant and at least one lipophilic surfactant.
  • the total amount of lipophilic component is the total amount in wt % of the lipophilic components including the mono-, di- and/or tri-glycerides and the lipophilic surfactants, if present in the composition.
  • the lipophilic surfactant includes the solubilizer and the lipophilic surfactant.
  • the total amount of hydrophilic surfactant is the total amount (in wt %) of the added hydrophilic surfactant and that hydrophilic surfactant formed in situ in an aqueous medium as a function of pH (e.g. intestinal pH) due to the hydrophilic ionized ionizable fatty acid (e.g. oleate) formed from lipophilic unionized ionizable fatty acid (oleic acid).
  • fatty acid such as oleic acid may be a good solubilizer for testosterone undecanoate
  • bioavailability and activity is substantially compromised with fatty acid containing compositions either by being unable to continue to solubilize the drug, or be inadequate facilitator for chylomicron related testosterone undecanoate absorption or can be slow to allow drug to partitioning out of the carrier.
  • hydrophilic surfactant can, but does not have to have appreciable solubilizing effect for the testosterone undecanoate present in the composition.
  • hydrophilic surfactants that can be included are non-ionic hydrophilic surfactants such as polysorbates, polyoxyethylene hydrogenated vegetable oils, polyoxyethylene vegetable oils; polyoxyethylene sorbitan fatty acid esters; polyoxyethylene-polyoxypropylene block copolymers; polyglycerol fatty acid esters; polyoxyethylene glycerides; polyoxyethylene sterols, derivatives and analogues thereof reaction mixtures of polyols and at least one member of the group consisting of fatty acids, glycerides, vegetable oils, hydrogenated vegetable oils, fractionated oils and sterols; tocopheryl polyethylene glycol succinates; sugar esters; sugar ethers; sucroglycerides; mixtures thereof; alkylglucosides; alkylmaltosides; alkylthioglucosides; lauryl
  • the hydrophilic surfactant can have at least one characteristic of: 1) being present in an amount such that it does not appreciably solubilize testosterone undecanoate present in the composition; or 2) the solubility of testosterone undecanoate in the hydrophilic surfactant at about 25° C., is less than 100 mg/gram or less, based on the total weight of the testosterone undecanoate and the solubilizer.
  • the hydrophilic surfactant can have at least one characteristic of: 1) being present in an amount such that it solubilizes less than 5 wt % of the testosterone undecanoate present in the composition; or 2) the solubility of testosterone undecanoate in the hydrophilic surfactant at about 25° C., is less than 100 mg/gram or less, based on the total weight of the testosterone undecanoate and the surfactant.
  • the hydrophilic surfactant can have at least one characteristic of: 1) the hydrophilic surfactant is present in an amount such that it solubilizes less than 5 wt % of the testosterone undecanoate present in the composition; or 2) the solubility of testosterone undecanoate in the hydrophilic surfactant at about 25° C., is about 50 mg/gram or less, based on the total weight of the testosterone undecanoate and the surfactant.
  • the hydrophilic surfactant can have a least one characteristic of: 1) the hydrophilic surfactant is present in an amount such that it solubilizes less than 5 wt % of the testosterone undecanoate present in the composition; or 2) the solubility of testosterone undecanoate in the hydrophilic surfactant at about 25° C. about 10 mg/gram or less, based on the total weight of the testosterone undecanoate and the surfactant.
  • the hydrophilic surfactant can have the characteristic of: 1) the hydrophilic surfactant is present in an amount such that it solubilizes less than 5 wt % of the testosterone undecanoate present in the composition; and 2) the solubility of testosterone undecanoate in the hydrophilic surfactant at about 25° C., is about 50 mg/gram or less, based on the total weight of the testosterone undecanoate and the surfactant.
  • compositions and oral dosage capsules can include at least one lipophilic surfactant.
  • lipophilic surfactants can be used including, but not limited to reaction mixtures of alcohols or polyalcohols with a variety of natural and/or hydrogenated oils such as PEG-5 hydrogenated castor oil, PEG-7 hydrogenated castor oil, PEG-9 hydrogenated castor oil, PEG-6 corn oil (Labrafil® M 2125 CS), PEG-6 almond oil (Labrafil®M 1966 CS), PEG-6 apricot kernel oil (Labrafil®M 1944 CS), PEG-6 olive oil (Labrafil®M 1980 CS), PEG-6 peanut oil (Labrafil®M 1969 CS), PEG-6 hydrogenated palm kernel oil (Labrafil®.
  • natural and/or hydrogenated oils such as PEG-5 hydrogenated castor oil, PEG-7 hydrogenated castor oil, PEG-9 hydrogenated castor oil, PEG-6 corn oil (Labrafil® M 2125 CS), PEG-6 almond oil (Labrafil®M 1966 CS), PEG
  • propylene glycol fatty acid esters such as propylene glycol monolaurate (Lauroglycol FCC), propylene glycol ricinoleate (Propymuls), propylene glycol monooleate (Myverol P-O6), propylene glycol dicaprylate/dicaprate (Captex® 200), and propylene glycol dioctanoate (Captex® 800), propylene glycol mono-caprylate (Capryol® 90); propylene glycol fatty acid esters, such as propylene glycol monolaurate (Lauroglycol FCC), propylene glycol ricinoleate (Propymuls), propylene glycol monooleate (Myverol P-O6), propylene glycol dicaprylate/dicaprate (Captex® 200), and propylene glycol dioctanoate (Captex® 800), propylene glycol mono-caprylate (Capryol® 90); propylene glycol fatty acid est
  • the lipophilic surfactant can be selected from the group consisting of propylene glycol mono caprylate, propylene glycol oleate, propylene glycol monostearate, propylene glycol monolaurate, propylene glycol mono-oleate, propylene glycol dicaprylate/dicaprate, sorbitan monooleate, PEG-5 hydrogenated castor oil, PEG-7 hydrogenated castor oil, PEG-9 hydrogenated castor oil, PEG-6 corn oil, PEG-6 almond oil, PEG-6 apricot kernel oil, PEG-6 olive oil, PEG-6 peanut oil, PEG-6 hydrogenated palm kernel oil, sorbitan monolaurate (Arlacel 20), sorbitan monopalmitate, sorbitan monooleate, sorbitan monostearate, sorbitan tristearate, sorbitan monolaurate, sorbitan monopalmitate, sorbitan monooleate, sorbitan trioleate, sorbitan sesqui
  • the pharmaceutical compositions and/or oral dosage capsules can include a solidifying agent.
  • a solidifying agent is a pharmaceutically acceptable additive that is in a solid physical state at 20° C.
  • solidifying agents facilitate the solidification of the pharmaceutical compositions of the present invention at temperatures around room temperature.
  • the compositions and capsule fill of the present invention can be non-liquid at standard temperature and pressure.
  • the composition and capsule fill can be semi-solid at standard temperature and pressure.
  • the composition and capsule fill can be solid at standard temperature and pressure.
  • the solidifying agent can comprise from about 0.1 wt % to about 25 wt % of the pharmaceutical composition or oral dosage capsule.
  • the solidifying agent can comprise about 2 wt % to about 20 wt % of the composition or oral dosage capsule. In yet a further embodiment, the solidifying agent can comprise about 3 wt % to about 15 wt % of the composition or oral dosage capsule. In still a further embodiment, the solidifying agent can comprise about 3 wt % to about 9 wt % of the capsule fill. In yet a further embodiment, the solidifying agent can comprise 6 wt % to 9 wt % of the capsule fill. In one embodiment, the solidifying agent can melt at a temperature of about 45° C. to about 75° C.
  • Non-limiting examples of solidifying agents that can be used include polyethylene glycols; sorbitol; gelatin; stearic acid; cetyl alcohol; cetosterayl alcohol; paraffin wax; polyvinyl alcohol; glyceryl stearates; glyceryl distearate; glyceryl monostearate; glyceryl palmitostearate; glyceryl behenate; waxes; hydrogenated castor oil; hydrogenated vegetable oil; bees wax, microcrystalline wax; sterols; phytosterols; cholesterol and mixtures thereof.
  • the solidifying agent includes a polyethylene glycol (PEG) having molecular weight from about 1000 to about 20,000 and their mixtures.
  • the solidifying agent includes one or more selected from the group consisting of polyethylene glycol; gelatin; stearic acid; polyvinyl alcohol; glyceryl stearates; glyceryl distearate; glyceryl monostearate; glyceryl palmitostearate; hydrogenated castor oil; hydrogenated vegetable oil and cholesterol.
  • the pharmaceutical composition can be a solid at about 20° C.
  • the solubilized solid and/or the undissolved crystalline testosterone undecanoate can act as a solidifying agent.
  • the oral compositions of the present invention can be formulated to take any dosage form commonly known in the pharmaceutical arts such as granules, tablet or capsule.
  • the oral dosage form can be a capsule having a pharmaceutical composition of the present invention disposed therein. Both soft and hard gelatin and non-gelatin capsules can be used.
  • the capsule size can be any size known in the art and can vary depending on the desired dosage amount.
  • the capsule can be a hard gelatin capsule having a fill volume of about 0.3 mL to about 1.1 mL.
  • the oral dosage capsules can be immediate release, extended release, targeted release, enteric release, delayed release dosage form or combinations thereof. In a specific embodiment, the oral dosage capsule can be a delayed release dosage form.
  • the capsule can have a ratio of the amount of testosterone undecanoate to the volume of the capsule fill can be about 80 mg/mL to about 750 g/mL. In another embodiment, the capsule can have a ratio of the amount of testosterone undecanoate to the volume of the capsule fill can be about 160 mg/mL to about 375 mg/mL.
  • the oral dosage capsules of the present invention can be formulated such that they have distinctive release profiles.
  • an oral dosage capsule can provide in vitro release of at least about 75 wt % of the testosterone undecanoate during the first 120 minutes when tested using about 1000 mL of 8% w/v Triton X-100 in water maintained at about 37 ⁇ 1° C. taken in a USP-Type II dissolution apparatus set at 100 rpm.
  • the oral dosage capsule can have an in vitro release profile such that 85 wt % or less of the testosterone undecanoate is released in the first 30 minutes, when measured using about 1000 mL of 8% w/v Triton X-100 in water maintained at about 37 ⁇ 1° C.
  • the oral dosage capsule can have an in vitro release profile such that 70 wt % or less of the testosterone undecanoate is released in the first 30 minutes, when measured using about 1000 mL of 8% w/v Triton X-100 in water maintained at about 37 ⁇ 1° C. taken in a USP-Type II dissolution apparatus set at 100 rpm.
  • the oral dosage capsule can have a in vitro release profile such that at least 35 wt % of the testosterone undecanoate is released in the first 30 minutes, when measured using about 1000 mL of 8% w/v Triton X-100 in water maintained at about 37 ⁇ 1° C.
  • the oral dosage capsule can have an in vitro release profile such that at least 40 wt % of the testosterone undecanoate is released in the first 30 minutes, when measured using about 1000 mL of 8% w/v Triton X-100 in water maintained at about 37 ⁇ 1° C. taken in a USP-Type II dissolution apparatus set at 100 rpm.
  • the dosage form can comprise two or more of populations of testosterone undecanoate compositions of the present invention.
  • at least one of the populations can be formulated to start releasing testosterone undecanoate immediately into a surrounding aqueous medium.
  • at least one the populations can be formulated to start releasing testosterone undecanoate after at least 2 hours.
  • at least one the populations can be formulated to release testosterone undecanoate after about 4 hours, or after about 6 hours, or after about 8 hours, or after about 10 hours.
  • At least one of the populations can be formulated to start releasing testosterone undecanoate immediately after oral administration to a human. In one particular case, at least one of the populations can be formulated to start releasing testosterone undecanoate in the duodenal region after oral administration to a human. In another particular case, at least one of the populations can be formulated to start releasing testosterone undecanoate in the small intestine after oral administration to a human.
  • At least one of the populations includes a pH sensitive substance.
  • at least one of the populations can be formulated to start releasing testosterone undecanoate at a pH of from about 1.0 to about 3.4.
  • at least one of the populations can be formulated to start releasing testosterone undecanoate at a pH of from about 3.5 to about 5.5.
  • at least one of the populations can be formulated to start releasing testosterone undecanoate at a pH of from about 5.6 to about 6.8.
  • at least one of the populations can be formulated to start releasing testosterone undecanoate at a pH about 7.0 or more.
  • the dosage form comprising two or more of populations of testosterone undecanoate compositions of the present invention is a capsule.
  • the dosage form is a capsule in capsule dosage form.
  • the dosage form is a tablet in capsule dosage form.
  • the dosage form is a granules or pellets or tablets or minitablets disposed in a capsule.
  • the oral dosage capsules of the present invention can be formulated such that, when administered to a human male they provide a serum total testosterone C avg ranging about 300 ng/dL to about 1100 ng/dL. In another embodiment, the oral dosage capsules can be formulated such that, upon single administration to a human male, they provide a serum total testosterone C avg ranging about 350 ng/dL to about 800 ng/dL. In another embodiment, the oral dosage capsules can be formulated such that, upon single administration to a human male, they provide a serum total testosterone C avg ranging from about 400 ng/dL to about 600 ng/dL. It is noted that such C avg value can be achieved based on administration every 12 hours or every 8 hours.
  • the oral dosage capsules can be formulated such that, upon single administration to a human male, they provide a serum testosterone undecanoate C avg of about 1.5 ng/mL to about 1 mcg/mL. In a further embodiment, the oral dosage capsules can be formulated such that, upon single administration to a human male, they provide a serum testosterone undecanoate C avg of about 3 ng/mL to about 850 ng/mL. In a further embodiment, the oral dosage capsules can be formulated such that, upon single administration to a human male, they provide a serum testosterone undecanoate C avg of about 10 ng/mL to about 850 ng/mL.
  • the capsule upon a single dose administration of the capsule to a subject the capsule provides a dose-normalized serum testosterone C max of about 3 ⁇ 10 ⁇ 6 dL ⁇ 1 or higher. In another embodiment, upon a single dose administration of the capsule to a subject the capsule provides a dose-normalized serum testosterone C avg of about 1.9 ⁇ 10 ⁇ 6 dL ⁇ 1 or higher. In yet another embodiment, upon a single dose administration of the capsule to a subject the capsule provides a dose-normalized serum testosterone C avg of about 2.7 ⁇ 10 ⁇ 6 dL ⁇ 1 or higher.
  • the oral dosage capsules can be formulated such that upon single administration d to a male human subject they provide a ratio of serum testosterone undecanoate C avg to serum total testosterone C avg of about 4:1 to about 75:1.
  • the oral dosage capsules can be formulated such that, upon single administration to a human male, they provide a ratio of serum testosterone undecanoate C avg to serum total testosterone C avg of about 20:1 to about 50:1.
  • the oral dosage capsules can be formulated such that, upon single administration to a human male, the oral dosage capsule provides a ratio of serum total testosterone C avg to dose of testosterone undecanoate of about 0.2 ⁇ 10 ⁇ 6 dL ⁇ 1 to about 20 ⁇ 10 ⁇ 6 dL ⁇ 1 .
  • a single dose of the testosterone undecanoate composition or oral dosage form can provide a serum total testosterone C avg of about 300 ng/dL or more from about 0.5 hours to about 24 hours after oral administration with a meal or a snack.
  • a single dose of a testosterone undecanoate composition or oral dosage capsule can provide a serum total testosterone C avg of about 300 ng/dL or more at about 20 hours after oral administration with a meal or a snack.
  • a single dose of the testosterone undecanoate composition can provide a serum total testosterone C avg of about 300 ng/dL or more at about 18 hours after oral administration with a meal or a snack.
  • a single dose of the testosterone undecanoate composition can provide a serum total testosterone C avg of about 300 ng/dL or more at about 16 hours after oral administration with a meal snack. In still a further embodiment, a single dose of the testosterone undecanoate composition can provide a serum total testosterone C avg of about 300 ng/dL or more at about 12 hours after administration after oral administration with a meal snack. In still a further embodiment, a single dose of the testosterone undecanoate composition can provide a serum total testosterone C avg of about 300 ng/dL or more at about 8 hours after oral administration with a meal or snack.
  • the meal that is administered with the composition or oral dosage form can be a standard meal or a snack.
  • compositions and oral dosage capsules disclosed herein can be, but do not have to be, orally administered with food.
  • the composition or oral dosage capsule can be administered with a meal, such as a meal that provides about 200 to about 1000 calories of energy.
  • the composition or oral dosage capsule can be administered with a standard meal.
  • the composition or oral dosage capsule can be administered with a meal that provides about 50% of the calories derived from the fat.
  • the composition or oral dosage capsule can be administered with a high-fat, high calorie meal.
  • the composition or oral dosage capsule can be administered with a meal that provides about 500 to about 1000 calories of energy.
  • the composition or oral dosage capsule can be administered with a meal that provides about 400 to about 700 calories derived from the fat therein.
  • the compositional make-up of the meals that are administered can vary depending on the tastes and dietary needs of a subject. However, in some situations it may be beneficial to administer the compositions and oral dosage forms with meals that provide no fat or up to about 50 g of fat.
  • the meal can provide about 10 g to about 50 g of fat.
  • the meal can provide about 30 g of fat.
  • the testosterone undecanoate dosage compositions and oral dosage capsules disclosed herein can be orally administered in a 24 hours' dosing regimen (also referred to as or a daily dosing regimen) that is suitable to the needs of the subject.
  • the 24 hours' dosing regimen can include administering the dosage forms after meals in the morning, at about noon, in the evening, at about night time or combinations thereof.
  • the 24 hours' dosing regimen can include dosing one or more dosage units at one or more administration times.
  • the pharmaceutical composition is administered as a single oral dosage capsule.
  • the testosterone undecanoate compositions and oral dosage capsules can provide increased bioavailability as compared to other testosterone undecanoate compositions and dosage forms.
  • the testosterone undecanoate oral dosage capsules can provide an in vitro release of less than about 85 wt % of the testosterone undecanoate within the first 30 minutes.
  • the testosterone undecanoate oral dosage capsules can provide an in vitro release of about 90 wt % or less testosterone undecanoate within the first 30 minutes.
  • the in vitro release is determined in about 1000 mL of 8% w/v Triton X-100 in water maintained at about 37° C. in an USPType-2 Apparatus at about 100 rpm.
  • these testosterone undecanoate oral dosage capsules i.e. those having the above release characteristics, provide at least a 10% increase in the testosterone undecanoate AUC after single oral dosages are administered to human males.
  • the increase is as compared to equivalent dosages of testosterone undecanoate in an immediate release dosage forms administered under same conditions.
  • Immediate release dosage forms are defined as being dosage forms which release more than 95 wt % of the testosterone undecanoate within the first 30 minutes using the same in vitro release conditions described above.
  • the testosterone undecanoate oral dosage capsules can provide at least a 15% increase in the testosterone undecanoate AUC as compared to an immediate release dosage oral dosage form.
  • the testosterone undecanoate oral dosage capsules disclosed herein can provide at least a 10% reduction in the inter-subject variability of the testosterone undecanoate C max , and/or the testosterone undecanoate AUC as compared to immediate release equivalent dosage containing oral dosage forms.
  • the testosterone undecanoate oral dosage capsules disclosed herein can provide 10% or more testosterone bioavailability in subjects as compared to immediate release equivalent dosed oral dosage forms.
  • compositions and oral dosage capsules of the present invention can be formulated such that upon administration of one or more capsules daily to each subject in a group of at least 24 hypogonadal males for a period of at least 84 days, the capsule provides a serum testosterone C avg of 300 ng/dL to 1100 ng/dL in at least 75% of the hypogonadal males in the group.
  • the capsule can be such that at least one of the following regarding the PK parameter of the administration is true: a) a serum testosterone C max of less than 1500 ng/dL in at least 85% of the hypogonadal males in the group; b) a serum testosterone C max of about 1800 ng/dL to about 2500 ng/dL in 5% or less of the hypogonadal males in the group; or c) a serum testosterone C max greater than 2500 ng/dL in about 1% or less of the hypogonadal males in the group.
  • the administration over the at least 84 days can be divided into two dosing regimens including an initial regimen in which an initial dose is administered and a maintenance regimen in which a maintenance dose is administered.
  • the daily dose of the maintenance regimen of the testosterone undecanoate can be about 45% to about 155% of the initial daily dose.
  • the daily dose of the maintenance regimen of the testosterone undecanoate can be about 66% to about 133% of the initial daily dose.
  • the daily dose of the maintenance regimen of the testosterone undecanoate can be about 75% to about 125% of the initial daily dose.
  • the capsule can be formulated such that the amount of the daily dose of the maintenance regimen is based on at least one testosterone undecanoate dose titration metric derived from the measurement of the serum testosterone concentration on at least one titration node day.
  • the titration node day can be any single day beginning on day 15 to day 84 after administration of the initial daily dose.
  • the titration node day can be any day from day 15 to day 21 after administration the daily dose of the initial regimen of the initial regimen.
  • the titration node day is any day from day 22 to day 30 after administration of the daily dose of the initial regimen of the initial regimen.
  • the titration node day can be any day from day 22 to day 30, from day 31 to day 63, or from day 64 to day 84 after administration of the daily dose of the initial regimen of the initial regimen.
  • the titration based on the titration metric can be such that if the testing of the metric is serum testosterone concentration and the testing is done at time (t) of 1 to less than 3 hours following administration the serum testosterone concentration is less than 12.0 ng/mL then the daily TU dose may need to be increased and if the serum testosterone concentration is more than 9.4 then the daily TU dose may need to be decreased.
  • the titration metric is serum testosterone concentration and the testing is done at time (t) ii) if at time (t) of 3 to less than 8 hours following administration the serum testosterone concentration is less than 4.1 ng/mL daily TU dose may need to be increased and if the serum testosterone concentration is more than 18.1 then the daily TU dose may need to be decreased.
  • the titration metric is serum testosterone concentration and the testing is done at time (t) of 8 to less than 12 hours following administration the serum testosterone concentration is less than 3.0 ng/mL then the daily TU dose may need to be increased and if the serum testosterone concentration is more than 7.8 then the daily TU dose may need to be decreased.
  • the titration metric is serum testosterone concentration and the testing is done at time (t) of 12 to less than 14 hours following administration the serum testosterone concentration is less than 1.4 ng/mL then the daily TU dose may need to be increased and if the serum testosterone concentration is more than 2.9, then the daily TU dose may need to be decreased.
  • the above titrations are only exemplary of the possible titrations that can be accomplished using the methods of the present invention. It is further noteworthy that the titrations can be used in conjunction with the methods taught herein including the disclosed methods of providing a serum concentration of testosterone within a target serum testosterone C ave range for a male subject.
  • the testosterone undecanoate compositions and oral dosage capsules disclosed herein can be used in conjunction with or as a component of a diagnostic or treatment kit that enables the diagnosis and treatment of a male patient in need of testosterone therapy.
  • the diagnostic or treatment kit may comprise the testosterone undecanoate composition or oral dosage capsule disclosed herein along with one or more other components, including, but not limited to 1) instructions to enable those ordinarily skilled in the art to prepare a dosage form for immediate dispensing to the subject in need of; 2) one or more containers filled with one or more of the ingredients of the oral pharmaceutical dosage forms of the invention.
  • Suitable containers include, for example, a bottle, a box, a blister card, a foil packet, or a combination thereof; 3) a tamper proof container or packaging; 4) other pharmaceutical dosage forms including other active agents including PDE-5 inhibitors and glucocorticosteroids; 5) Notice or printed instructions: in a form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of the manufacture, use, or sale for human administration to treat a condition that could be treated by oral testosterone therapy; 6) A “planner” for monitoring and tracking administration of the oral dosage forms; 7) Containers for storing and transporting the components of the kit; 8) total testosterone or free testosterone testing kits; 9) Sex Hormone binding globulin, SHBG, testing kits; 10) Body mass index testing materials to identify high risk patients; 11) tests for identifying patients with hypogonadism; 12) tests to assess testicular function or impotency; 13) test for bone mineral density/osteo
  • compositions and oral dosage capsules disclosed herein can be co-administered with other active agents in order to treat a target condition.
  • One or more co-administered active agents can be admixed with the testosterone undecanoate containing compositions and/or oral dosage forms of the current invention.
  • phosphodiesterase type 5 (PDE-5) inhibitors such as sildenafil citrate, tadalafil, vardenafil, avanafil, lodenafil, mirodenafil, udenafil, and the like, are used to block the degradative action of phosphodiesterase type 5 enzyme on cyclic GMP in the smooth muscle cells lining the blood vessels supplying the corpus cavernosum of the penis and are frequently used to treat erectile dysfunction.
  • compositions and oral dosage forms of the present invention could be co-administered with the compositions and oral dosage forms of the present invention in order to provide improved clinical outcomes through synergistic pharmacological action as measured by improved (sooner, better and longer lasting) erection, potency, libido, mood, body mass, etc. in males relative to administration of the testosterone or the co-administered PDE-5 alone.
  • the testosterone undecanoate compositions and oral dosage capsules can also be co-administered with one or more other active agents such as aromatase inhibitors (for example letrozole, anastrozole, exemestane, fadrozole, vorozole, formestane etc.), dopamine agonists (for example apomorphine, bromocriptine, cabergoline, pergolide, ropinirole, rotigotine, pramipexole, fenoldopam etc.), prostaglandins (for example alprostadil), alpha blockers (for example yohimbine, phentolamine), vasodilators (for example minoxidil) and the like, for improved clinical outcomes through synergistic pharmacological action as measured by improvements in one or more of the secondary sexual characteristics in males such as sexual activity, potency, libido, erection etc., mood, body mass and the like, relative to administration of either the testosterone or the co
  • the subjects receiving the dosage form of this invention are expected to improve in quality of life.
  • the patient reported outcome may be employed to measure the improvement in other levels apart from primary (serum testosterone C ave ) and secondary (serum testosterone C max , C min , C trough , etc) outcomes which are typically quantified by the PK profiles.
  • the improvements in the symptoms are usually monitored by ranking or scoring, dairy recording etc, by the subject being treated and/or the partner or spouse of the subject, in a timely manner before and during the therapy.
  • the dosage forms and the methods of current invention improve sexual symptoms including but not limited to sexual activity engagement, sexual thoughts or fantasies; feel of sexual desire; frequency of experience of morning erections; maintaining erections as long as desired; hardness of erection; ejaculation; enjoyment/satisfaction of sexual activity.
  • the dosage form and the methods of current invention improves or enhances the physical and physiological symptoms and body energy level as assessed by the level of happiness with the body looks; body muscle mass, body weight and weakness/strength of muscles; level of tiredness; level of physical tiredness; level of energy; level of exhaustion and the like.
  • the dosage form and the methods of the current invention improves the symptoms related to the sleep symptoms and memory/cognition as assessed by quality of sleep at night; frequency of sleep restfulness; number of wake-up times during the night; frequency of feeling of satisfactory rest, frequency of accidental doze off during the day; frequency of purposely taken naps during the day; focus attention to tasks; level of forgetfulness; desire or ambition to take on new projects; short attention span; successful/efficient completion of tasks.
  • compositions of the current invention can be formulated to provide a gastro-retentive dosage form.
  • the gastro-retentive dosage form is a capsule.
  • the gastro-retentive dosage form is retained in the upper gastro-intestinal tract for at least one hour post-dosing.
  • the gastro-retentive dosage form is retained in the upper gastro-intestinal tract for at least two hours post-dosing.
  • the gastro-retentive dosage form is retained in the upper gastro-intestinal tract for at least 4 hours post-dosing.
  • the gastro-retentive dosage form is formulated to float in the stomach after dosing.
  • the gastro-retentive dosage form is formulated to expand when it comes in contact with aqueous medium to at least 1.3 times its size compared to its size when it is not in contact with the aqueous use environment. In another embodiment, the gastro-retentive dosage form is formulated to adhere to the lining of the stomach wall after dosing.
  • compositions and the oral dosage capsules of the current invention can also include one or more of other additives selected from binders, bufferants, diluents, disintegrants, flavors, colorants, taste-masking agents, resins, pH modifiers, lubricants, glidants, thickening agent, opacifying agent, humectants, desiccants, effervescing agents, plasticizing agents and the like.
  • other additives selected from binders, bufferants, diluents, disintegrants, flavors, colorants, taste-masking agents, resins, pH modifiers, lubricants, glidants, thickening agent, opacifying agent, humectants, desiccants, effervescing agents, plasticizing agents and the like.
  • compositions and oral dosage capsules of the present invention a method for providing a serum concentration of testosterone within a target serum testosterone concentration C ave range for a male subject is also provided. It is noted that the compositions and oral dosage capsules of the present invention can be used in the conjunction with this method and that the teachings regarding the compositions and their administration provided above can be applied and used in connection with the methods disclosed here.
  • the method includes the step of orally administering to the male subject an initial regimen including a daily dose of a testosterone undecanoate-containing composition.
  • the testosterone undecanoate comprises about 14 wt % to about 35 wt % of the testosterone undecanoate-containing composition and the daily dose provides about 350 mg to about 650 mg of testosterone undecanoate to the male subject.
  • the method includes a step of determining a dose titration metric based on a measurement of serum testosterone concentration for the male subject on at least one titration node day within the initial regimen.
  • the method further includes the step of orally administering to the male subject a maintenance regimen including a daily dose of testosterone undecanoate-containing composition that comprises about 14 wt % to about 35 wt % of the testosterone undecanoate.
  • the maintenance regimen provides a daily dose of testosterone undecanoate to the subject based on the titration metric determined on the at least one titration node day of the initial regimen and is sufficient to provide a serum testosterone plasma concentration that is closer to or within the target range.
  • the method for providing a serum concentration of testosterone within a target serum testosterone concentration C ave range for a male subject can further include the steps of determining a dose titration metric based on a measurement of serum testosterone concentration for the male subject on at least one titration node day within the maintenance regimen. Following the determination of the titration metric the method includes the step of orally administering to the male subject a second maintenance regimen including a daily dose of testosterone undecanoate-containing composition, wherein the testosterone undecanoate-containing composition comprises about 14 wt % to about 35 wt % of the testosterone undecanoate-containing composition.
  • the second maintenance regimen provides a daily dose of testosterone undecanoate to the subject based on the titration metric determined on the at least one titration node day of the maintenance regimen sufficient to provide a serum testosterone plasma concentration within the target range.
  • the steps of determining the titration metric and administering an additional maintenance regimen can be repeated as needed in order to achieve the a serum testosterone concentration within the target range.
  • the daily dose administered in the initial regimen can be the same dosage amount as the daily dose administered in the maintenance regimen.
  • the daily dose of the maintenance regimen can provide an amount of testosterone undecanoate that is about 45% to about 155% of that of the initial daily dose.
  • the daily dose of the maintenance regimen can provide an amount of testosterone undecanoate that is about 66% to about 133% of that of the daily dose in the initial regimen.
  • the daily dose of the maintenance regimen can provide an amount of testosterone undecanoate that is about 75% to about 125% of that of the daily dose in the initial regimen.
  • the determination step or steps of the above described methods can be any single day from day after day 15 of the initial regimen.
  • the titration node day can be any single day from day 15 to day 21 following the start of the initial regimen.
  • the titration node day can be any single day from day 21 to day 30 following the start of the initial regimen.
  • the titration node day is any single day from day 31 to day 63 following the start of the initial regimen.
  • the titration node day can be any single day from day 64 to day 84 following the start of the initial regimen.
  • subjects with whom the methods of the present invention can be used can be those in need of testosterone therapy.
  • the male subject with whom the method is being used can have a serum testosterone C avg of less than 300 ng/dL before beginning of the initial regimen.
  • the male subject can have a serum testosterone C avg of less than 200 ng/dL before beginning of the initial regimen.
  • the male subject can have a serum testosterone C avg of less than 300 ng/dL before beginning of the maintenance regimen.
  • the male subject can have a serum testosterone C avg of less than 200 ng/dL before beginning of the maintenance regimen.
  • the target serum testosterone C ave range can vary depending on the subject and his particular needs and physiological parameters.
  • the target serum testosterone C ave range can be about 300 ng/dL to 1100 ng/dL and is achieved by the method on or after day 84 following the start of the initial regimen.
  • the target serum testosterone C ave range can be about 300 ng/dL to about 1100 ng/dL and is achieved by the method on or after day 120 following the start of the initial regimen.
  • the target serum testosterone C ave range can be about 300 ng/dL to about 1100 ng/dL and can be achieved by the method on or after day 180 following the start of the initial regimen.
  • the method of the present invention can be such that the method can provide a serum testosterone C avg in the range of 300 ng/dL to 1100 ng/dL in 75% or more of hypogonadal males after 84 days from the start of the initial regimen, based on a minimum group size of 24 hypogonadal males.
  • the method can provide a serum testosterone C max of 1500 ng/dL or less in less than or equal to 85% of hypogonadal males based on a minimum group size of 24 hypogonadal males.
  • the method provides a serum testosterone C max in the range of 1800 ng/dL to 2500 ng/dL in about 5% or less of hypogonadal males after 84 days from the start of the initial regimen based on a minimum group size of 24 hypogonadal males.
  • the method can provide a serum testosterone C max of 2500 ng/dL in about 1% or less of hypogonadal males after 84 days from the start of the initial regimen based on a minimum group size of 24 hypogonadal males.
  • the method can provide a steady state ratio of serum testosterone C max to C ave of 2.7 or less based on single subject administration.
  • the above disclosed methods provide for initial and maintenance regimens that include daily dose amounts that can be provided as twice-a-day administrations or divided into multi-dosage administrations.
  • a multi-dosage administration is utilized to provide the daily dose amount of testosterone undecanoate the dosages can be equal or unequal and can be administered with or without meals, depending of the designated regimen.
  • the dosages, whether twice-a-day or multi-time dosages, are administered with a meal or a snack the meal or snack can include about 15 g to about 60 g of fat.
  • the method provides for administration of the daily dose during the maintenance regimen as including twice-a-day administration of the testosterone undecanoate-containing composition in conjunction with meals.
  • the meal administered with the testosterone undecanoate-containing composition can have a total calorie content of about 350 and 1200 K calories with about 30% to about 60% of the calories in the meal being derived from fat.
  • the method can provide, at steady state, a dose-normalized serum testosterone C max of about 3 ⁇ 10 ⁇ 6 dL ⁇ 1 or higher when administered to a male subject with meals daily in two divided doses.
  • the formulations provide 440 mg to 490 mg of testosterone undecanoate per day to an individual when administered twice-a-day as one or two unit dosage forms per administration (e.g., 2-4 unit dosage forms per day).
  • the formulations provide about 450 mg or about 474 mg of testosterone undecanoate per day to an individual when administered twice-a-day as one or two unit dosage forms per administration (e.g., 2-4 unit dosage forms per day).
  • the weight percent refers to the weight percent of the capsule fill composition.
  • Glyceryl mono or di- linoleate Vegetable — — — — Y — — — — — — — — — — glycerides (e.g. Glyceryl mono or di- oleate) Glyceryl — — — — — — — — — — mono or di- stearate Glyceryl — — — — — — — — — — — — palmito- stearate (9Z)-Octadec- — — — — — — — — — — — — 9-enoic acid Octadecanoic — — — — — — — — — — — — acid (9Z,12Z)- — — — — — — — — — — — — 9,12-
  • Carrier I Compositions composed of solubilizer (Propylene glycol mono or di- laurate), hydrophilic additives, and other additives for Composition A to E Carrier Compositions (w/w %) Hydrophilic additives 1 (e.g. % Release in Cremophor 2 RH 8% Triton Propylene 40, Tween 3 80, Aqueous Media glycol SLS, Poloxamer 4 Other additives 7 ⁇ 50% in 2 Comp. mono or 407, Polymer 5 , and/ Anti- Solidify Combi- Total % hrs and ⁇ 75% I No.
  • solubilizer Propylene glycol mono or di- laurate
  • hydrophilic additives e.g. % Release in Cremophor 2 RH 8% Triton Propylene 40, Tween 3 80, Aqueous Media glycol SLS, Poloxamer 4
  • Carrier II Compositions composed of solubilizer (Propylene glycol mono or di- caprylate), hydrophilic additives, and other additives for Composition A to E Carrier Compositions (w/w %) Hydrophilic additives 1 (e.g. % Release in Cremophor 2 RH 8% Triton Propylene 40, Tween 3 80, Aqueous Media glycol SLS, Poloxamer 4 Other additives 7 ⁇ 50% in 2 Comp. mono or 407, Polymer 5 , and/ Anti- Solidify Combi- Total % hrs and ⁇ 75% II No.
  • solubilizer Propylene glycol mono or di- caprylate
  • hydrophilic additives e.g. % Release in Cremophor 2 RH 8% Triton Propylene 40, Tween 3 80, Aqueous Media glycol SLS, Poloxamer 4
  • Carrier III Compositions composed of solubilizer (Corn glycerides: e.g. Glyceryl mono or di-linoleate), hydrophilic additives, and other additives for Composition A to E Carrier Compositions (w/w %) Hydrophilic additives 1 (e.g. % Release in Corn Cremophor 2 RH 8% Triton glycerides 40, Tween 3 80, Aqueous Media (e.g. Glyceryl SLS, Poloxamer 4 Other additives 7 ⁇ 50% in 2 Comp. mono or 407, Polymer 5 , and/ Anti- Solidify Combi- Total % hrs and ⁇ 75% III No.
  • solubilizer Corn glycerides: e.g. Glyceryl mono or di-linoleate
  • hydrophilic additives e.g. % Release in Corn Cremophor 2 RH 8% Triton glycerides 40, Tween 3 80, Aqueous Media (e.
  • Carrier IV Compositions composed of solubilizer (Vegetable glycerides: e.g. Glyceryl mono or di-oleate), hydrophilic additives, and other additives for Composition A to E Carrier Compositions (w/w %) Hydrophilic additives 1 (e.g. % Release in Vegetable Cremophor 2 RH 8% Triton glycerides 40, Tween 3 80, Aqueous Media (e.g. Glyceryl SLS, Poloxamer 4 Other additives 7 ⁇ 50% in 2 Comp. mono or 407, Polymer 5 , and/ Anti- Solidify Combi- Total % hrs and ⁇ 75% IV No.
  • solubilizer e.g. Glyceryl mono or di-oleate
  • hydrophilic additives e.g. % Release in Vegetable Cremophor 2 RH 8% Triton glycerides 40, Tween 3 80
  • Aqueous Media e.g. Glyceryl
  • Carrier V Compositions composed of solubilizer (Glyceryl mono or di-stearate), hydrophilic additives, and other additives for Composition A to E Carrier Compositions (w/w %) Hydrophilic additives 1 (e.g. % Release in Cremophor 2 RH 8% Triton 40, Tween 3 80, Aqueous Media Glyceryl SLS, Poloxamer 4 Other additives 7 ⁇ 50% in 2 Comp. mono or 407, Polymer 5 , and/ Anti- Solidify Combi- Total % hrs and ⁇ 75% V No.
  • solubilizer Glyceryl mono or di-stearate
  • hydrophilic additives e.g. % Release in Cremophor 2 RH 8% Triton 40, Tween 3 80, Aqueous Media Glyceryl SLS, Poloxamer 4
  • Carrier VI Compositions composed of solubilizer (Glyceryl palmito-stearate), hydrophilic additives, and other additives for Composition A to E Carrier Compositions (w/w %) Hydrophilic additives 1 (e.g. % Release in Cremophor 2 RH 8% Triton 40, Tween 3 80, Aqueous Media Glyceryl SLS, Poloxamer 4 Other additives 7 ⁇ 50% in 2 Comp. palmito- 407, Polymer 5 , and/ Anti- Solidify Combi- Total % hrs and ⁇ 75% VI No.
  • solubilizer Glyceryl palmito-stearate
  • hydrophilic additives e.g. % Release in Cremophor 2 RH 8% Triton 40, Tween 3 80, Aqueous Media Glyceryl SLS, Poloxamer 4
  • Carrier VII Compositions composed of solubilizer ((9Z)-Octadec-9-enoic acid), hydrophilic additives, and other additives for Composition A to E Carrier Compositions (w/w %) Hydrophilic additives 1 (e.g. % Release in Cremophor 2 RH 8% Triton 40, Tween 3 80, Aqueous Media SLS, Poloxamer 4 Other additives 7 ⁇ 50% in 2 Comp. (9Z)-Octadec- 407, Polymer 5 , and/ Anti- Solidify Combi- Total % hrs and ⁇ 75% VII No.
  • solubilizer ((9Z)-Octadec-9-enoic acid)
  • hydrophilic additives e.g. % Release in Cremophor 2 RH 8% Triton 40, Tween 3 80, Aqueous Media SLS, Poloxamer 4
  • Carrier VIII Compositions composed of solubilizer (octadecanoic acid), hydrophilic additives, and other additives for Composition A to E Carrier Compositions (w/w %) Hydrophilic additives 1 (e.g. % Release in Cremophor 2 RH 8% Triton 40, Tween 3 80, Aqueous Media SLS, Poloxamer 4 Other additives 7 ⁇ 50% in 2 Comp. Octadec- 407, Polymer 5 , and/ Anti- Solidify Combi- Total % hrs and ⁇ 75% VIII No.
  • solubilizer octadecanoic acid
  • hydrophilic additives e.g. % Release in Cremophor 2 RH 8% Triton 40, Tween 3 80, Aqueous Media SLS, Poloxamer 4
  • Other additives 7 ⁇ 50% in 2 Comp. Octadec- 407, Polymer 5 , and/ Anti- Solidify Combi- Total % hrs and ⁇ 75%
  • Carrier IX Compositions composed of solubilizer ((9Z,12Z)-9,12-Octadecadienoic acid), hydrophilic additives, and other additives for Composition A to E Carrier Compositions (w/w %) Hydrophilic additives 1 (e.g. % Release in Cremophor 2 RH 8% Triton 40, Tween 3 80, Aqueous Media (9Z,12Z)-9,12- SLS, Poloxamer 4 Other additives 7 ⁇ 50% in 2 Comp. Octadeca- 407, Polymer 5 , and/ Anti- Solidify Combi- Total % hrs and ⁇ 75% IX No.
  • solubilizer ((9Z,12Z)-9,12-Octadecadienoic acid)
  • hydrophilic additives e.g. % Release in Cremophor 2 RH 8% Triton 40, Tween 3 80, Aqueous Media (9Z,12Z)-9,12- SLS, Poloxamer
  • Carrier X Compositions composed of solubilizer (Peppermint oil), hydrophilic additives, and other additives for Composition A to E Carrier Compositions (w/w %) Hydrophilic additives 1 (e.g. % Release in Cremophor 2 RH 8% Triton 40, Tween 3 80, Aqueous Media SLS, Poloxamer 4 Other additives 7 ⁇ 50% in 2 Comp. Pepper- 407, Polymer 5 , and/ Anti- Solidify Combi- Total % hrs and ⁇ 75% X No.
  • Carrier XI Compositions composed of solubilizer (Omega-3 EPA/DHA), hydrophilic additives, and other additives for Composition A to E Carrier Compositions (w/w %) Hydrophilic additives 1 (e.g. % Release in Cremophor 2 RH 8% Triton 40, Tween 3 80, Aqueous Media SLS, Poloxamer 4 Other additives 7 ⁇ 50% in 2 Comp. Omega-3 407, Polymer 5 , and/ Anti- Solidify Combi- Total % hrs and ⁇ 75% XI No.
  • Carrier XII Compositions composed of solubilizer (Vitamin E), hydrophilic additives, and other additives for Composition A to E Carrier Compositions (w/w %) Hydrophilic additives 1 (e.g. % Release in Cremophor 2 RH 8% Triton 40, Tween 3 80, Aqueous Media SLS, Poloxamer 4 Other additives 7 ⁇ 50% in 2 Comp. 407, Polymer 5 , and/ Anti- Solidify Combi- Total % hrs and ⁇ 75% XII No.
  • Vitamin E or combinations 6 oxidant 8 agent 9 nations 10 in Carrier in 4 hrs a 90-100 — 0-1 0-10 0-10 100 Yes b 45-99 1-40 0-1 0-15 0-15 100 Yes 1-25 0-1 15-20 15-20 100 Yes c 85-99 1-4.5 0-1 0-10 0-10 100 Yes d 80-95 5-10 0-1 0-10 0-10 100 Yes e 70-90 10-20 0-1 0-10 0-10 100 Yes f 60-80 20-30 0-1 0-10 0-10 100 Yes 10-20 0-1 10-20 10-20 100 Yes g 50-80 30-40 0-1 0-10 0-10 100 Yes 20-30 0-1 0-10 0-10 100 Yes 10-20 0-1 10-20 10-20 100 Yes g 50-80 30-40 0-1 0-10 0-10 100 Yes 20-30 0-1 0-10 0-10 100 Yes 10-20 0-1 10-20 10-20 100 Yes 0-1
  • Carrier XIII Compositions composed of a combination of solubilizers, hydrophilic additives, and other additives for Composition A to E Carrier Compositions (w/w %) Combination of solubilizers ⁇ (e.g. oleic Hydrophilic acid and GDS, additives 1 (e.g. % Release in oleic acid and Cremophor 2 RH 8% Triton peppermint 40, Tween 3 80, Aqueous Media oil, Magnoliaine SLS, Poloxamer 4 Other additives 7 ⁇ 50% in 2 Comp. 35-1 and 407, Polymer 5 , and/ Anti- Solidifying Combi- Total % hrs and ⁇ 75% XIII No.
  • solubilizers ⁇ e.g. oleic Hydrophilic acid and GDS
  • additives 1 e.g. % Release in oleic acid and Cremophor 2 RH 8% Triton peppermint 40, Tween 3 80, Aqueous Media oil,
  • the formulations provide 440 mg to 490 mg of testosterone undecanoate per day to an individual when administered twice-a-day as one or two unit dosage forms per administration (e.g., 2-4 unit dosage forms per day). In another preferred embodiment, the formulations provide about 450 mg or about 474 mg of testosterone undecanoate per day to an individual when administered twice-a-day as one or two unit dosage forms per administration (e.g., 2-4 unit dosage forms per day).
  • the weight percent refers to the weight percent of the capsule fill composition.
  • the monoglyceride can be glyceryl monolinoleate. In another aspect, the monoglyceride can be glyceryl mono-oleate.
  • the polyoxyl hydrogenated vegetable oil is a polyoxyl hydrogenated castor oil. In another aspect, polyoxyl hydrogenated vegetable oil is polyoxyl 40 hydrogenated castor oil. In another aspect, polyoxyl hydrogenated vegetable oil is polyoxyl 35 hydrogenated castor oil.
  • the additive can be chosen from adsorbing agents, anti-adherents, anticoagulants, antifoaming agents, anti-caking agents, anti-static agents, binders, bile acids, bufferants, bulking agents, chelating agents, coagulants, colorants, opaquants, coolants, cryoprotectants, diluents, dehumidifying agents, desiccants, desensitizers, disintegrants, dispersing agents, enzyme inhibitors, fillers, hydrating agent, super disintegrants, gums, mucilages, hydrogen bonding agents, enzymes, flavorants, humectants, humidifying agents, ion-exchange resins, lubricants, plasticizers, pH modifying agents, preservatives, organic solvents, spreading agent, stabilizers, suspending agent, thickeners, viscosity increasing agents, waxes, and mixtures thereof.
  • Monoglyceride/Diglyceride Hydrogenated Vegetable Oil 10 20-80% 0-20% Polyoxyl q.s.
  • Monoglyceride/Diglyceride Hydrogenated Vegetable Oil 11 20-80% 0-20% Polyoxyl q.s.
  • Monoglyceride/Diglyceride Hydrogenated Vegetable Oil 12 20-80% 0-20% Polyoxyl q.s.
  • Monoglyceride/Diglyceride Hydrogenated Vegetable Oil 13 20-80% 0-20% Polyoxyl q.s.
  • Monoglyceride/Diglyceride Hydrogenated Vegetable Oil 14 20-80% 0-20% Polyoxyl q.s.
  • the solubilizer can be glyceryl monolinoleate. In another aspect, the solubilizer can be glyceryl mono-oleate. In another aspect, the solubilizer can be glyceryl palmitostearate. In one aspect, the solubilizer is a mono-diglyceride of medium chain fatty acids (e.g., caprylic and capric)
  • the polyoxyl hydrogenated vegetable oil is a polyoxyl hydrogenated castor oil. In another aspect, the polyoxyl hydrogenated vegetable oil is polyoxyl 40 hydrogenated castor oil. In another aspect, the polyoxyl hydrogenated vegetable oil is polyoxyl 35 hydrogenated castor oil.
  • the additive can be chosen from adsorbing agents, anti-adherents, anticoagulants, antifoaming agents, anti-caking agents, anti-static agents, binders, bile acids, bufferants, bulking agents, chelating agents, coagulants, colorants, opaquants, coolants, cryoprotectants, diluents, dehumidifying agents, desiccants, desensitizers, disintegrants, dispersing agents, enzyme inhibitors, fillers, hydrating agent, super disintegrants, gums, mucilages, hydrogen bonding agents, enzymes, flavorants, humectants, humidifying agents, ion-exchange resins, lubricants, plasticizers, pH modifying agents, preservatives, organic solvents, spreading agent, stabilizers, suspending agent, thickeners, viscosity increasing agents, waxes, and mixtures thereof.
  • the solubilizer can be glyceryl monolinoleate. In another aspect, the solubilizer can be glyceryl mono-oleate. In one aspect, the solubilizer is a tocopherol. In another aspect, the solubilizer can be glyceryl palmitostearate. In one aspect, the solubilizer is chosen from any one or more of those described herein. In one aspect, the dispersant is chosen from any one of those described herein. In one aspect, the dispersant is a polyoxyl hydrogenated vegetable oil. In one aspect, the polyoxyl hydrogenated vegetable oil is a polyoxyl hydrogenated castor oil.
  • the polyoxyl hydrogenated vegetable oil is polyoxyl 40 hydrogenated castor oil. In another aspect, the polyoxyl hydrogenated vegetable oil is polyoxyl 35 hydrogenated castor oil.
  • the additive can be chosen from adsorbing agents, anti-adherents, anticoagulants, antifoaming agents, anti-caking agents, anti-static agents, binders, bile acids, bufferants, bulking agents, chelating agents, coagulants, colorants, opaquants, coolants, cryoprotectants, diluents, dehumidifying agents, desiccants, desensitizers, disintegrants, dispersing agents, enzyme inhibitors, fillers, hydrating agent, super disintegrants, gums, mucilages, hydrogen bonding agents, enzymes, flavorants, humectants, humidifying agents, ion-exchange resins, lubricants, plasticizers, pH modifying agents, preservatives, organic solvents, spreading agent, stabilizer
  • the solubilizer can be glyceryl monolinoleate. In another aspect, the solubilizer can be glyceryl mono-oleate. In another aspect, the solubilizer can be glyceryl palmitostearate.
  • the lipophilic surfactant is chosen from any one or more of those described therein.
  • the hydrophilic surfactant is an ionic hydrophilic surfactant. In one aspect, the hydrophilic surfactant is a non-ionic hydrophilic surfactant.
  • the hydrophilic surfactant is a polyoxyl hydrogenated vegetable oil. In one aspect, the polyoxyl hydrogenated vegetable oil is a polyoxyl hydrogenated castor oil.
  • polyoxyl hydrogenated vegetable oil is polyoxyl 40 hydrogenated castor oil.
  • polyoxyl hydrogenated vegetable oil is polyoxyl 35 hydrogenated castor oil.
  • hydrophilic surfactant is any one of those described herein.
  • the additive can be chosen from adsorbing agents, anti-adherents, anticoagulants, antifoaming agents, anti-caking agents, anti-static agents, binders, bile acids, bufferants, bulking agents, chelating agents, coagulants, colorants, opaquants, coolants, cryoprotectants, diluents, dehumidifying agents, desiccants, desensitizers, disintegrants, dispersing agents, enzyme inhibitors, fillers, hydrating agent, super disintegrants, gums, mucilages, hydrogen bonding agents, enzymes, flavorants, humectants, humidifying agents, ion-exchange resins, lubricants, plasticizers, pH modifying agents, preservatives, organic solvents, spreading agent, stabilizers, suspending agent, thickeners, viscosity increasing agents, waxes, and mixtures thereof.
  • compositions in the Tables above releases (a) at least 80% or more at 1 or 2 hours; (b) less than 95% or 90% at 0.25 hours; (c) about 100% at 4, 3, 2, 1, 0.5, or 0.25 hours or (d) a combination of one, two, or three of (a)-(c) when tested using USP type 2 apparatus in about e.g., 1000 mL 8% Triton X100 solution in water at a specific temperature (e.g., 37° C.) at 100 rpm.
  • the composition of this embodiment releases (a) at least 75% or more at 2 hours and (b) less than 95% or 90% at 0.25 hours.
  • compositions in the Tables above have carrier components chosen so as to have a release profile as described in this paragraph.
  • the active pharmaceutical ingredient is fully solubilized in the composition. In another aspect, the active pharmaceutical ingredient is not fully solubilized as described herein.
  • a testosterone undecanoate containing composition was prepared by using the components set forth in Table I.
  • the composition is prepared by weighing all of the components, except the testosterone undecanoate, into a clean stainless steel container and mixed together at about 50° C. to about 70° C., using a stirrer.
  • the testosterone undecanoate (TU) is added and stirred into the mixture of other components until the testosterone undecanoate dissolves.
  • a predetermined quantity of this fill material is disposed into a capsule (for example, hard gelatin capsule) to get the required testosterone undecanoate dose per dosage unit.
  • the capsules are allowed to cool at room temperature, banded (if required) and packaged in a HDPE bottle and tightly closed with an appropriate lid.
  • a testosterone undecanoate containing composition was prepared similarly to the method described in Example 1 using the components set forth in Table II.
  • Example 2 Composition mg/capsule Testosterone Undecanoate 225 Solubilizer (e.g. Maize oil glyceride) 260 Dispersant (e.g. lauroglycol) 665 TU-loading (wt %) of capsule fill ⁇ 19.6%
  • Solubilizer e.g. Maize oil glyceride
  • Dispersant e.g. lauroglycol
  • TU-loading wt %
  • Testosterone undecanoate containing composition were prepared similarly to the method described in Example 1 using the components set forth in Tables III and IV
  • a testosterone undecanoate containing composition was prepared similarly to the method described in Example 1 using the components set forth in Table V
  • a testosterone undecanoate containing composition was prepared by using the components set forth in Table VI and a method similar to that described in Example 1.
  • Solubilizer e.g. Maize oil 490 glycerides
  • Dispersant e.g. polysorbate 80
  • Solidifying agent e.g. polyethylene 45 glycol, 8000 or PEG 8000
  • TU-Loading per capsule 26.3%
  • a testosterone undecanoate containing composition was prepared by using the components set forth in Table VII and a method similar to that described in Example 1.
  • a testosterone undecanoate containing composition was prepared by using the components set forth in Table VIII and a method similar to that described in Example 1.
  • a testosterone undecanoate containing composition was prepared using the components set forth in Table IX and a method similar to that described in Example 1.
  • Solubilizer e.g. Maize oil glycerides
  • Solubilizer e.g. ⁇ -tocopherol
  • Solidifying agent - e.g. Glyceryl distearate; 25 Percirol ® ATO 5
  • TU- loading (wt %) of capsule fill 34.8%
  • a testosterone undecanoate containing composition can be prepared by using the components set forth in Table X by a method as follows: The required quantity of the glyceryl distearate or glyceryl monostearate and the PEG 8000 are placed in a stainless steel container and heated to about 50 to 70° C. to get a molten mixture. The testosterone undecanoate is added and stirred till it completely dissolves. A predetermined weight of the molten mixture is disposed into capsules and allowed to congeal at room temperature, banded and packed.
  • the oral dosage capsules of Example 10, which contains non-dissolved TU, can provide, upon single administration along with food to a human male, a testosterone undecanoate AUC that is about 20% higher as compared to a composition that does not include the glyceryl distearate (Percirol® ATO 5) or glyceryl monostearate.
  • a testosterone undecanoate AUC that is about 20% higher as compared to a composition that does not include the glyceryl distearate (Percirol® ATO 5) or glyceryl monostearate.
  • Example 10 can also be optionally modified so that a dispersant such as a disintegrating agent (e.g. Crospovidone at about 150 mg for every 100 mg TU dose) can be uniformly suspended under stirring in the molten testosterone undecanoate solution.
  • a dispersant such as a disintegrating agent (e.g. Crospovidone at about 150 mg for every 100 mg TU dose) can be uniformly suspended under stirring in the molten testosterone undecanoate solution.
  • This suspension can be further allowed to cooled and passed through ASTM 30 mesh get granulates or particulates which can be either filled in a capsule or compressed to a tablet.
  • a testosterone undecanoate containing composition wherein at least 50% of the testosterone undecanoate is dissolved is prepared by using the components set forth in Table XI and a method similar to that described under Example 1.
  • Testosterone undecanoate formulations of Examples 12 through 19 were prepared by using the components set forth in Table XII and by a method similar to that described under Example 1. Additionally, indicated amounts of the respective formulations were filled into hard gelatin capsules and the testosterone undecanoate release from capsules is measured using a USP Type-II apparatus at about 100 rpm in about 1000 mL of 8% w/w solution of Triton X100 in water, maintained at about 37° C. The results of the release testing are also shown in Table XII.
  • Testosterone undecanoate formulations of Examples 20 through 25 were prepared by using the components set forth in Table XIII and by a method similar to that described under Example 1. Additionally, indicated amounts of the respective formulations were filled into hard gelatin capsules and the testosterone undecanoate release from capsules was tested in about 1000 mL of 8% w/w solution of Triton X100 in water, maintained at about 37° C., using a USP Type-II apparatus at about 100 rpm. The results of the release testing are also shown in Table XIII.
  • Testosterone undecanoate formulations of Examples 26 through 29 were prepared by using the components set forth in Table XIV and a method similar to that described under Example 1. Additionally, indicated amounts of the respective formulations were filled into hard gelatin capsules and the testosterone undecanoate released from capsules was tested in about 1000 mL of 8% w/w solution of Triton X100 in water, maintained at about 37° C., using a USP Type-II apparatus at about 100 rpm. The results of the release testing are also shown in Table XIV.
  • Testosterone undecanoate formulations Examples 30 through 35 can be prepared by using the components set forth in Table XV and by a method similar to that described in Example 1. Additionally, indicated amounts of the respective formulations can be encapsulated in gelatin capsules and the testosterone undecanoate release from the capsules tested in about 1000 mL of 8% w/w solution of Triton X100 in water, maintained at about 37° C., using a USP Type-II apparatus at about 100 rpm. The results of the release testing are also shown in Table XV.
  • Examples 30 through 35 demonstrate the importance of the choice of the solubilizers of the current invention and their levels to achieve greater testosterone undecanoate loading and yet maintain the solubilization of the testosterone undecanoate in the composition and/or the dosage form.
  • compositions of the current invention can be further adsorbed onto one or more substrate materials such as, for example, lactose, magnesium aluminosilicate, colloidal silicon dioxide, starch, microcrystalline cellulose, alkyl celluloses etc., whereby a free flowing powder/granule form is obtained which can be used as a granules, or disposed into capsule, or pressed into tablet.
  • substrate materials such as, for example, lactose, magnesium aluminosilicate, colloidal silicon dioxide, starch, microcrystalline cellulose, alkyl celluloses etc.
  • the amount of the substrate material can be from about 15% to about 40% of the weight of the composition. In one embodiment, the amount of the substrate material can be from about 20% to about 35% of the weight of the formed granule or powder.
  • the method of making such adsorbed testosterone undecanoate compositions can include pouring the liquid compositions on the substrate material under and continuous mixing at room temperature or at about 50° C.-70° C., depending on the composition. After cooling, the adsorbed composition can be disposed into capsule or pressed into tablet.
  • Table XVI illustrates examples of the freely flowable adsorbed solubilized testosterone undecanoate compositions.
  • Example 17 and Example 21 A preliminary stability evaluation with respect to the change in potency and/or appearance of the potential primary degradation products was carried out with the compositions of Example 17 and Example 21, both filled in hard gelatin capsules at 200 mg/per capsule and 75 mg per capsules.
  • the capsules were packed in HDPE bottles and staged for stability studies in the environmental chambers maintained at 25° C./60% RH.
  • the primary degradation products were determined by a HPLC analysis method after about three months' storage and the results shown in Table XVII.
  • Testosterone undecanoate-containing compositions were prepared by using the components set forth in Tables XVIII and XIX. It should be noted that Example 40 and 47 are expressed as wt % of Examples 17 and 12 respectively.
  • the compositions are prepared by weighing all of the components, except the testosterone undecanoate, into a clean stainless steel container and mixed together at about 50° C. to about 70° C., using a stirrer.
  • the testosterone undecanoate (TU) is added and stirred into the mixture of other components until the testosterone undecanoate dissolves.
  • a predetermined quantity of the “capsule fill” was disposed into a capsule (for example, hard gelatin capsule) to get the required testosterone undecanoate dose per dosage unit.
  • the capsules are allowed to cool at room temperature, banded (if required) and packaged in a HDPE bottle and tightly closed with an appropriate lid.
  • compositions can also include one or more components such as anti-oxidants (e.g. BHA, BHT, Ascorbyl palmitate, vitamin E or combinations), flavorings etc.
  • anti-oxidants e.g. BHA, BHT, Ascorbyl palmitate, vitamin E or combinations
  • compositions can also include one or more components such as anti-oxidants (e.g. BHA, BHT, Ascorbyl palmitate, vitamin E or combinations), flavorings etc.
  • anti-oxidants e.g. BHA, BHT, Ascorbyl palmitate, vitamin E or combinations
  • compositions of tables XVIII and XIX include descriptions of the amounts of testosterone undecanoate that is dissolved at room temperature (RT).
  • the percent (%) of the undissolved testosterone undecanoate in the composition at RT can range from about 5 wt % to about 60 wt %.
  • Examples 44 to 47 do not include a solidifying agent and that about 100 wt % of the testosterone undecanoate is dissolved in these composition. All Examples, except Examples 44 to 47, are free of ionizable fatty acids such as oleic acid.
  • the clinical testing for the select inventive composition examples were conducted in a randomized study in human subjects including hypogonadal males having a pre-test screening serum testosterone concentrations less than 3 ng/mL ( ⁇ 10.5 nmol/L).
  • the study compositions were administered orally with at least 240 mL of water about 30 minutes after starting a standardized meal (at least 30-35% fat) which was preceded by a fast of about 10 hours.
  • the testing methodology allows for determining single dose pharmacokinetic profiles. Dose amounts as well as mean pharmacokinetic parameters from the testing are shown in the Table XX.
  • Examples 40, 41, and 45 provide a higher dose normalized C max and a lower C max to C ave ratio after single dose administration (one half of the daily dose) as compared to Example 47, which is a lower loading, fully dissolved composition.
  • Examples 40, 41, 44, and 45 were clinically tested in a randomized study in human male subjects, including hypogonadal males having pre-test serum testosterone concentrations less than 3 ng/mL ( ⁇ 10.5 nmol/L).
  • the example compositions were administered orally with at least 240 mL of water about 30 minutes after starting a standardized meal (at least 30-35% fat) which was preceded by a fast of about 10 hours.
  • the testing methodology allows for determining single dose PK profiles. Dose amounts as well as mean pharmacokinetic parameters from the testing are shown in Table XXI.
  • Example 40 and 41 provide a desirable higher dose normalized C ave (bioavailability) after single dose administration (one half of the daily dose) as compared to Examples 44 and 45. It is noteworthy that the testosterone undecanoate in Examples 44 and 45 is fully dissolved in the composition at room temperature. Further, each of Examples 44 and 45 contain oleic acid, an ionizable fatty acid, and each is devoid of solids (i.e. a solidifying agent and/or undissolved testosterone undecanoate).
  • PK performance parameters related to target pharmacokinetic criteria in a group of hypogonadal men with no dose titration i.e. the maintenance dose is the same as the initial TU daily dose
  • the data shown is based on normal patient distribution and a variability of about 50%, although similar results are expected if the variability changes up to an additional 15%.
  • the simulated PK performance parameters allow for the C ave and C max to be measured on different days after day 84.
  • Table XXII shows the estimated parameters for Examples 40, 41 and 45, including various daily dose quantities for the compositions and capsules Example 40.
  • Example 40-B and Example 41 provide PK performance criteria which may not require individualized titrations of the daily dose.
  • PK performance parameters were estimated related to target PK criteria in a group of hypogonadal men using a dose titration metric of C ave with a single dose titration.
  • the estimated data is based on normal patient distribution and a variability of about 50%. Similar results are expected if the variability in PK parameters and/or maintenance dose changes by up to 15% or the selection of another relevant dose-sensitive metric, such as C t , C max , C min , or the like.
  • the simulated PK performance parameters allow for the C ave and C max to be measured on different days after day 90.
  • Table XXIII shows the estimated parameters for Examples 40 and 45, including various daily dose quantities for the compositions and capsules of Example 40.
  • Example 40A 40E 40D Example 45 Total TU initial Daily Dose 150 360-460 1000 580-650 (IDD), mg Maintenance Daily Dose (% ⁇ 40 ⁇ 40 ⁇ 40 ⁇ 40 of IDD) PK Performance Criteria Group PK Performance Results C avg in the range of 300-1100 ⁇ 75% >75% ⁇ 75% ⁇ 75% ng/dL C max is less than 1500 ng/dL >85% >85% ⁇ 85% ⁇ 85% C max in the range between ⁇ 5% ⁇ 5% >5% >5% 1800 and 2500 ng/dL C max greater than 2500 ng/dL ⁇ 1% ⁇ 1% >1% >1% >1% >1%
  • pharmacokinetic performance parameters were estimated related to target PK criteria in a group of hypogonadal men using a dose titration metric of C ave and two dose titrations.
  • the estimated data is based on normal patient distribution and a variability of about 50%. Similar results are expected if the variability in PK parameters and/or maintenance dose changes by up to 15% or the selection of another relevant dose-sensitive metric, such as C t , C max , C min , or the like.
  • the simulated PK performance parameters allow for the C ave and C max to be measured on different days after day 90.
  • Table XXIV shows the estimated parameters for Examples 40 and 45, including various daily dose quantities for the compositions and capsules Example 40.
  • Example 40A 40E 40C Example 45 Total TU Initial Daily (IDD) 150 360-460 1000 580-650 Maintenance Daily Dose (% ⁇ 40 ⁇ 40 ⁇ 40 ⁇ 40 of IDD) PK Performance Criteria Group PK Performance Results C avg in the range of 300-1100 ⁇ 75% >75% ⁇ 75% >75% ng/dL C max is less than 1500 ng/dL >85% >85% ⁇ 85% >85% C max in the range between ⁇ 5% ⁇ 5% >5% ⁇ 5% 1800 and 2500 ng/dL C max greater than 2500 ng/dL ⁇ 1% ⁇ 1% >1% >1% >1% >1%
  • pharmacokinetic performance parameters were estimated related to target pharmacokinetic criteria in a group of hypogonadal men using a dose titration metric of C ave and three dose titrations.
  • the estimated data is based on normal patient distribution and a variability of about 50%. Similar results are expected if the variability in PK parameters and/or maintenance dose changes by up to 15% or the selection of another relevant dose-sensitive metric, such as C t , C max , C min , or the like.
  • the simulated PK performance parameters allow for the C ave and C max to be measured on different days after day 90.
  • Table XXV shows the estimated parameters for Examples 40, 41, 44 and 45, including various daily dose quantities for the compositions and capsules Example 40.
  • C t is the serum total testosterone concentration upon single testosterone administration, after “t” hours post-dose of a typical single daily dose of a BID daily dose regimen on the titration node day.
  • Table XXVI shows a titration table based on the titration metric C t for various time points following administration.
  • the “t” can be within about 1 hour.
  • Dosing regimens were prepared based on simulated data from observed steady state PK data in hypogonadal men assuming a normal patient distribution and with variability of about 50% and a dose titration metric of C ave . Similar results can be expected if the variability in PK parameters changes by up to 15% using titration metric C t relationship given in Table-XXVI.
  • Safety assessment can be done any day the C max is measured for a testosterone product (typically, 84 days from initial dosing).
  • efficacy assessments can be done any day the C ave is measured for a testosterone product (typically, 84 days from initial dosing).
  • the regimens are prepared based on the understanding that the C ave and C max can be measured on different days.
  • the dosing regimen methodology disclosed herein for subjects in need of dose adjustments includes appropriate initial daily dose, maintenance dose and titration metric result in effective testosterone therapy (C ave >300 ng/dL) for optimal efficacy, while maintaining the C max ⁇ 1500 ng/dL in order to provide a better safety profile.
  • dosing method E for a compositions of this invention would require more than three titrations which would generally be inconvenient, impractical, and may not allow for high levels of patient compliance.

Abstract

The present disclosure is drawn to pharmaceutical compositions and oral dosage capsules containing testosterone undecanoate, as well as related methods. The capsule includes a capsule shell and a capsule fill. The capsule fill can include a solubilizer and about 14 wt % to about 35 wt % testosterone undecanoate based on the total capsule fill. The oral dosage capsule is such that when a single oral administration to a male subject of one or more capsules with a total testosterone undecanoate daily dose of about 350 mg to about 650 mg it provides a ratio of serum testosterone Cmax to serum testosterone Cave of about 2.7 or less. In yet another embodiment, a method for providing a serum concentration of testosterone within a target serum testosterone concentration Cave range for a male subject is provided.

Description

    PRIORITY DATA
  • This application is a continuation-in-part of U.S. patent application Ser. No. 15/270,357, filed Sep. 20, 2016 which is a continuation U.S. Ser. No. 14/952,796, filed Nov. 25, 2015 (now U.S. Pat. No. 9,480,690, issued Nov. 1, 2016) which is a continuation of U.S. Ser. No. 14/801,674, filed Jul. 16, 2015, which is a continuation of Ser. No. 14/691,229, filed Apr. 20, 2015 (now U.S. Pat. No. 9,205,057, issued Dec. 8, 2015) which is a continuation of U.S. Ser. No. 13/485,807 filed May 31, 2012 (now U.S. Pat. No. 9,034,858, issued May 19, 2015) which is a continuation-in-part of U.S. Ser. No. 12/957,206, filed on Nov. 30, 2010 and of Patent Cooperation Treaty Application Serial No. PCT/US2011/62538, filed on Nov. 30, 2011, each of which is incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to testosterone undecanoate containing pharmaceutical compositions and oral dosage capsules as well as associated methods. Accordingly, this invention involves the fields of chemistry, pharmaceutical sciences, medicine and other health sciences.
  • BACKGROUND OF THE INVENTION
  • Male hypogonadism is a serious condition affecting mostly aging men. The common reasons for hypogonadism in men could be physiological abnormality involving among other factors, improper functioning or growth of the gonads and/or the pituitary-hypothalamus regulatory systems, and aging. Many of the abnormalities that are identified to be commonly associated with the low or decreased testosterone levels include impaired sexual function and/or libido, metabolic syndrome which may be a combination of abdominal obesity, high blood pressure, insulin resistance, lipid disorders; high risk of cardiovascular diseases; reduced bone mass/mineral density and muscle weakness and or degeneration affecting the musculoskeletal system. Other effects of low testosterone levels include negative changes in body composition, depression and other psychological disorders. The average human male produces 4-7 mg of testosterone per day in a circadian pattern, with maximal plasma levels attained in early morning and minimal levels in the evening. It is generally recognized that in a normal adult man of age 17 to 54 years, the serum total testosterone (T) is between about 300 ng/dL to about 1100 ng/dL and this range is referred to as the eugonadal range. Restoration of testosterone levels to the eugonadal range typically corrects many of the cited clinical abnormalities associated with hypogonadism or low testosterone levels.
  • While oral administration is the most preferred and patient friendly route for administration, the effective oral delivery of testosterone as testosterone and its esters remains a challenge. This is due to extremely poor bioavailability of testosterone, which requires very high dosing as well as frequent dosing due to the short serum half-life. These problems with orally administered testosterone products are primarily due to first pass metabolism. Further, direct oral delivery of testosterone has also been known to cause enzyme induction resulting in potential drug-drug interactions.
  • Currently, modified testosterones, in form of a methyl analogue of testosterone, and as an undecanoate ester, testosterone undecanoate (TU) are available for oral administration for patients in need of testosterone therapy. However, liver damage including cholestasis, peliosis hepatitis, nodular regenerative hyperplasia, and primary hepatic tumors has been reported with use of methyl testosterone. Testosterone undecanoate is a prodrug which gets converted to testosterone in vivo. Testosterone undecanoate containing products are available in some countries as liquid filled soft-gelatin capsule containing 40 mg of fully solubilized testosterone undecanoate. Testosterone undecanoate is extremely lipophilic (calculated log P of ˜6.5) with a water solubility of <0.3 ng/ml and a melting point around 62° C. It is generally believed that in order to promote lymphatic absorption for better safety profile and to facilitate effective oral delivery of testosterone undecanoate, the testosterone undecanoate must be presented in a bioacceptable solubilizer. Accordingly, research continues into the development of testosterone oral delivery products that can have high drug load and provide for practical unit oral dosage forms.
  • SUMMARY OF THE INVENTION
  • The present disclosure is drawn to pharmaceutical compositions and oral dosage capsules containing testosterone undecanoate, as well as related methods. In one embodiment, a pharmaceutical capsule for oral delivery is provided. The capsule includes a capsule shell and a capsule fill. The capsule fill can include a solubilizer and about 14 wt % to about 35 wt % testosterone undecanoate based on the total weight of the capsule fill. In some aspects, the oral dosage capsule is such that upon a single oral administration to a male subject of one or more capsules with a total testosterone undecanoate daily dose of about 350 mg to about 650 mg it provides a ratio of serum testosterone Cmax to serum testosterone Cave of about 2.7 or less.
  • In another embodiment, a method for providing a serum concentration of testosterone within a target serum testosterone concentration Cave range for a male subject is provided. The method can include the step of orally administering to the male subject a daily dose of a testosterone undecanoate-containing composition. The testosterone undecanoate can comprise about 14 wt % to about 35 wt % of the testosterone undecanoate-containing composition and the daily dose provided can be about 350 mg to about 420 mg of testosterone undecanoate to the male subject.
  • In yet a further embodiment, a method for providing a serum concentration of testosterone within a target serum testosterone concentration Cave range for a male subject is provided. The method can include the step of orally administering to the male subject an initial regimen including a daily dose of a testosterone undecanoate-containing composition. The testosterone undecanoate can comprise about 14 wt % to about 35 wt % of the testosterone undecanoate-containing composition and the daily dose provided can be about 350 mg to about 650 mg of testosterone undecanoate to the male subject. After the initial regimen, the method can include a step of determining the serum testosterone concentration for the male subject on at least one titration node day within the initial regimen. The method further can include the step of orally administering to the male subject a maintenance regimen including a daily dose of testosterone undecanoate-containing composition that comprises about 14 wt % to about 35 wt % of the testosterone undecanoate. The maintenance regimen can provide a daily dose of testosterone undecanoate to the subject based on the serum testosterone concentration determined by a titration metric on the at least one titration node day of the initial regimen and the said maintenance daily dose can be sufficient to provide a serum testosterone plasma concentration within the target range. The method can provide a steady state ratio of serum testosterone Cmax to Cave of 2.7 or less.
  • The applicants of the present invention have found that capsule fill compositions with a lower TU loading of less than 14% w/w are unsuitable for optimal activity due to inadequate bioavailability which also leads to larger dosage forms/doses for therapy in treatment of symptoms related to male hypogonadism. Consistent with the compositions of the present invention, higher loading dosage forms, i.e. those with ≥14% w/w of TU based on total capsule fill, resulted in a superior Cave per mg testosterone undecanoate administered, thus providing improved performance of oral testosterone undecanoate as a testosterone therapy.
  • Further, it was also found that it is not necessary to fully dissolve the testosterone undecanoate in the capsule fill, but rather only about at least 80 wt % of the testosterone undecanoate may need to be dissolved with at least 5% of testosterone undecanoate possibly needing to remain undissolved in order to achieve a desired and efficient Cave per mg of testosterone undecanoate administered. Therefore, in one embodiment in order to facilitate improved activity, an oral capsule with TU loading in the range of about 14 wt % to about 18% is provided in which no more than 80 wt % of the TU in the capsule fill is dissolved. This capsule can result in dose normalized Cmax suggesting superior bioavailability yet manageable Cmax for effective therapeutics.
  • DETAILED DESCRIPTION
  • Before the present testosterone undecanoate compositions, oral dosage capsules and related methods of use are disclosed and described, it is to be understood that this invention is not limited to the particular process steps and materials disclosed herein, but is extended to equivalents thereof, as would be recognized by those ordinarily skilled in the relevant arts. It should also be understood that terminology employed herein is used for the purpose of describing particular embodiments only and is not intended to be limiting.
  • It should be noted that, the singular forms “a,” “an,” and, “the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to “an excipient” includes reference to one or more of such excipients, and reference to “the carrier” includes reference to one or more of such carriers.
  • Definitions
  • As used herein, the term “treatment,” when used in conjunction with the administration of pharmaceutical compositions and oral dosage capsules containing testosterone undecanoate, refers to the administration of the oral dosage capsules and pharmaceutically acceptable composition to subjects who are either asymptomatic or symptomatic. In other words, “treatment” can both be to reduce or eliminate symptoms associated with a condition present in a subject, or it can be prophylactic treatment, i.e. to prevent the occurrence of the symptoms in a subject. Such prophylactic treatment can also be referred to as prevention of the condition.
  • As used herein, the terms “formulation” and “composition” are used interchangeably and refer to a mixture of two or more compounds, elements, or molecules. In some aspects the terms “formulation” and “composition” may be used to refer to a mixture of one or more active agents with a carrier or other excipients. Furthermore, the term “dosage form” can include one or more formulation(s) or composition(s) provided in a format for administration to a subject. When any of the above terms is modified by the term “oral” such terms refer to compositions, formulations, or dosage forms formulated and intended for oral administration to subjects.
  • As used herein, the term “fatty acid” refers to unionized carboxylic acids with a long aliphatic tail (chain), either saturated or unsaturated, conjugated or non-conjugated.
  • Unless otherwise specified, the term C8 to C22 fatty acid glycerides refers to a mixture of mono-, di-, and/or tri-glycerol esters of medium to long chain (C8 to C22) fatty acids.
  • As used herein, the term “dispersant” refers to any pharmaceutically acceptable additive that enables the contents of the compositions and/or oral dosage capsules to disperse in an aqueous medium. The extent of dispersion in an aqueous medium can be determined spectrophotometrically from the absorbance exhibited by the dispersion at a wavelength of about 400 nm. For example, the dispersion of the composition (with or without the testosterone undecanoate) of the current invention in about 0.2 mM sodium lauryl sulphate solution in water, has an absorbance of about 0.6 or less at about 400 nm wavelength, when the ratio of the composition to the sodium lauryl sulphate solution is about 1:2000. In a specific embodiment, the dispersion of the composition (with or without the testosterone undecanoate) of the current invention in about 0.2 mM sodium lauryl sulphate solution in water, has an absorbance of about 0.3 or less at about 400 nm wavelength, when the ratio of the composition to the sodium lauryl sulphate solution is about 1:5000. In another embodiment, the composition can produce a fine dispersion upon dilution in an aqueous medium without the need of a hydrophilic surfactant.
  • Further, as used herein, the dispersant of the current invention is at least one selected from the group of hydrophilic surfactant or lipophilic surfactant. In one embodiment, the dispersant includes a hydrophilic surfactant.
  • As used herein, the term “solidifying agent” or “solidifying additive” are used interchangeably and refer to a pharmaceutically acceptable additive that is in a solid physical state at 20° C. Similarly, a “solid lipophilic additive” refers to a lipophilic compound or component that is in a solid physical state at 20° C. and/or renders the composition or dosage form non-liquid, such as solid or semi-solid.
  • As used herein, the terms “solubilized” and “solubility,” when used to describe the state of testosterone undecanoate with respect to a composition and/or capsule fill, refer to the absence of testosterone undecanoate crystals in the composition or oral dosage form when observed under hot-stage microscope over a temperature of about 25° C. to about 65° C., or the absence of crystalline testosterone undecanoate melting related peak (about 62 to about 65° C.) when the composition or oral dosage form is subjected to differential scanning calorimetry. Similarly, the solubility of testosterone undecanoate in a particular compound, e.g. a solubilizer, is the amount of testosterone undecanoate dissolved to form a visibly clear solution at a specified temperature such as about 25° C. or about 37° C. With this definition in mind, compositions having crystalline forms of testosterone undecanoate at about room temperature would be considered to have an unsolubilized fraction and a solubilized fraction of testosterone undecanoate which fraction includes testosterone undecanoate in a solid state that is not crystalline such as amorphous and solid solution which are solubilized but undissolved.
  • As used herein, “undissolved” or “non-dissolved” can be used interchangeably and when one is used to describe the state of testosterone undecanoate with respect to a composition and/or capsule fill refers to the testosterone undecanoate in a non-liquid testosterone undecanoate-containing composition that is solubilized (such as non-crystalline) and non-solubilized such as crystalline TU. The solubility of TU in the composition can be estimated based on the individual solubility in the composition components. For solubilizers that are viscous or non-liquid at room temperature, TU solubility in the composition at room temperature (RT) may be estimated based on the observed values at higher temperature such as at 37° C.
  • As used herein, “dissolved” when used to describe the state of testosterone undecanoate (TU) with respect to a composition or capsule fill refers to a testosterone undecanoate-containing liquid solution having no undissolved testosterone undecanoate.
  • As used herein, the term “lipophilic,” refers to compounds that are not freely soluble in water; and the term “lipophilic surfactant” refers to surfactants that have HLB values of about 10 or less. Conversely, the term “hydrophilic” refers to compounds that are soluble in water; and term “hydrophilic surfactant” refers to surfactants that have HLB values of more than about 10.
  • As used herein, the term “ionizable fatty acid” refers to a fatty acid compound that changes its HLB as a function of pH. For example oleic acid is predominantly lipophilic at lower pH values (such as those found in the stomach) but becomes a predominantly hydrophilic at higher pH values (such as those found in the intestine).
  • As used herein, “subject” refers to a mammal that may benefit from the administration of a drug composition or method of this invention. Examples of subjects include humans. In one aspect, the subject can be a human male. In another embodiment, the subject can be a hypogonadal male. As used herein, the testosterone deficiency or hypogonadism in a male human subject (hypogonadal male) refers to a condition wherein the average baseline plasma testosterone concentration (T-Cavg-B) is about 300 ng/dL or less. However in some instances, testosterone deficiency or hypogonadism in a male human subject refers to a condition wherein the average baseline plasma testosterone concentration is about 400 ng/dL or less.
  • A used herein, a “responder” is a subject who responds to exogenous oral testosterone undecanoate therapy. “Responder analysis” is the assessment of the effectiveness of testosterone undecanoate therapy in a group of subjects deemed to get benefits of oral TU therapy.
  • As used herein, “group” or “group of subjects” refers to a collection of at least 24 human male subjects who receive and respond to exogenous oral administration of the compositions disclosed herein, namely testosterone undecanoate-containing compositions. In one aspect, the group can include at least 100 or at least 300 male subjects. In another aspect, the group can include at least 1000 male subjects. In another embodiment, the subjects can be hypogonadal subjects.
  • The term “oral administration” represents any method of administration in which an active agent can be administered by swallowing, chewing, or sucking of the dosage form. The composition of the current inventions can be admixed with food or drink prior to being orally consumed.
  • As used herein, a “dosing regimen” or “regimen” such as an “initial dosing regimen” or a “maintenance dosing regimen” refers to how, when, how much, and for how long a dose of the compositions of the present invention can be administered to a subject. For example, an initial dosing regimen for a hypogonadal male subject may provide for a total daily dose of 600 mg administered in two divided doses at least 12 hours apart (e.g. once with breakfast and once with dinner) with meals having about 25-55 g of fat content repeated daily for 30 days.
  • As used herein, “daily dose” refers to the amount of active agent (e.g. testosterone undecanoate) administered to a subject over a 24 hour period of time. The daily dose can be administered two or more administrations during the 24 hour period. In one embodiment, the daily dose provides for two administrations in a 24 hour period. With this in mind, an “initial dose” or initial daily dose” refers to a dose administered during the initial regimen or period of a dosing regimen. An initial dose includes both the very first dose during the initial regimen as well as the subsequent doses during the same initial regimen. Similarly, a “maintenance dose” or “maintenance daily dose” refers to a dose administered during a maintenance regimen of a dosing regimen. It is worth noting that the maintenance dose follows a dose titration based on the serum testosterone determination on a titration node day, however the maintenance dose does not need to be of a different quantity as the initial dose or the previous maintenance dose (in the case of multiple titrations).
  • As used herein, “non-liquid” when used to refer to the state of a composition disclosed herein refers to the physical state of the composition as being a semi-solid or solid.
  • As used herein, “solid” and “semi-solid” refers to the physical state of a composition that supports its own weight at standard temperature and pressure, and has adequate viscosity or structure to not freely flow. Semi-solid materials may conform to the shape of a container under applied pressure.
  • As used herein, “titration” or “dose titration” or “dose adjustment” are used interchangeably and refer to an increase or decrease of the total daily dose of testosterone undecanoate administered to a subject, typically based on the response of the subject to the exogenous administered testosterone undecanoate. The dose can be increased or decreased based on the measurement of serum testosterone concentration after a steady state has been achieved.
  • As used herein, “steady state” refers to the achievement of a stable response in serum total testosterone levels to exogenously administered testosterone undecanoate, typically achieved after at least 15 days following the start of a dosing regimen.
  • In some embodiments, the titration can also include the adjustment of the way the total daily dose is administered such as whether it is administered as two or three doses within a 24 hour period, whether it is administered with a meal, with a meal with a particular fat content, or at a particular hour of the day.
  • As used herein, “initial daily dose” (IDD) or “Daily dose of the initial regimen” is a dose of testosterone undecanoate administered daily to a subject in need of testosterone therapy. The initial daily dose may be administered in two or more intervals over a 24 hour period, e.g. twice-a-day. Similarly, “maintenance daily dose” or “daily dose of the maintenance regiment” is a dose of testosterone undecanoate administered daily to a subject in need of testosterone therapy as determined based on measurement of the titration node day titration metric and is the daily dose going forward within a few days of measurement unless a dose change is needed based on a another titration node day measurements. During a maintenance regime there may be two or more daily doses administered which at some point during the regime would be considered to be the maintenance daily dose.
  • As used herein, “titration node” or “titration node day” are used interchangeably and refer to a day on which a serum sample is drawn from a subject for measurement of the serum testosterone concentrations in order to determine whether a testosterone undecanoate dose titration is necessary and what the titration type might need to be. The measured serum testosterone levels may also be used to determine dose a titration metric to be utilized in deciding dose titration needs for an individual subject. As dosing regimens can include one or more titration node day the term may refer to a first titration node during a dosing regimen (e.g. between the initial dosing regimen and the maintenance dosing regimen) or it can refer to a subsequent titration node day between a maintenance dosing regimen and a subsequent maintenance dosing regimen.
  • As used herein, “titration day” refers to the day when administration of a newly titrated (adjusted) dose is initiated. It should be noted that one or more titrations can be conducted to arrive at a maintenance daily dose in a maintenance regimen. Thus, the maintenance daily dose and regimen is considered to be the dose based on the last or most recent titration.
  • As used herein, “titration metric” is a pharmacokinetic (PK) parameter determined from a serum sample on a titration node day. The PK parameter used as the titration metric can include the serum testosterone Cmax, Cavg, Cmin, Cpre-dose or Ct (serum concentration at a particular time of day). The titration metric can be used to aid in the determination of whether a dose titration is necessary, its magnitude, and other factors possibly included in the titration adjustment.
  • As used herein, the terms “release” and “release rate” are used interchangeably to refer to the discharge or liberation of a substance, including without limitation a drug, from the dosage form into a surrounding environment such as an aqueous medium either in vitro or in vivo.
  • As used herein, an “effective amount” or a “therapeutically effective amount” of a drug refers to a non-toxic, but sufficient amount of the drug, to achieve therapeutic results in treating a condition for which the drug is known to be effective. It is understood that various biological factors may affect the ability of a substance to perform its intended task. Therefore, an “effective amount” or a “therapeutically effective amount” may be dependent in some instances on such biological factors. Further, while the achievement of therapeutic effects may be measured by a physician or other qualified medical personnel using evaluations known in the art, it is recognized that individual variation and response to treatments may make the achievement of therapeutic effects a somewhat subjective decision. The determination of an effective amount is well within the ordinary skill in the art of pharmaceutical sciences and medicine. See, for example, Meiner and Tonascia, “Clinical Trials: Design, Conduct, and Analysis,” Monographs in Epidemiology and Biostatistics, Vol. 8 (1986), incorporated herein by reference.
  • As used herein, the term “delayed release” refers to the release into an aqueous solution of the testosterone undecanoate from the composition or oral dosage form in a time delayed manner attributed either to the inherent nature of the composition or to a coating which may surround the composition or the oral dosage form. A traditional gelatin or non-gelatin non-enteric capsule shell does not alone constitute a delayed release mechanism. In one embodiment, the delayed release is such that about 20% or less of the testosterone undecanoate is released within the first 15 minutes after the composition is contacted by the aqueous solution.
  • The terms “plasma testosterone concentration,” “testosterone concentration in the blood,” and “serum testosterone concentration” are used interchangeably and refer to the “total” testosterone concentration which is the sum of the bioavailable testosterone including free and protein-bound testosterone concentrations. As with any bio-analytical measure, for increased consistency the method employed to measure initial serum testosterone levels should be consistent with the method used to monitor and re-measure serum testosterone levels during clinical testing and testosterone therapy for a subject. Unless otherwise stated, “testosterone concentration” refers to serum total testosterone concentration.
  • As used herein, of the average serum testosterone concentration can be determined using methods and practices known in the art. For example, the average baseline plasma testosterone concentration of a human male is the arithmetic mean of the total plasma testosterone concentrations determined on at least two consecutive time points that are reasonably spaced from each other, for example from about 1 hour to about 168 hours apart. In a particular case, the plasma testosterone concentration can be determined on at least two consecutive times that are about 12 hours to about 48 hours apart. In another particular method, the plasma testosterone concentration of the human male can be determined at a time between about 5 o'clock and about 11 o'clock in the morning. Further, the plasma testosterone concentration can be the determined by standard analytical procedures and methods available in the art, such as for example, automated or manual immunoassay methods, liquid chromatography or liquid chromatography-tandem mass spectrometry (LC-MSMS) etc.
  • As used herein, the term AUC0-t is the area under the curve of a plasma-versus-time graph determined for the analyte from the time 0 to time “t”.
  • As used herein, the term “Cavg,” “Cave,” or “C-average” are used interchangeably, and is determined as the AUC0-t or the mean AUC divided by the time period (t). For example, Cavg-8 h is the average plasma concentration over a period of 8 hours post-dosing determined by dividing the AUC0-8 value by 8. Similarly, Cavg-12 h is the average plasma concentration over a period of 12 hours post-dosing determined by dividing the AUC0-12 value by 12; Cavg-24 h is the average plasma concentration over a period of 24 hours post-dosing determined by dividing the AUC0-24 h, value by 24, and so on. Unless otherwise stated, all Cave values are considered to be Cave-24 h.
  • As used herein, “Ct” refers to the serum concentration of testosterone at time “t” prior to or after administration of the dosage of the current invention. The time “t” is generally in hours, unless otherwise specified. For example, a Ct of “C(−2 to 0) refers to serum testosterone concentration measured in sample collected between the time of about 2 hours before and just immediately prior to dosage administration to the subject tested. Similarly, Ct of “C(2 to 4)” refers to serum testosterone concentration measured in sample collected between the time of about 2 hours and 4 hours after administration of a dosage to the subject tested.
  • As used herein “SIF” or “simulated intestinal fluid” refers to “intestinal fluid, simulated TS” in accordance with the USP. In one embodiment, the SIF does not contain pancreatic enzyme. In another embodiment, SIF may be a fed or fasted simulated intestinal aqueous solution comprising phosphatidyl choline and from about 2 mM to 20 mM bile salts.
  • As used herein “SGF” or “simulated gastric fluid” refers to “Gastric fluid, Simulated TS” in accordance with the USP. In one embodiment, the SGF does not contain the enzyme pepsin. In another embodiment, the SGF may also be a simple 0.1 N HCl solution in water.
  • As used herein, “free of” or “substantially free of” of a particular compound or compositions refers to the absence of any separately added portion of the referenced compound or composition. Free of or substantially free of can include the presence of 1 wt % or less (based on total composition weight) of the referenced compound which may be present as a component or impurity of one or more of the ingredients.
  • As used herein, the term “about” is used to provide flexibility to a numerical range endpoint by providing that a given value may be “a little above” or “a little below” the endpoint. As used herein, a plurality of items, structural elements, compositional elements, and/or materials may be presented in a common list for convenience. However, these lists should be construed as though each member of the list is individually identified as a separate and unique member. Thus, no individual member of such list should be construed as a de facto equivalent of any other member of the same list solely based on their presentation in a common group without indications to the contrary.
  • As used herein, a plurality of items, structural elements, compositional elements, and/or materials may be presented in a common list for convenience. However, these lists should be construed as though each member of the list is individually identified as a separate and unique member. Thus, no individual member of such list should be construed as a de facto equivalent of any other member of the same list solely based on their presentation in a common group without indications to the contrary.
  • Concentrations, amounts, levels and other numerical data may be expressed or presented herein in a range format. It is to be understood that such a range format is used merely for convenience and brevity and thus should be interpreted flexibly to include not only the numerical values explicitly recited as the limits of the range, but also to include all the individual numerical values or sub-ranges or decimal units encompassed within that range as if each numerical value and sub-range is explicitly recited. As an illustration, a numerical range of “about 1 to about 5” should be interpreted to include not only the explicitly recited values of about 1 to about 5, but also include individual values and sub-ranges within the indicated range. Thus, included in this numerical range are individual values such as 2, 3, and 4 and sub-ranges such as from 1-3, from 2-4, and from 3-5, etc., as well as 1, 2, 3, 4, and 5, individually. This same principle applies to ranges reciting only one numerical value as a minimum or a maximum. Furthermore, such an interpretation should apply regardless of the breadth of the range or the characteristics being described.
  • Invention
  • Reference will now be made in detail to preferred embodiments of the invention. While the invention will be described in conjunction with the preferred embodiments, it will be understood that it is not intended to limit the invention to those preferred embodiments. To the contrary, it is intended to cover alternatives, variants, modifications, and equivalents as may be included within the spirit and scope of the invention as defined by the appended claims.
  • In one embodiment, a pharmaceutical capsule for oral delivery is provided. The capsule includes a capsule shell and a capsule fill. The capsule fill can include a solubilizer and about 14 wt % to about 35 wt % testosterone undecanoate based on the total weight of capsule fill. The oral dosage capsule is such that when a single oral administration to a male subject of one or more capsules with a total testosterone undecanoate daily dose of about 350 mg to about 650 mg it provides a ratio of serum testosterone Cmax to serum testosterone Cave of about 2.7 or less.
  • The compositions and oral dosage capsules of the present invention can be used to treat subjects, particularly human males, or even more particularly males who suffer from testosterone deficiency or hypogonadism. Accordingly, in one embodiment of the present invention, a method for providing a serum concentration of testosterone within a target serum testosterone concentration Cave range for a male subject is provided. The method includes the step of orally administering to the male subject a daily dose of a testosterone undecanoate-containing composition. The testosterone undecanoate comprises about 14 wt % to about 35 wt % of the testosterone undecanoate-containing composition and the daily dose provides about 350 mg to about 420 mg of testosterone undecanoate to the male subject. In one specific embodiment, the testosterone undecanoate comprises about 14 wt % to about 18 wt % of the testosterone undecanoate-containing composition.
  • Testosterone deficiency is typically associated with a particular condition that is the source or causes the deficiency. The compositions and oral dosage capsules of the present invention can be used to treat any condition associated with testosterone deficiency, including complete absence, of endogenous testosterone. Examples of conditions associated with testosterone deficiency that can be treated using the oral dosage capsules and/or compositions of the present invention include, but are not limited to congenital or acquired primary hypogonadism, hypogonadotropic hypogonadism, cryptorchidism, bilateral torsion, orchitis, vanishing testis syndrome, orchidectomy, Klinefelter's syndrome, post castration, eunuchoidism, hypopituitarism, endocrine impotence, infertility due to spermatogenic disorders, impotence, male sexual dysfunction (MSD) including conditions such as premature ejaculation, erectile dysfunction, decreased libido, and the like, micropenis and constitutional delay, penile enlargement, appetite stimulation, testosterone deficiency associated with chemotherapy, testosterone deficiency associated with toxic damage from alcohol, testosterone deficiency associated with toxic damage from heavy metal, osteoporosis associated with androgen deficiency, and combinations thereof.
  • Other conditions that can be treated by the compositions and oral dosage forms disclosed herein include idiopathic gonadotropin, LHRH deficiency, or pituitary hypothalamic injury from tumors, trauma, or radiation. Typically, these subjects have low serum testosterone levels but have gonadotropins in the normal or low range. In one embodiment, the compositions or oral dosage forms may be used to stimulate puberty in carefully selected males with clearly delayed puberty not secondary to pathological disorder. In another embodiment, the compositions and oral dosage forms may be used in female-to-male transsexuals in order to maintain or restore male physical and sexual characteristics including body muscle mass, muscle tone, bone density, body mass index (BMI), enhanced energy, motivation and endurance, restoring psychosexual activity etc. In some embodiments, the testosterone undecanoate compositions and oral dosage capsules may be useful in providing hormonal male contraception.
  • Additionally, testosterone therapy can also be used to improve the quality of life of subjects suffering from conditions such as decreased libido, diminishing memory, anemia due to marrow failure, renal failure, chronic respiratory or cardiac failure, steroid-dependent autoimmune disease, muscle wasting associated with various diseases such as AIDS, preventing attacks of hereditary angioedema or urticaria; andropause, and palliating terminal breast cancer. In some situations, certain biomarkers such as for example, increased SHBG levels, can be used to diagnose a subject who may be in need of testosterone therapy. These biomarkers can be associated with conditions/disease states such as anorexia nervosa, hyperthyroidism, hypogonadism, androgen insensitivity/deficiency, alcoholic hepatic cirrhosis, primary biliary cirrhosis, and the like.
  • Subjects that can be treated by the testosterone undecanoate compositions and oral dosage capsule of the present disclosure can be any human male in need thereof. In particular, in one embodiment, the human male may be at least 14 years of age. In another embodiment, the human male is an adult of at least age 30. In a further embodiment, the subject can be an adult male of at least age 50. In yet a further embodiment, the subject can be an adult male of at least age 60.
  • As discussed above, the compositions and oral dosage capsules disclosed herein can be used to treat testosterone deficiency in human males. In one embodiment, the human male being treated can have an average baseline plasma testosterone concentration of about 400 ng/dL or less. In another embodiment, the human male being treated can have an average baseline plasma testosterone concentration of about 350 ng/dL or less. In another embodiment, the human male being treated can have an average baseline plasma testosterone concentration of about 300 ng/dL or less. In another embodiment, the human male being treated can have an average baseline plasma testosterone concentration of about 250 ng/dL or less. In still another embodiment, the human male being treated can have an average baseline plasma testosterone concentration of about of about 190 ng/dL or less. In still a further embodiment, the human male has an average baseline plasma testosterone concentration of about 400 ng/dL or less, along with a co-morbid condition of insulin resistance.
  • Further, there are several biomarkers that can be used to identify patients who need testosterone therapy through the administration of the compositions and/or dosage forms of the current invention. Accordingly, in one embodiment, the human male being treated can have a low density lipoproteins (LDL) level in greater than about 130 mg/dL of blood. In another embodiment, the human male being treated can have a high density lipoproteins (HDL) level less than about 40 mg/dL of blood. In still another embodiment, the human male being treated can have a total cholesterol level greater than about 220 mg/dL of blood. In yet a further embodiment, the human male being treated can have an average TG (triglycerides) levels greater than 250 mg/dL of blood. In one embodiment, the testosterone undecanoate dosage forms of the current invention can be administered to human male whose bioavailable or free or un-bound plasma estradiol levels are about 20 pg/mL or less. In another embodiment, dosage forms of the current invention can be administered to human male who has a ratio of the bioavailable or free or unbound plasma testosterone level to the bioavailable or free or un-bound plasma estradiol level at about 100 or less.
  • The testosterone undecanoate compositions and oral dosage capsules of the current invention can be administered orally to a human male who has an average body mass index (BMI) of about 28 kg/m2 or more. In another embodiment, the human male has an average BMI of about 30 kg/m2 or more. In another embodiment, the human male has an average BMI of about 37 kg/m2 or more. In a further embodiment, the subject male being treated can have a serum sex hormone binding globulin (SHBG) levels of about 40 nmol/L or more. In yet another embodiment, the human male being treated can have a serum SHBG levels of about 60 nmol/L or more.
  • It was found that the pharmaceutical compositions and oral dosage capsules of the present invention have the ability to provide for increased stability of the testosterone undecanoate present in the formulation. In particular, the pharmaceutical compositions and oral dosage capsules of the present invention can provide for superior stability with respect to the degradation of the testosterone undecanoate that can occur during storage as compared to other formulation containing lower testosterone undecanoate concentration. In one embodiment, the pharmaceutical compositions and oral dosage capsules of the present invention can have increased stability such that, when stored for a period of at least three months there is at least 20% less degradation of the testosterone undecanoate as compared to testosterone undecanoate containing compositions having less than 14 wt % testosterone undecanoate. In another embodiment, the pharmaceutical compositions and oral dosage capsules of the present invention can have increased stability such that, when stored for a period of at least three months there is at least 20% less degradation of the testosterone undecanoate as compared to testosterone undecanoate containing compositions having less than 16 wt % testosterone undecanoate.
  • Further, it has been discovered that the pharmaceutical compositions and oral dosage capsules disclosed herein can provide therapeutically effective treatment without the need to include oils, triglycerides, and/or hydrophilic surfactants. Accordingly, in one embodiment, the pharmaceutical compositions and oral dosage capsules can be free of oil. In another embodiment, the pharmaceutical composition and oral dosage capsules can be free of triglycerides. In one embodiment, the composition or capsule fill can comprise 25 wt % or less of total triglycerides. In one embodiment, the composition or capsule fill can be free of ionizable fatty acids. In another embodiment, the composition or capsule fill can be free of oleic acid. Without wishing to be bound by theory, it is believed that testosterone undecanoate-containing compositions or capsule fill that comprise greater than 25 wt % triglycerides have a higher dependence on digestion upon oral administration than do those in which the triglycerides comprise 25 wt % or less of the total composition or capsule fill. In one embodiment, the capsule fill can comprise 15 wt % or less of triglycerides. In yet another embodiment, the capsule fill can comprise 10 wt % or less of triglycerides. In yet a further embodiment, the capsule fill can comprise 5 wt % or less of triglycerides.
  • In yet a further embodiment, the pharmaceutical compositions and oral dosage capsules can be free of hydrophilic surfactants. In yet a further embodiment, the composition can include a hydrophilic surfactant as a dispersant and the hydrophilic surfactant can be present in an amount such that it does not appreciably solubilize the testosterone undecanoate in the composition. A hydrophilic surfactant is said to “not appreciably solubilize” testosterone undecanoate when it solubilizes 5 wt % or less of the testosterone undecanoate in the composition or the dosage form. In one embodiment, a hydrophilic surfactant is deemed to “not appreciably solubilize” testosterone undecanoate when it solubilizes 2 wt % or less of the testosterone undecanoate in the composition or oral dosage capsule. In all of these embodiments, the pharmaceutical compositions and oral dosage capsules can still be capable of providing the necessary dispersion and pharmacokinetics parameters to effectively treat testosterone deficiency.
  • The testosterone undecanoate can be present in the pharmaceutical compositions and oral dosage capsules in amounts sufficient to comprise 14 wt % to about 35 wt % of the composition or capsule fill. In one embodiment, the testosterone undecanoate can make up about 15 wt % to about 30 wt % of the composition or oral dosage capsule. In yet a further embodiment, the oral dosage capsule can comprise about 18 wt % to about 25 wt % of the composition or oral dosage capsule. In still a further embodiment, the compositions and/or capsule fill material can be such that the testosterone undecanoate comprises about 14 wt % to about 18 wt % of the total composition or capsule fill. In one embodiment, at least 35 wt % of the testosterone undecanoate in the composition or capsule fill can be in dissolved form. In yet another embodiment, at least about 66 wt % of the testosterone undecanoate in the capsule fill or composition can be present in dissolved form. In another embodiment, at least about 5 wt % of the testosterone undecanoate in the capsule fill or composition can be present in undissolved form.
  • The oral dosage capsules of the present application can include dosages of testosterone undecanoate of at least 50 mg. The oral dosage capsules of the present application can include dosages of testosterone undecanoate of about 80 mg to about 400 mg. In another embodiment, the oral dosage capsule can include about 80 mg to about 140 mg testosterone undecanoate. In another embodiment, the oral dosage capsule can include about 120 mg to about 300 mg testosterone undecanoate. In yet a further embodiment, the oral dosage capsule can include about 150 mg to about 250 mg of testosterone undecanoate. With this in mind, the compositions and oral dosage capsule can be used as part of dosing regimens to provide daily doses of about 250 mg to about 650 mg per day, preferably, daily doses of about 350 mg to about 650 mg per day
  • The solubilizers used in the pharmaceutical compositions and oral dosage capsules of the present invention play role in the ability of the formulation to provide the desired therapeutic characteristics. Solubilizers that can be used can be selected from a variety of compounds and mixtures of compounds that have the ability to facilitate loading of testosterone undecanoate. The solubilizer can comprise about 50 wt % to about 86 wt % of the composition or capsule fill. In one embodiment, the solubilizer can comprise about 55 wt % to about 82 wt % of the pharmaceutical composition or oral dosage capsule. In another embodiment, the solubilizer can comprise about 60 wt % to about 80 wt % of the pharmaceutical composition or oral dosage capsule. In one embodiment, the solubilizer can be such that the testosterone undecanoate can have solubility in the solubilizer, at about 37° C., of about 250 mg/g to about 750 mg/g (mg testosterone undecanoate/gram of solubilizer and testosterone undecanoate).
  • Non-limiting examples of solubilizers that can be used include C8 to C22 fatty acid glycerides, omega fatty acids, and mixtures thereof. In one embodiment, the C8 to C22 fatty acid glycerides can include C8 to C22 medium and/or long chain monoglycerides, medium and/or long chain diglycerides, or mixtures of a mixture of medium and/or long chain monoglycerides and medium and/or long chain diglycerides. In another embodiment, the solubilizer can consist essentially of medium and/or long chain monoglycerides and/or diglycerides. Medium to long chain monoglycerides and diglycerides refers to compounds having chain lengths of C8 to C22. In one embodiment, the mixture of monoglycerides and diglycerides can have chain lengths of C8 to about C13. In another embodiment, the mixture of monoglycerides and diglycerides can have chain lengths of about C14 to about C22. When the solubilizer includes C8 to C22 fatty acid glycerides, monoglycerides can comprise at least about 40 wt % of the C8 to C22 fatty acid glycerides (such as commercially available Maisine® 35-1, Capmul® MCM, Peceol,® and the like). In another embodiment, the monoglycerides can comprise at least about 60 wt % of the C8 to C22 fatty acid glycerides. In yet a further embodiment, the monoglycerides can comprise at least about 80 wt % of the C8 to C22 fatty acid glycerides.
  • Non-limiting examples of C8 to C22 fatty acid glycerides that can be used as solubilizers in pharmaceutical compositions and oral dosage capsules of the present invention include monoglycerides and/or diglycerides derived from sources such as maize oil, poppy seed oil, safflower oil, sunflower oil, borage seed oil, coconut oil, palm kernel oil, castor oil, and mixtures thereof. Although not essential, the solubilizer can also include a triglyceride. The triglyceride can be a medium and/or long chain triglyceride, or mixture thereof, and can be present alone or with other solubilizers. The triglycerides can be selected from a variety of well-known pharmaceutically acceptable triglycerides including, but not limited to vegetable oils such as peanut oil, safflower oil, sunflower oil, olive oil, castor oil, corn oil, maize oil, flax seed oil, wheat-germ oil and the like, or their hydrogenated derivatives and their mixtures thereof. Additional triglyceride sources can include animal derived oils such as fish oil, seal oil, whale oil, and the like, triglycerides of C8-C22 fatty acids or their mixtures; triglycerides of C8-C13 fatty acids; triglycerides of C14-C22 fatty acids. In one embodiment, the composition can include a fatty acid triglyceride and the testosterone undecanoate can comprise at least about 25 wt % of the composition. In another embodiment, the triglyceride can be castor oil. In yet a further embodiment, the castor oil can comprise about 45 wt % or less of the total composition. In yet another embodiment, the castor oil can comprise about 40 wt % or less of the solubilizer. In a further embodiment, the composition can be free of castor oil. In one embodiment of the invention, the solubilizer can include a glyceryl palmitostearate, a glyceryl stearate, a glyceryl distearate, glyceryl monostearate, or a combination thereof.
  • In another aspect of the invention, the solubilizer can include a C8 to C22 fatty acid glycerides that is monoglycerides and/or diglycerides of capric acid, caprylic acid, or mixtures thereof. In another embodiment, the solubilizer can include a C8 to C22 fatty acid glycerides that is a monoglycerides and/or diglycerides of linoleic acid, oleic acid, or mixtures thereof. Other examples of C8 to C22 fatty acids that can be used include capric acid, pelargonic acid, caprylic acid, undecanoic acid, lauric acid, myristic acid, palmitic acid, stearic acid, oleic acid, linoleic acid, linolenic acid, arachodonic acid, eicosapentaenoic acid, docosahexanoic acid, and mixtures thereof. In one embodiment, the C8 to C22 fatty acid can be capric acid, caprylic acid, undecanoic acid, lauric acid, myristic acid, palmitic acid, stearic acid, oleic acid, linoleic acid, linolenic acid or mixtures thereof. In another embodiment, the C8 to C22 fatty acid can be selected from the group consisting of capric acid, caprylic acid, oleic acid, linoleic acid, and mixtures thereof. In one embodiment, the composition or capsule fill can be free of ionizable fatty acids. In another embodiment, the composition or capsule fill can be free of oleic acid.
  • In a further embodiment, the solubilizer can include an alcohol. Non-limiting examples of alcohols that can be used as solubilizers include tocopherol, ethyl alcohol, isopropanol, butanol, benzyl alcohol, ethylene glycol, propylene glycol, butanediol, glycerol, pentaerythritol, transcutol, dimethyl isosorbide, polyethylene glycol and mixtures thereof. In one embodiment, the solubilizer can be ethyl alcohol, benzyl alcohol, tocopherol, and mixtures thereof.
  • The pharmaceutical compositions and oral dosage capsules can also include a dispersant. In one aspect of the invention, the dispersant can be a hydrophilic surfactant having an HLB value of greater than 10, a lipophilic surfactant having an HLB value of 10 or less, or combinations thereof. In one embodiment, the compositions and oral dosage forms can include at least one hydrophilic surfactant. In another embodiment the capsule fill includes at least one hydrophilic surfactant and at least one lipophilic surfactant.
  • Unlike dosage forms containing ionizable components such as fatty acids (e.g. oleic acid), which are prone to being ionized at higher pH values thereby becoming charged and serving as a hydrophilic surfactant, it has been found that for improved bioavailability or activity of TU for testosterone therapy it can be useful for a composition's performance to be robust with regards to inter-conversion between hydrophilic and hydrophobic species as determined by the absence of ionized ionizable fatty acid due to pH changes such as encountered in the gastro-intestinal tract.
  • The total amount of lipophilic component is the total amount in wt % of the lipophilic components including the mono-, di- and/or tri-glycerides and the lipophilic surfactants, if present in the composition. In one embodiment, the lipophilic surfactant includes the solubilizer and the lipophilic surfactant. The total amount of hydrophilic surfactant is the total amount (in wt %) of the added hydrophilic surfactant and that hydrophilic surfactant formed in situ in an aqueous medium as a function of pH (e.g. intestinal pH) due to the hydrophilic ionized ionizable fatty acid (e.g. oleate) formed from lipophilic unionized ionizable fatty acid (oleic acid).
  • Therefore, even though fatty acid such as oleic acid may be a good solubilizer for testosterone undecanoate, its bioavailability and activity is substantially compromised with fatty acid containing compositions either by being unable to continue to solubilize the drug, or be inadequate facilitator for chylomicron related testosterone undecanoate absorption or can be slow to allow drug to partitioning out of the carrier.
  • When present, the hydrophilic surfactant can, but does not have to have appreciable solubilizing effect for the testosterone undecanoate present in the composition. Non-limiting examples of hydrophilic surfactants that can be included are non-ionic hydrophilic surfactants such as polysorbates, polyoxyethylene hydrogenated vegetable oils, polyoxyethylene vegetable oils; polyoxyethylene sorbitan fatty acid esters; polyoxyethylene-polyoxypropylene block copolymers; polyglycerol fatty acid esters; polyoxyethylene glycerides; polyoxyethylene sterols, derivatives and analogues thereof reaction mixtures of polyols and at least one member of the group consisting of fatty acids, glycerides, vegetable oils, hydrogenated vegetable oils, fractionated oils and sterols; tocopheryl polyethylene glycol succinates; sugar esters; sugar ethers; sucroglycerides; mixtures thereof; alkylglucosides; alkylmaltosides; alkylthioglucosides; lauryl macrogolglycerides; polyoxyethylene alkyl ethers; polyoxyethylene alkylphenols; polyethylene glycol fatty acids esters; polyethylene glycol glycerol fatty acid esters; polyoxyethylene sorbitan fatty acid esters; polyoxyethylene-polyoxypropylene block copolymers such as poloxamer—108, 188, 217, 238, 288, 338, 407, 124, 182, 183, 212, 331, or 335, or combinations thereof; ionic hydrophilic surfactants such as sodium dodecyl sulphate, docusate sodium; bile acid, cholic acid, deoxycholic acid, chenodeoxycholic acid, salts thereof, and mixtures thereof. In one embodiment, the pharmaceutical composition or oral dosage form can be substantially free of hydrophilic surfactants.
  • In one embodiment, the hydrophilic surfactant can have at least one characteristic of: 1) being present in an amount such that it does not appreciably solubilize testosterone undecanoate present in the composition; or 2) the solubility of testosterone undecanoate in the hydrophilic surfactant at about 25° C., is less than 100 mg/gram or less, based on the total weight of the testosterone undecanoate and the solubilizer.
  • In one embodiment, the hydrophilic surfactant can have at least one characteristic of: 1) being present in an amount such that it solubilizes less than 5 wt % of the testosterone undecanoate present in the composition; or 2) the solubility of testosterone undecanoate in the hydrophilic surfactant at about 25° C., is less than 100 mg/gram or less, based on the total weight of the testosterone undecanoate and the surfactant. In another embodiment, the hydrophilic surfactant can have at least one characteristic of: 1) the hydrophilic surfactant is present in an amount such that it solubilizes less than 5 wt % of the testosterone undecanoate present in the composition; or 2) the solubility of testosterone undecanoate in the hydrophilic surfactant at about 25° C., is about 50 mg/gram or less, based on the total weight of the testosterone undecanoate and the surfactant. In yet a further embodiment, the hydrophilic surfactant can have a least one characteristic of: 1) the hydrophilic surfactant is present in an amount such that it solubilizes less than 5 wt % of the testosterone undecanoate present in the composition; or 2) the solubility of testosterone undecanoate in the hydrophilic surfactant at about 25° C. about 10 mg/gram or less, based on the total weight of the testosterone undecanoate and the surfactant. In yet a further embodiment, the hydrophilic surfactant can have the characteristic of: 1) the hydrophilic surfactant is present in an amount such that it solubilizes less than 5 wt % of the testosterone undecanoate present in the composition; and 2) the solubility of testosterone undecanoate in the hydrophilic surfactant at about 25° C., is about 50 mg/gram or less, based on the total weight of the testosterone undecanoate and the surfactant.
  • As discussed above, in some embodiments the compositions and oral dosage capsules can include at least one lipophilic surfactant. Various lipophilic surfactants can be used including, but not limited to reaction mixtures of alcohols or polyalcohols with a variety of natural and/or hydrogenated oils such as PEG-5 hydrogenated castor oil, PEG-7 hydrogenated castor oil, PEG-9 hydrogenated castor oil, PEG-6 corn oil (Labrafil® M 2125 CS), PEG-6 almond oil (Labrafil®M 1966 CS), PEG-6 apricot kernel oil (Labrafil®M 1944 CS), PEG-6 olive oil (Labrafil®M 1980 CS), PEG-6 peanut oil (Labrafil®M 1969 CS), PEG-6 hydrogenated palm kernel oil (Labrafil®. M 2130 BS), PEG-6 palm kernel oil (Labrafil® M 2130 CS), PEG-6 triolein (Labrafil® M 2735 CS), PEG-8 corn oil (Labrafil® WL 2609 BS), PEG-20 corn glycerides (Crovol® M40), PEG-20 almond glycerides (Crovol® A40), lipophilic polyoxyethylene-polyoxypropylene block co-polymers (Pluronic® L92, L101, L121 etc.); propylene glycol fatty acid esters, such as propylene glycol monolaurate (Lauroglycol FCC), propylene glycol ricinoleate (Propymuls), propylene glycol monooleate (Myverol P-O6), propylene glycol dicaprylate/dicaprate (Captex® 200), and propylene glycol dioctanoate (Captex® 800), propylene glycol mono-caprylate (Capryol® 90); propylene glycol oleate (Lutrol OP2000); propylene glycol myristate; propylene glycol mono stearate; propylene glycol hydroxy stearate; propylene glycol ricinoleate; propylene glycol isostearate; propylene glycol mono-oleate; propylene glycol dicaprylate/dicaprate; propylene glycol dioctanoate; propylene glycol caprylate-caprate; propylene glycol dilaurate; propylene glycol distearate; propylene glycol dicaprylate; propylene glycol dicaprate; mixtures of propylene glycol esters and glycerol esters such as mixtures composed of the oleic acid esters of propylene glycol and glycerol (Arlacel® 186); sterol and sterol derivatives such as cholesterol, sitosterol, phytosterol, PEG-5 soya sterol, PEG-10 soya sterol, PEG-20 soya sterol, and the like; glyceryl palmitostearate, glyceryl stearate, glyceryl distearate, glyceryl monostearate, or a combination thereof; sorbitan fatty acid esters such as sorbitan monolaurate (Arlacel 20), sorbitan monopalmitate (Span-40), sorbitan monooleate (Span-80), sorbitan monostearate, and sorbitan tristearate, sorbitan monolaurate, sorbitan monopalmitate, sorbitan monooleate, sorbitan trioleate, sorbitan sesquioleate, sorbitan tristearate, sorbitan monoisostearate, sorbitan sesquistearate, and the like; and mixtures thereof. It is important to note that some lipophilic surfactants may also function as the solubilizer component of the compositions and oral dosage forms.
  • In one embodiment, the lipophilic surfactant can be selected from the group consisting of propylene glycol mono caprylate, propylene glycol oleate, propylene glycol monostearate, propylene glycol monolaurate, propylene glycol mono-oleate, propylene glycol dicaprylate/dicaprate, sorbitan monooleate, PEG-5 hydrogenated castor oil, PEG-7 hydrogenated castor oil, PEG-9 hydrogenated castor oil, PEG-6 corn oil, PEG-6 almond oil, PEG-6 apricot kernel oil, PEG-6 olive oil, PEG-6 peanut oil, PEG-6 hydrogenated palm kernel oil, sorbitan monolaurate (Arlacel 20), sorbitan monopalmitate, sorbitan monooleate, sorbitan monostearate, sorbitan tristearate, sorbitan monolaurate, sorbitan monopalmitate, sorbitan monooleate, sorbitan trioleate, sorbitan sesquioleate, sorbitan tristearate, sorbitan monoisostearate, and combinations thereof.
  • In another aspect of the invention, the pharmaceutical compositions and/or oral dosage capsules, namely the capsule fill, can include a solidifying agent. As defined above, a solidifying agent is a pharmaceutically acceptable additive that is in a solid physical state at 20° C. Typically solidifying agents facilitate the solidification of the pharmaceutical compositions of the present invention at temperatures around room temperature. The compositions and capsule fill of the present invention, including those with solidifying agents, can be non-liquid at standard temperature and pressure. In one embodiment, the composition and capsule fill can be semi-solid at standard temperature and pressure. In yet another embodiment, the composition and capsule fill can be solid at standard temperature and pressure. When present, the solidifying agent can comprise from about 0.1 wt % to about 25 wt % of the pharmaceutical composition or oral dosage capsule. In another embodiment, the solidifying agent can comprise about 2 wt % to about 20 wt % of the composition or oral dosage capsule. In yet a further embodiment, the solidifying agent can comprise about 3 wt % to about 15 wt % of the composition or oral dosage capsule. In still a further embodiment, the solidifying agent can comprise about 3 wt % to about 9 wt % of the capsule fill. In yet a further embodiment, the solidifying agent can comprise 6 wt % to 9 wt % of the capsule fill. In one embodiment, the solidifying agent can melt at a temperature of about 45° C. to about 75° C. Non-limiting examples of solidifying agents that can be used include polyethylene glycols; sorbitol; gelatin; stearic acid; cetyl alcohol; cetosterayl alcohol; paraffin wax; polyvinyl alcohol; glyceryl stearates; glyceryl distearate; glyceryl monostearate; glyceryl palmitostearate; glyceryl behenate; waxes; hydrogenated castor oil; hydrogenated vegetable oil; bees wax, microcrystalline wax; sterols; phytosterols; cholesterol and mixtures thereof. In one embodiment, the solidifying agent includes a polyethylene glycol (PEG) having molecular weight from about 1000 to about 20,000 and their mixtures. In another embodiment the solidifying agent includes one or more selected from the group consisting of polyethylene glycol; gelatin; stearic acid; polyvinyl alcohol; glyceryl stearates; glyceryl distearate; glyceryl monostearate; glyceryl palmitostearate; hydrogenated castor oil; hydrogenated vegetable oil and cholesterol. In one embodiment, the pharmaceutical composition can be a solid at about 20° C. In yet a further embodiment, the solubilized solid and/or the undissolved crystalline testosterone undecanoate can act as a solidifying agent.
  • The oral compositions of the present invention can be formulated to take any dosage form commonly known in the pharmaceutical arts such as granules, tablet or capsule. In one embodiment, the oral dosage form can be a capsule having a pharmaceutical composition of the present invention disposed therein. Both soft and hard gelatin and non-gelatin capsules can be used. The capsule size can be any size known in the art and can vary depending on the desired dosage amount. In one embodiment, the capsule can be a hard gelatin capsule having a fill volume of about 0.3 mL to about 1.1 mL. The oral dosage capsules can be immediate release, extended release, targeted release, enteric release, delayed release dosage form or combinations thereof. In a specific embodiment, the oral dosage capsule can be a delayed release dosage form. In one embodiment, the capsule can have a ratio of the amount of testosterone undecanoate to the volume of the capsule fill can be about 80 mg/mL to about 750 g/mL. In another embodiment, the capsule can have a ratio of the amount of testosterone undecanoate to the volume of the capsule fill can be about 160 mg/mL to about 375 mg/mL.
  • The oral dosage capsules of the present invention can be formulated such that they have distinctive release profiles. In one embodiment, an oral dosage capsule can provide in vitro release of at least about 75 wt % of the testosterone undecanoate during the first 120 minutes when tested using about 1000 mL of 8% w/v Triton X-100 in water maintained at about 37±1° C. taken in a USP-Type II dissolution apparatus set at 100 rpm. In another embodiment, the oral dosage capsule can have an in vitro release profile such that 85 wt % or less of the testosterone undecanoate is released in the first 30 minutes, when measured using about 1000 mL of 8% w/v Triton X-100 in water maintained at about 37±1° C. taken in a USP-Type II dissolution apparatus set at 100 rpm. In further embodiment, the oral dosage capsule can have an in vitro release profile such that 70 wt % or less of the testosterone undecanoate is released in the first 30 minutes, when measured using about 1000 mL of 8% w/v Triton X-100 in water maintained at about 37±1° C. taken in a USP-Type II dissolution apparatus set at 100 rpm. In an additional embodiment, the oral dosage capsule can have a in vitro release profile such that at least 35 wt % of the testosterone undecanoate is released in the first 30 minutes, when measured using about 1000 mL of 8% w/v Triton X-100 in water maintained at about 37±1° C. taken in a USP-Type II dissolution apparatus set at 100 rpm. In still an additional embodiment, the oral dosage capsule can have an in vitro release profile such that at least 40 wt % of the testosterone undecanoate is released in the first 30 minutes, when measured using about 1000 mL of 8% w/v Triton X-100 in water maintained at about 37±1° C. taken in a USP-Type II dissolution apparatus set at 100 rpm.
  • In one aspect, the dosage form can comprise two or more of populations of testosterone undecanoate compositions of the present invention. In one embodiment, at least one of the populations can be formulated to start releasing testosterone undecanoate immediately into a surrounding aqueous medium. In another embodiment, at least one the populations can be formulated to start releasing testosterone undecanoate after at least 2 hours. In another embodiment, at least one the populations can be formulated to release testosterone undecanoate after about 4 hours, or after about 6 hours, or after about 8 hours, or after about 10 hours.
  • In yet a further embodiment, at least one of the populations can be formulated to start releasing testosterone undecanoate immediately after oral administration to a human. In one particular case, at least one of the populations can be formulated to start releasing testosterone undecanoate in the duodenal region after oral administration to a human. In another particular case, at least one of the populations can be formulated to start releasing testosterone undecanoate in the small intestine after oral administration to a human.
  • In yet a further embodiment, at least one of the populations includes a pH sensitive substance. In a particular case, at least one of the populations can be formulated to start releasing testosterone undecanoate at a pH of from about 1.0 to about 3.4. In another particular case, at least one of the populations can be formulated to start releasing testosterone undecanoate at a pH of from about 3.5 to about 5.5. In another particular case, at least one of the populations can be formulated to start releasing testosterone undecanoate at a pH of from about 5.6 to about 6.8. In another particular case, at least one of the populations can be formulated to start releasing testosterone undecanoate at a pH about 7.0 or more.
  • In yet another aspect, the dosage form comprising two or more of populations of testosterone undecanoate compositions of the present invention is a capsule. In a particular case, the dosage form is a capsule in capsule dosage form. In another particular case the dosage form is a tablet in capsule dosage form. In another particular case, the dosage form is a granules or pellets or tablets or minitablets disposed in a capsule.
  • The oral dosage capsules of the present invention can be formulated such that, when administered to a human male they provide a serum total testosterone Cavg ranging about 300 ng/dL to about 1100 ng/dL. In another embodiment, the oral dosage capsules can be formulated such that, upon single administration to a human male, they provide a serum total testosterone Cavg ranging about 350 ng/dL to about 800 ng/dL. In another embodiment, the oral dosage capsules can be formulated such that, upon single administration to a human male, they provide a serum total testosterone Cavg ranging from about 400 ng/dL to about 600 ng/dL. It is noted that such Cavg value can be achieved based on administration every 12 hours or every 8 hours. Similarly, the oral dosage capsules can be formulated such that, upon single administration to a human male, they provide a serum testosterone undecanoate Cavg of about 1.5 ng/mL to about 1 mcg/mL. In a further embodiment, the oral dosage capsules can be formulated such that, upon single administration to a human male, they provide a serum testosterone undecanoate Cavg of about 3 ng/mL to about 850 ng/mL. In a further embodiment, the oral dosage capsules can be formulated such that, upon single administration to a human male, they provide a serum testosterone undecanoate Cavg of about 10 ng/mL to about 850 ng/mL. In one embodiment, upon a single dose administration of the capsule to a subject the capsule provides a dose-normalized serum testosterone Cmax of about 3×10−6 dL−1 or higher. In another embodiment, upon a single dose administration of the capsule to a subject the capsule provides a dose-normalized serum testosterone Cavg of about 1.9×10−6 dL−1 or higher. In yet another embodiment, upon a single dose administration of the capsule to a subject the capsule provides a dose-normalized serum testosterone Cavg of about 2.7×10−6 dL−1 or higher.
  • In another aspect, the oral dosage capsules can be formulated such that upon single administration d to a male human subject they provide a ratio of serum testosterone undecanoate Cavg to serum total testosterone Cavg of about 4:1 to about 75:1. In a further embodiment, the oral dosage capsules can be formulated such that, upon single administration to a human male, they provide a ratio of serum testosterone undecanoate Cavg to serum total testosterone Cavg of about 20:1 to about 50:1. In yet another embodiment, the oral dosage capsules can be formulated such that, upon single administration to a human male, the oral dosage capsule provides a ratio of serum total testosterone Cavg to dose of testosterone undecanoate of about 0.2×10−6 dL−1 to about 20×10−6 dL−1.
  • In one embodiment, a single dose of the testosterone undecanoate composition or oral dosage form can provide a serum total testosterone Cavg of about 300 ng/dL or more from about 0.5 hours to about 24 hours after oral administration with a meal or a snack. In a further embodiment, a single dose of a testosterone undecanoate composition or oral dosage capsule can provide a serum total testosterone Cavg of about 300 ng/dL or more at about 20 hours after oral administration with a meal or a snack. In yet a further embodiment, a single dose of the testosterone undecanoate composition can provide a serum total testosterone Cavg of about 300 ng/dL or more at about 18 hours after oral administration with a meal or a snack. In still a further embodiment, a single dose of the testosterone undecanoate composition can provide a serum total testosterone Cavg of about 300 ng/dL or more at about 16 hours after oral administration with a meal snack. In still a further embodiment, a single dose of the testosterone undecanoate composition can provide a serum total testosterone Cavg of about 300 ng/dL or more at about 12 hours after administration after oral administration with a meal snack. In still a further embodiment, a single dose of the testosterone undecanoate composition can provide a serum total testosterone Cavg of about 300 ng/dL or more at about 8 hours after oral administration with a meal or snack. The meal that is administered with the composition or oral dosage form can be a standard meal or a snack.
  • The compositions and oral dosage capsules disclosed herein can be, but do not have to be, orally administered with food. In one embodiment, the composition or oral dosage capsule can be administered with a meal, such as a meal that provides about 200 to about 1000 calories of energy. In another embodiment, the composition or oral dosage capsule can be administered with a standard meal. In another embodiment, the composition or oral dosage capsule can be administered with a meal that provides about 50% of the calories derived from the fat. In another embodiment, the composition or oral dosage capsule can be administered with a high-fat, high calorie meal. In another embodiment, the composition or oral dosage capsule can be administered with a meal that provides about 500 to about 1000 calories of energy. In another embodiment, the composition or oral dosage capsule can be administered with a meal that provides about 400 to about 700 calories derived from the fat therein. The compositional make-up of the meals that are administered can vary depending on the tastes and dietary needs of a subject. However, in some situations it may be beneficial to administer the compositions and oral dosage forms with meals that provide no fat or up to about 50 g of fat. In one embodiment, the meal can provide about 10 g to about 50 g of fat. In yet a further embodiment, the meal can provide about 30 g of fat. The testosterone undecanoate dosage compositions and oral dosage capsules disclosed herein can be orally administered in a 24 hours' dosing regimen (also referred to as or a daily dosing regimen) that is suitable to the needs of the subject. The 24 hours' dosing regimen can include administering the dosage forms after meals in the morning, at about noon, in the evening, at about night time or combinations thereof. The 24 hours' dosing regimen can include dosing one or more dosage units at one or more administration times. In one embodiment, the pharmaceutical composition is administered as a single oral dosage capsule.
  • The testosterone undecanoate compositions and oral dosage capsules can provide increased bioavailability as compared to other testosterone undecanoate compositions and dosage forms. In some embodiments, the testosterone undecanoate oral dosage capsules can provide an in vitro release of less than about 85 wt % of the testosterone undecanoate within the first 30 minutes. In another embodiment, the testosterone undecanoate oral dosage capsules can provide an in vitro release of about 90 wt % or less testosterone undecanoate within the first 30 minutes. The in vitro release is determined in about 1000 mL of 8% w/v Triton X-100 in water maintained at about 37° C. in an USPType-2 Apparatus at about 100 rpm. It has been discovered that these testosterone undecanoate oral dosage capsules, i.e. those having the above release characteristics, provide at least a 10% increase in the testosterone undecanoate AUC after single oral dosages are administered to human males. The increase is as compared to equivalent dosages of testosterone undecanoate in an immediate release dosage forms administered under same conditions. Immediate release dosage forms are defined as being dosage forms which release more than 95 wt % of the testosterone undecanoate within the first 30 minutes using the same in vitro release conditions described above. Further, in one embodiment, the testosterone undecanoate oral dosage capsules can provide at least a 15% increase in the testosterone undecanoate AUC as compared to an immediate release dosage oral dosage form.
  • In another embodiment, the testosterone undecanoate oral dosage capsules disclosed herein can provide at least a 10% reduction in the inter-subject variability of the testosterone undecanoate Cmax, and/or the testosterone undecanoate AUC as compared to immediate release equivalent dosage containing oral dosage forms. In another embodiment, the testosterone undecanoate oral dosage capsules disclosed herein can provide 10% or more testosterone bioavailability in subjects as compared to immediate release equivalent dosed oral dosage forms.
  • The pharmaceutical compositions and oral dosage capsules of the present invention can be formulated such that upon administration of one or more capsules daily to each subject in a group of at least 24 hypogonadal males for a period of at least 84 days, the capsule provides a serum testosterone Cavg of 300 ng/dL to 1100 ng/dL in at least 75% of the hypogonadal males in the group. Additionally, under such an administration regimen the capsule can be such that at least one of the following regarding the PK parameter of the administration is true: a) a serum testosterone Cmax of less than 1500 ng/dL in at least 85% of the hypogonadal males in the group; b) a serum testosterone Cmax of about 1800 ng/dL to about 2500 ng/dL in 5% or less of the hypogonadal males in the group; or c) a serum testosterone Cmax greater than 2500 ng/dL in about 1% or less of the hypogonadal males in the group.
  • In one embodiment, the administration over the at least 84 days can be divided into two dosing regimens including an initial regimen in which an initial dose is administered and a maintenance regimen in which a maintenance dose is administered. In one aspect of this embodiment, the daily dose of the maintenance regimen of the testosterone undecanoate can be about 45% to about 155% of the initial daily dose. In another aspect of the embodiment, the daily dose of the maintenance regimen of the testosterone undecanoate can be about 66% to about 133% of the initial daily dose. In another aspect of the embodiment, the daily dose of the maintenance regimen of the testosterone undecanoate can be about 75% to about 125% of the initial daily dose. In yet a further aspect of this embodiment, the capsule can be formulated such that the amount of the daily dose of the maintenance regimen is based on at least one testosterone undecanoate dose titration metric derived from the measurement of the serum testosterone concentration on at least one titration node day. The titration node day can be any single day beginning on day 15 to day 84 after administration of the initial daily dose. In one embodiment, the titration node day can be any day from day 15 to day 21 after administration the daily dose of the initial regimen of the initial regimen. In yet another embodiment, the titration node day is any day from day 22 to day 30 after administration of the daily dose of the initial regimen of the initial regimen. In further embodiments, the titration node day can be any day from day 22 to day 30, from day 31 to day 63, or from day 64 to day 84 after administration of the daily dose of the initial regimen of the initial regimen.
  • In one aspect, the titration based on the titration metric can be such that if the testing of the metric is serum testosterone concentration and the testing is done at time (t) of 1 to less than 3 hours following administration the serum testosterone concentration is less than 12.0 ng/mL then the daily TU dose may need to be increased and if the serum testosterone concentration is more than 9.4 then the daily TU dose may need to be decreased. In another aspect, the titration metric is serum testosterone concentration and the testing is done at time (t) ii) if at time (t) of 3 to less than 8 hours following administration the serum testosterone concentration is less than 4.1 ng/mL daily TU dose may need to be increased and if the serum testosterone concentration is more than 18.1 then the daily TU dose may need to be decreased. In another aspect, the titration metric is serum testosterone concentration and the testing is done at time (t) of 8 to less than 12 hours following administration the serum testosterone concentration is less than 3.0 ng/mL then the daily TU dose may need to be increased and if the serum testosterone concentration is more than 7.8 then the daily TU dose may need to be decreased. In yet a further aspect, the titration metric is serum testosterone concentration and the testing is done at time (t) of 12 to less than 14 hours following administration the serum testosterone concentration is less than 1.4 ng/mL then the daily TU dose may need to be increased and if the serum testosterone concentration is more than 2.9, then the daily TU dose may need to be decreased. The above titrations are only exemplary of the possible titrations that can be accomplished using the methods of the present invention. It is further noteworthy that the titrations can be used in conjunction with the methods taught herein including the disclosed methods of providing a serum concentration of testosterone within a target serum testosterone Cave range for a male subject.
  • The testosterone undecanoate compositions and oral dosage capsules disclosed herein can be used in conjunction with or as a component of a diagnostic or treatment kit that enables the diagnosis and treatment of a male patient in need of testosterone therapy. The diagnostic or treatment kit may comprise the testosterone undecanoate composition or oral dosage capsule disclosed herein along with one or more other components, including, but not limited to 1) instructions to enable those ordinarily skilled in the art to prepare a dosage form for immediate dispensing to the subject in need of; 2) one or more containers filled with one or more of the ingredients of the oral pharmaceutical dosage forms of the invention. Suitable containers include, for example, a bottle, a box, a blister card, a foil packet, or a combination thereof; 3) a tamper proof container or packaging; 4) other pharmaceutical dosage forms including other active agents including PDE-5 inhibitors and glucocorticosteroids; 5) Notice or printed instructions: in a form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of the manufacture, use, or sale for human administration to treat a condition that could be treated by oral testosterone therapy; 6) A “planner” for monitoring and tracking administration of the oral dosage forms; 7) Containers for storing and transporting the components of the kit; 8) total testosterone or free testosterone testing kits; 9) Sex Hormone binding globulin, SHBG, testing kits; 10) Body mass index testing materials to identify high risk patients; 11) tests for identifying patients with hypogonadism; 12) tests to assess testicular function or impotency; 13) test for bone mineral density/osteoporosis; 14) test for hair density 15) test for muscle mass and strength; 16) test for determining erectile dysfunction; 17) test for decreased libido; 18) test for fatigue, depression, mood disorders or irritability; 19) test for infertility; 20) test for prostate condition.
  • The oral dosage compositions and oral dosage capsules disclosed herein can be co-administered with other active agents in order to treat a target condition. One or more co-administered active agents can be admixed with the testosterone undecanoate containing compositions and/or oral dosage forms of the current invention. For example, phosphodiesterase type 5 (PDE-5) inhibitors, such as sildenafil citrate, tadalafil, vardenafil, avanafil, lodenafil, mirodenafil, udenafil, and the like, are used to block the degradative action of phosphodiesterase type 5 enzyme on cyclic GMP in the smooth muscle cells lining the blood vessels supplying the corpus cavernosum of the penis and are frequently used to treat erectile dysfunction. Such compounds could be co-administered with the compositions and oral dosage forms of the present invention in order to provide improved clinical outcomes through synergistic pharmacological action as measured by improved (sooner, better and longer lasting) erection, potency, libido, mood, body mass, etc. in males relative to administration of the testosterone or the co-administered PDE-5 alone. The testosterone undecanoate compositions and oral dosage capsules can also be co-administered with one or more other active agents such as aromatase inhibitors (for example letrozole, anastrozole, exemestane, fadrozole, vorozole, formestane etc.), dopamine agonists (for example apomorphine, bromocriptine, cabergoline, pergolide, ropinirole, rotigotine, pramipexole, fenoldopam etc.), prostaglandins (for example alprostadil), alpha blockers (for example yohimbine, phentolamine), vasodilators (for example minoxidil) and the like, for improved clinical outcomes through synergistic pharmacological action as measured by improvements in one or more of the secondary sexual characteristics in males such as sexual activity, potency, libido, erection etc., mood, body mass and the like, relative to administration of either the testosterone or the co-administered active agent alone.
  • In another aspect, the subjects receiving the dosage form of this invention are expected to improve in quality of life. The patient reported outcome may be employed to measure the improvement in other levels apart from primary (serum testosterone Cave) and secondary (serum testosterone Cmax, Cmin, Ctrough, etc) outcomes which are typically quantified by the PK profiles. For measuring the pharmacodynamic related efficacy outcomes, the improvements in the symptoms are usually monitored by ranking or scoring, dairy recording etc, by the subject being treated and/or the partner or spouse of the subject, in a timely manner before and during the therapy.
  • Accordingly, in one embodiment the dosage forms and the methods of current invention improve sexual symptoms including but not limited to sexual activity engagement, sexual thoughts or fantasies; feel of sexual desire; frequency of experience of morning erections; maintaining erections as long as desired; hardness of erection; ejaculation; enjoyment/satisfaction of sexual activity. In another embodiment the dosage form and the methods of current invention improves or enhances the physical and physiological symptoms and body energy level as assessed by the level of happiness with the body looks; body muscle mass, body weight and weakness/strength of muscles; level of tiredness; level of physical tiredness; level of energy; level of exhaustion and the like.
  • In another embodiment the dosage form and the methods of the current invention improves the symptoms related to the sleep symptoms and memory/cognition as assessed by quality of sleep at night; frequency of sleep restfulness; number of wake-up times during the night; frequency of feeling of satisfactory rest, frequency of accidental doze off during the day; frequency of purposely taken naps during the day; focus attention to tasks; level of forgetfulness; desire or ambition to take on new projects; short attention span; successful/efficient completion of tasks.
  • In a further aspect, the compositions of the current invention can be formulated to provide a gastro-retentive dosage form. In one embodiment, the gastro-retentive dosage form is a capsule. In another embodiment, the gastro-retentive dosage form is retained in the upper gastro-intestinal tract for at least one hour post-dosing. In another embodiment, the gastro-retentive dosage form is retained in the upper gastro-intestinal tract for at least two hours post-dosing. In another embodiment, the gastro-retentive dosage form is retained in the upper gastro-intestinal tract for at least 4 hours post-dosing. In another embodiment, the gastro-retentive dosage form is formulated to float in the stomach after dosing. In another embodiment, the gastro-retentive dosage form is formulated to expand when it comes in contact with aqueous medium to at least 1.3 times its size compared to its size when it is not in contact with the aqueous use environment. In another embodiment, the gastro-retentive dosage form is formulated to adhere to the lining of the stomach wall after dosing.
  • The compositions and the oral dosage capsules of the current invention can also include one or more of other additives selected from binders, bufferants, diluents, disintegrants, flavors, colorants, taste-masking agents, resins, pH modifiers, lubricants, glidants, thickening agent, opacifying agent, humectants, desiccants, effervescing agents, plasticizing agents and the like.
  • In addition to the compositions and oral dosage capsules of the present invention, a method for providing a serum concentration of testosterone within a target serum testosterone concentration Cave range for a male subject is also provided. It is noted that the compositions and oral dosage capsules of the present invention can be used in the conjunction with this method and that the teachings regarding the compositions and their administration provided above can be applied and used in connection with the methods disclosed here. The method includes the step of orally administering to the male subject an initial regimen including a daily dose of a testosterone undecanoate-containing composition. The testosterone undecanoate comprises about 14 wt % to about 35 wt % of the testosterone undecanoate-containing composition and the daily dose provides about 350 mg to about 650 mg of testosterone undecanoate to the male subject. After the initial regimen, the method includes a step of determining a dose titration metric based on a measurement of serum testosterone concentration for the male subject on at least one titration node day within the initial regimen. The method further includes the step of orally administering to the male subject a maintenance regimen including a daily dose of testosterone undecanoate-containing composition that comprises about 14 wt % to about 35 wt % of the testosterone undecanoate. The maintenance regimen provides a daily dose of testosterone undecanoate to the subject based on the titration metric determined on the at least one titration node day of the initial regimen and is sufficient to provide a serum testosterone plasma concentration that is closer to or within the target range.
  • In one embodiment, the method for providing a serum concentration of testosterone within a target serum testosterone concentration Cave range for a male subject can further include the steps of determining a dose titration metric based on a measurement of serum testosterone concentration for the male subject on at least one titration node day within the maintenance regimen. Following the determination of the titration metric the method includes the step of orally administering to the male subject a second maintenance regimen including a daily dose of testosterone undecanoate-containing composition, wherein the testosterone undecanoate-containing composition comprises about 14 wt % to about 35 wt % of the testosterone undecanoate-containing composition. The second maintenance regimen provides a daily dose of testosterone undecanoate to the subject based on the titration metric determined on the at least one titration node day of the maintenance regimen sufficient to provide a serum testosterone plasma concentration within the target range. Following the second maintenance regimen, the steps of determining the titration metric and administering an additional maintenance regimen can be repeated as needed in order to achieve the a serum testosterone concentration within the target range.
  • In the above method, the daily dose administered in the initial regimen can be the same dosage amount as the daily dose administered in the maintenance regimen. In one embodiment, the daily dose of the maintenance regimen can provide an amount of testosterone undecanoate that is about 45% to about 155% of that of the initial daily dose. In another embodiment, the daily dose of the maintenance regimen can provide an amount of testosterone undecanoate that is about 66% to about 133% of that of the daily dose in the initial regimen. In yet another embodiment, the daily dose of the maintenance regimen can provide an amount of testosterone undecanoate that is about 75% to about 125% of that of the daily dose in the initial regimen.
  • The determination step or steps of the above described methods can be any single day from day after day 15 of the initial regimen. In one embodiment, the titration node day can be any single day from day 15 to day 21 following the start of the initial regimen. In another embodiment, the titration node day can be any single day from day 21 to day 30 following the start of the initial regimen. In yet a further embodiment, the titration node day is any single day from day 31 to day 63 following the start of the initial regimen. In still a further embodiment, the titration node day can be any single day from day 64 to day 84 following the start of the initial regimen.
  • As discussed previously, subjects with whom the methods of the present invention can be used can be those in need of testosterone therapy. In one aspect, the male subject with whom the method is being used can have a serum testosterone Cavg of less than 300 ng/dL before beginning of the initial regimen. In another aspect, the male subject can have a serum testosterone Cavg of less than 200 ng/dL before beginning of the initial regimen. In yet another aspect, the male subject can have a serum testosterone Cavg of less than 300 ng/dL before beginning of the maintenance regimen. In yet a further aspect, the male subject can have a serum testosterone Cavg of less than 200 ng/dL before beginning of the maintenance regimen. Similarly, the target serum testosterone Cave range can vary depending on the subject and his particular needs and physiological parameters. In one embodiment, the target serum testosterone Cave range can be about 300 ng/dL to 1100 ng/dL and is achieved by the method on or after day 84 following the start of the initial regimen. In another embodiment, the target serum testosterone Cave range can be about 300 ng/dL to about 1100 ng/dL and is achieved by the method on or after day 120 following the start of the initial regimen. In yet a further embodiment, the target serum testosterone Cave range can be about 300 ng/dL to about 1100 ng/dL and can be achieved by the method on or after day 180 following the start of the initial regimen.
  • The above described method can provide desirable pharmacokinetic parameters based on administration to a group of subjects. In one embodiment, the method of the present invention can be such that the method can provide a serum testosterone Cavg in the range of 300 ng/dL to 1100 ng/dL in 75% or more of hypogonadal males after 84 days from the start of the initial regimen, based on a minimum group size of 24 hypogonadal males. In another embodiment, the method can provide a serum testosterone Cmax of 1500 ng/dL or less in less than or equal to 85% of hypogonadal males based on a minimum group size of 24 hypogonadal males. In yet a further embodiment, the method provides a serum testosterone Cmax in the range of 1800 ng/dL to 2500 ng/dL in about 5% or less of hypogonadal males after 84 days from the start of the initial regimen based on a minimum group size of 24 hypogonadal males. In yet a further embodiment, the method can provide a serum testosterone Cmax of 2500 ng/dL in about 1% or less of hypogonadal males after 84 days from the start of the initial regimen based on a minimum group size of 24 hypogonadal males. In one embodiment, the method can provide a steady state ratio of serum testosterone Cmax to Cave of 2.7 or less based on single subject administration.
  • The above disclosed methods provide for initial and maintenance regimens that include daily dose amounts that can be provided as twice-a-day administrations or divided into multi-dosage administrations. When a multi-dosage administration is utilized to provide the daily dose amount of testosterone undecanoate the dosages can be equal or unequal and can be administered with or without meals, depending of the designated regimen. In one aspect, when the dosages, whether twice-a-day or multi-time dosages, are administered with a meal or a snack the meal or snack can include about 15 g to about 60 g of fat. In one embodiment, the method provides for administration of the daily dose during the maintenance regimen as including twice-a-day administration of the testosterone undecanoate-containing composition in conjunction with meals. In one embodiment, the meal administered with the testosterone undecanoate-containing composition can have a total calorie content of about 350 and 1200 K calories with about 30% to about 60% of the calories in the meal being derived from fat. In another embodiment, the method can provide, at steady state, a dose-normalized serum testosterone Cmax of about 3×10−6 dL−1 or higher when administered to a male subject with meals daily in two divided doses.
  • Exemplary Formulations Embodiments
  • Provided in this section are formulations. In a preferred embodiment, the formulations provide 440 mg to 490 mg of testosterone undecanoate per day to an individual when administered twice-a-day as one or two unit dosage forms per administration (e.g., 2-4 unit dosage forms per day). For example, 440, 441, 442, 443, 444, 445, 446, 447, 448, 449, 450, 451, 452, 453, 454, 455, 456, 457, 458, 459 In another preferred embodiment, the formulations provide about 450 mg or about 474 mg of testosterone undecanoate per day to an individual when administered twice-a-day as one or two unit dosage forms per administration (e.g., 2-4 unit dosage forms per day). In the tables below the weight percent refers to the weight percent of the capsule fill composition.
  • TABLE 1
    Drug + Carriers
    Compositions (w/w %) Ratio of API:Carrier
    Testosterone in a pharmaceutical
    Composition undecanoate* Carrier composition
    A  8-10 80-92 1.0:9.0-1.0:11.5
    B 10-14 86-90 1.0:4.0-1.0:9.0
    C 15-20 80-85 1.0:4.0-1.0:5.7
    D 20-25 75-80 1.0:3.0-1.0:4.0
    E 25-50 50-75 1.0:1.0-1.0:3.0
    *As active pharmaceutical ingredient (API), it can be untreated, sieved (PS <450 micron), milled (PS <150 micron), micronized (1 micron < PS < 25 micron), or nanosized (PS <1 micron).
  • TABLE 2
    Carrier Components
    Carrier No.
    Component I II III IV V VI VII VIII IX X XI XII XIII
    Solubi- Propylene Y
    lizer glycol mono
    or di-laurate
    Propylene Y
    glycol mono
    or di-caprylate
    Corn glycerides Y
    (e.g. Glyceryl
    mono or di-
    linoleate)
    Vegetable Y
    glycerides
    (e.g. Glyceryl
    mono or di-
    oleate)
    Glyceryl Y
    mono or di-
    stearate
    Glyceryl Y
    palmito-
    stearate
    (9Z)-Octadec- Y
    9-enoic acid
    Octadecanoic Y
    acid
    (9Z,12Z)- Y
    9,12-Octa-
    decadienoic
    acid
    Peppermint oil Y
    Omega-3 Y
    EPA/DHA
    Vitamin E Y
    Combinations* Y
    Hydro- Cremophor, Y Y Y Y Y Y Y Y Y Y Y Y Y
    philic Tween, SLS,
    Add. Poloxamer,
    Polymer, and/or
    combinations
    Other Anti-oxidant, q.s. q.s. q.s. q.s. q.s. q.s. q.s. q.s. q.s. q.s. q.s. q.s. q.s.
    Add. solidifier,
    flow agent,
    solvent, and/or
    combinations
    *Combinations of solubilizers can be a combination of 2 or more solubilizers that are listed in this table as well as include propylene glycol, polyethylene glycol, glycerol, sorbitol, DMA, and so on.
    Add. = additive.
    Y = Yes.
  • Carrier I. Compositions composed of solubilizer (Propylene glycol mono or di-
    laurate), hydrophilic additives, and other additives for Composition A to E
    Carrier Compositions (w/w %)
    Hydrophilic
    additives1 (e.g. % Release in
    Cremophor2 RH 8% Triton
    Propylene 40, Tween3 80, Aqueous Media
    glycol SLS, Poloxamer4 Other additives7 ≥50% in 2
    Comp. mono or 407, Polymer5, and/ Anti- Solidify Combi- Total % hrs and ≥75%
    I No. di-laurate or combinations6) oxidant8 agent9 nations10 in Carrier in 4 hrs
    a  90-100 0-1 0-10 0-10 100 Yes
    b 45-99  1-40 0-1 0-15 0-15 100 Yes
     1-25 0-1 15-20  15-20  100 No
    c 85-99 1-4.5 0-1 0-10 0-10 100 Yes
    d 80-95  5-10 0-1 0-10 0-10 100 Yes
    e 70-90 10-20 0-1 0-10 0-10 100 Yes
    f 60-80 20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
    g 50-80 30-40 0-1 0-10 0-10 100 Yes
    20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
    • 1 Hydrophilic additives can be, but are not limited to ones listed in this table, e.g., hydrophilic surfactants having an HLB value of greater than 10, which are PEG-8 caprylic/capric glycerides (Labrasol), lauroyl macrogol-32 glyceride (Gelucire 44/14), stearoyl macrogol glyceride (Gelucire 50/13), sodium dioctyl sulfosuccinate, polyethylene glycol fatty acids mono- and di-ester mixtures, polyethylene glycol 1000 tocopherol succinate, phytosterols, phytosterol fatty acid esters, lanosterol PEG-24 cholesterol ether, PEG-30 soya sterol, PEG-25 phytosterol, PEG-30 cholestanol, and so on.
    • 2 Cremophor includes, but is not limited to, Cremophor RH 40, but Cremophor EL, RH 40, and RH 60.
    • 3 Tween includes, but is not limited to, Tween 80, but Tween 20, 60, and 80.
    • 4 Poloxamer includes, but is not limited to Poloxamer 407, but Poloxamer 124, 188, 234, 335, and 407.
    • 5 Polymer includes, but is not limited to, Polyethylene glycol, Hydroxypropyl cellulose, Hydroxypropylmethyl cellulose, Hydroxypropylmethyl cellulose acetate succinate, Polyvinylpyrrolidone, Polyvinyl acetate, Polylactic-co-glycolic acid, Polyvinyl caprolactame, Carbomer, and a combination thereof
    • 6 Combinations of hydrophilic additives can be 2 or more hydrophilic additives.
    • 7 Other additives can be, but are not limited to ones listed in this table, e.g., adsorbing agents, anti-adherents, anticoagulants, antifoaming agents, anti-caking agents, anti-static agents, binders, bile acids, bufferants, bulking agents, chelating agents, coagulants, colorants, opaquants, coolants, cryoprotectants, diluents, dehumidifying agents, desiccants, desensitizers, disintegrants, dispersing agents, enzyme inhibitors, fillers, hydrating agent, super disintegrants, gums, mucilages, hydrogen bonding agents, enzymes, flavorants, humectants, humidifying agents, ion-exchange resins, lubricants, plasticizers, pH modifying agents, preservatives, organic solvents, spreading agent, stabilizers, suspending agent, thickeners, viscosity increasing agents, waxes, and so on.
    • 8 Anti-oxidant can be, but is not limited to, ascorbyl palmitate, ascorbic acid, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), propyl gallate, cysteine, sodium metabisulfite (SMB), thiol derivatives, alpha-tocopherol, and so on.
    • 9 Solidifying (solidify) agent can be, but is not limited, to PEG 3350, PEG 4000, PEG 6000, PEG 8000, Poloxamer 188, Poloxamer 407, cetyl esters, wax, beeswax, glyceryl monostearate, glyceryl distearate, glyceryl palmitostearate, stearic acid, and so on.
    • 10 Combinations of other additives can be 2 or more other additives.
      • Descriptions for from 1 to 10 are applied to tables of all carrier compositions (from Carrier I to Carrier XIII tables) shown below.
  • Carrier II. Compositions composed of solubilizer (Propylene glycol mono or di-
    caprylate), hydrophilic additives, and other additives for Composition A to E
    Carrier Compositions (w/w %)
    Hydrophilic
    additives1 (e.g. % Release in
    Cremophor2 RH 8% Triton
    Propylene 40, Tween3 80, Aqueous Media
    glycol SLS, Poloxamer4 Other additives7 ≥50% in 2
    Comp. mono or 407, Polymer5, and/ Anti- Solidify Combi- Total % hrs and ≥75%
    II No. di-caprylate or combinations6) oxidant8 agent9 nations10 in Carrier in 4 hrs
    a  90-100 0-1 0-10 0-10 100 Yes
    b 45-99  1-40 0-1 0-15 0-15 100 Yes
     1-25 0-1 15-20  15-20  100 No
    c 85-99 1-4.5 0-1 0-10 0-10 100 Yes
    d 80-95  5-10 0-1 0-10 0-10 100 Yes
    e 70-90 10-20 0-1 0-10 0-10 100 Yes
    f 60-80 20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
    g 50-80 30-40 0-1 0-10 0-10 100 Yes
    20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
  • Carrier III. Compositions composed of solubilizer (Corn glycerides: e.g. Glyceryl mono
    or di-linoleate), hydrophilic additives, and other additives for Composition A to E
    Carrier Compositions (w/w %)
    Hydrophilic
    additives1 (e.g. % Release in
    Corn Cremophor2 RH 8% Triton
    glycerides 40, Tween3 80, Aqueous Media
    (e.g. Glyceryl SLS, Poloxamer4 Other additives7 ≥50% in 2
    Comp. mono or 407, Polymer5, and/ Anti- Solidify Combi- Total % hrs and ≥75%
    III No. di-linoleate) or combinations6) oxidant 8 agent9 nations10 in Carrier in 4 hrs
    a  90-100 0-1 0-10 0-10 100 Yes
    b 45-99  1-40 0-1 0-15 0-15 100 Yes
     1-25 0-1 15-20  15-20  100 No
    c 85-99 1-4.5 0-1 0-10 0-10 100 Yes
    d 80-95  5-10 0-1 0-10 0-10 100 Yes
    e 70-90 10-20 0-1 0-10 0-10 100 Yes
    f 60-80 20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
    g 50-80 30-40 0-1 0-10 0-10 100 Yes
    20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
  • Carrier IV. Compositions composed of solubilizer (Vegetable glycerides: e.g. Glyceryl
    mono or di-oleate), hydrophilic additives, and other additives for Composition A to E
    Carrier Compositions (w/w %)
    Hydrophilic
    additives1 (e.g. % Release in
    Vegetable Cremophor2 RH 8% Triton
    glycerides 40, Tween3 80, Aqueous Media
    (e.g. Glyceryl SLS, Poloxamer4 Other additives7 ≥50% in 2
    Comp. mono or 407, Polymer5, and/ Anti- Solidify Combi- Total % hrs and ≥75%
    IV No. di-oleate) or combinations6) oxidant 8 agent9 nations10 in Carrier in 4 hrs
    a  90-100 0-1 0-10 0-10 100 Yes
    b 45-99  1-40 0-1 0-15 0-15 100 Yes
     1-25 0-1 15-20  15-20  100 No
    c 85-99 1-4.5 0-1 0-10 0-10 100 Yes
    d 80-95  5-10 0-1 0-10 0-10 100 Yes
    e 70-90 10-20 0-1 0-10 0-10 100 Yes
    f 60-80 20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
    g 50-80 30-40 0-1 0-10 0-10 100 Yes
    20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
  • Carrier V. Compositions composed of solubilizer (Glyceryl mono or di-stearate),
    hydrophilic additives, and other additives for Composition A to E
    Carrier Compositions (w/w %)
    Hydrophilic
    additives1 (e.g. % Release in
    Cremophor2 RH 8% Triton
    40, Tween3 80, Aqueous Media
    Glyceryl SLS, Poloxamer4 Other additives7 ≥50% in 2
    Comp. mono or 407, Polymer5, and/ Anti- Solidify Combi- Total % hrs and ≥75%
    V No. di-stearate or combinations6) oxidant 8 agent9 nations10 in Carrier in 4 hrs
    a  90-100 0-1 0-10 0-10 100 No
    b 45-99  1-40 0-1 0-15 0-15 100 No
     1-25 0-1 15-20  15-20  100 No
    c 85-99 1-4.5 0-1 0-10 0-10 100 No
    d 80-95  5-10 0-1 0-10 0-10 100 No
    e 70-90 10-20 0-1 0-10 0-10 100 No
    f 60-80 20-30 0-1 0-10 0-10 100 No
    10-20 0-1 10-20  10-20  100 No
    g 50-80 30-40 0-1 0-10 0-10 100 No
    20-30 0-1 0-10 0-10 100 No
    10-20 0-1 10-20  10-20  100 No
  • Carrier VI. Compositions composed of solubilizer (Glyceryl palmito-stearate),
    hydrophilic additives, and other additives for Composition A to E
    Carrier Compositions (w/w %)
    Hydrophilic
    additives1 (e.g. % Release in
    Cremophor2 RH 8% Triton
    40, Tween3 80, Aqueous Media
    Glyceryl SLS, Poloxamer4 Other additives7 ≥50% in 2
    Comp. palmito- 407, Polymer5, and/ Anti- Solidify Combi- Total % hrs and ≥75%
    VI No. stearate or combinations6) oxidant 8 agent9 nations10 in Carrier in 4 hrs
    a  90-100 0-1 0-10 0-10 100 No
    b 45-99  1-40 0-1 0-15 0-15 100 No
     1-25 0-1 15-20  15-20  100 No
    c 85-99 1-4.5 0-1 0-10 0-10 100 No
    d 80-95  5-10 0-1 0-10 0-10 100 No
    e 70-90 10-20 0-1 0-10 0-10 100 No
    f 60-80 20-30 0-1 0-10 0-10 100 No
    10-20 0-1 10-20  10-20  100 No
    g 50-80 30-40 0-1 0-10 0-10 100 No
    20-30 0-1 0-10 0-10 100 No
    10-20 0-1 10-20  10-20  100 No
  • Carrier VII. Compositions composed of solubilizer ((9Z)-Octadec-9-enoic
    acid), hydrophilic additives, and other additives for Composition A to E
    Carrier Compositions (w/w %)
    Hydrophilic
    additives1 (e.g. % Release in
    Cremophor2 RH 8% Triton
    40, Tween3 80, Aqueous Media
    SLS, Poloxamer4 Other additives7 ≥50% in 2
    Comp. (9Z)-Octadec- 407, Polymer5, and/ Anti- Solidify Combi- Total % hrs and ≥75%
    VII No. 9-enoic acid or combinations6) oxidant 8 agent9 nations10 in Carrier in 4 hrs
    a  90-100 0-1 0-10 0-10 100 Yes
    b 45-99  1-40 0-1 0-15 0-15 100 Yes
     1-25 0-1 15-20  15-20  100 No
    c 85-99 1-4.5 0-1 0-10 0-10 100 Yes
    d 80-95  5-10 0-1 0-10 0-10 100 Yes
    e 70-90 10-20 0-1 0-10 0-10 100 Yes
    f 60-80 20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
    g 50-80 30-40 0-1 0-10 0-10 100 Yes
    20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
  • Carrier VIII. Compositions composed of solubilizer (octadecanoic acid),
    hydrophilic additives, and other additives for Composition A to E
    Carrier Compositions (w/w %)
    Hydrophilic
    additives1 (e.g. % Release in
    Cremophor2 RH 8% Triton
    40, Tween3 80, Aqueous Media
    SLS, Poloxamer4 Other additives7 ≥50% in 2
    Comp. Octadec- 407, Polymer5, and/ Anti- Solidify Combi- Total % hrs and ≥75%
    VIII No. anoic acid or combinations6) oxidant 8 agent9 nations10 in Carrier in 4 hrs
    a  90-100 0-1 0-10 0-10 100 No
    b 45-99  1-40 0-1 0-15 0-15 100 No
     1-25 0-1 15-20  15-20  100 No
    c 85-99 1-4.5 0-1 0-10 0-10 100 No
    d 80-95  5-10 0-1 0-10 0-10 100 No
    e 70-90 10-20 0-1 0-10 0-10 100 No
    f 60-80 20-30 0-1 0-10 0-10 100 No
    10-20 0-1 10-20  10-20  100 No
    g 50-80 30-40 0-1 0-10 0-10 100 No
    20-30 0-1 0-10 0-10 100 No
    10-20 0-1 10-20  10-20  100 No
  • Carrier IX. Compositions composed of solubilizer ((9Z,12Z)-9,12-Octadecadienoic
    acid), hydrophilic additives, and other additives for Composition A to E
    Carrier Compositions (w/w %)
    Hydrophilic
    additives1 (e.g. % Release in
    Cremophor2 RH 8% Triton
    40, Tween3 80, Aqueous Media
    (9Z,12Z)-9,12- SLS, Poloxamer4 Other additives7 ≥50% in 2
    Comp. Octadeca- 407, Polymer5, and/ Anti- Solidify Combi- Total % hrs and ≥75%
    IX No. dienoic acid or combinations6) oxidant 8 agent9 nations10 in Carrier in 4 hrs
    a  90-100 0-1 0-10 0-10 100 Yes
    b 45-99  1-40 0-1 0-15 0-15 100 Yes
     1-25 0-1 15-20  15-20  100 No
    c 85-99 1-4.5 0-1 0-10 0-10 100 Yes
    d 80-95  5-10 0-1 0-10 0-10 100 Yes
    e 70-90 10-20 0-1 0-10 0-10 100 Yes
    f 60-80 20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
    g 50-80 30-40 0-1 0-10 0-10 100 Yes
    20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
  • Carrier X. Compositions composed of solubilizer (Peppermint oil),
    hydrophilic additives, and other additives for Composition A to E
    Carrier Compositions (w/w %)
    Hydrophilic
    additives1 (e.g. % Release in
    Cremophor2 RH 8% Triton
    40, Tween3 80, Aqueous Media
    SLS, Poloxamer4 Other additives7 ≥50% in 2
    Comp. Pepper- 407, Polymer5, and/ Anti- Solidify Combi- Total % hrs and ≥75%
    X No. mint oil or combinations6) oxidant 8 agent9 nations10 in Carrier in 4 hrs
    a  90-100 0-1 0-10 0-10 100 Yes
    b 45-99  1-40 0-1 0-15 0-15 100 Yes
     1-25 0-1 15-20  15-20  100 Yes
    c 85-99 1-4.5 0-1 0-10 0-10 100 Yes
    d 80-95  5-10 0-1 0-10 0-10 100 Yes
    e 70-90 10-20 0-1 0-10 0-10 100 Yes
    f 60-80 20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 Yes
    g 50-80 30-40 0-1 0-10 0-10 100 Yes
    20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 Yes
  • Carrier XI. Compositions composed of solubilizer (Omega-3 EPA/DHA),
    hydrophilic additives, and other additives for Composition A to E
    Carrier Compositions (w/w %)
    Hydrophilic
    additives1 (e.g. % Release in
    Cremophor2 RH 8% Triton
    40, Tween3 80, Aqueous Media
    SLS, Poloxamer4 Other additives7 ≥50% in 2
    Comp. Omega-3 407, Polymer5, and/ Anti- Solidify Combi- Total % hrs and ≥75%
    XI No. EPA/DHA or combinations6) oxidant 8 agent9 nations10 in Carrier in 4 hrs
    a  90-100 0-1 0-10 0-10 100 Yes
    b 45-99  1-40 0-1 0-15 0-15 100 Yes
     1-25 0-1 15-20  15-20  100 No
    c 85-99 1-4.5 0-1 0-10 0-10 100 Yes
    d 80-95  5-10 0-1 0-10 0-10 100 Yes
    e 70-90 10-20 0-1 0-10 0-10 100 Yes
    f 60-80 20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
    g 50-80 30-40 0-1 0-10 0-10 100 Yes
    20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
  • Carrier XII. Compositions composed of solubilizer (Vitamin E), hydrophilic
    additives, and other additives for Composition A to E
    Carrier Compositions (w/w %)
    Hydrophilic
    additives1 (e.g. % Release in
    Cremophor2 RH 8% Triton
    40, Tween3 80, Aqueous Media
    SLS, Poloxamer4 Other additives7 ≥50% in 2
    Comp. 407, Polymer5, and/ Anti- Solidify Combi- Total % hrs and ≥75%
    XII No. Vitamin E or combinations6) oxidant 8 agent9 nations10 in Carrier in 4 hrs
    a  90-100 0-1 0-10 0-10 100 Yes
    b 45-99  1-40 0-1 0-15 0-15 100 Yes
     1-25 0-1 15-20  15-20  100 Yes
    c 85-99 1-4.5 0-1 0-10 0-10 100 Yes
    d 80-95  5-10 0-1 0-10 0-10 100 Yes
    e 70-90 10-20 0-1 0-10 0-10 100 Yes
    f 60-80 20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 Yes
    g 50-80 30-40 0-1 0-10 0-10 100 Yes
    20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 Yes
  • Carrier XIII. Compositions composed of a combination of solubilizers,
    hydrophilic additives, and other additives for Composition A to E
    Carrier Compositions (w/w %)
    Combination
    of solubilizers
    (e.g. oleic Hydrophilic
    acid and GDS, additives1 (e.g. % Release in
    oleic acid and Cremophor2 RH 8% Triton
    peppermint 40, Tween3 80, Aqueous Media
    oil, maisine SLS, Poloxamer4 Other additives7 ≥50% in 2
    Comp. 35-1 and 407, Polymer5, and/ Anti- Solidifying Combi- Total % hrs and ≥75%
    XIII No. GMC, etc) or combinations6) oxidant 8 agent9 nations10 in Carrier in 4 hrs
    a  90-100 0-1 0-10 0-10 100 Yes
    b 45-99  1-40 0-1 0-15 0-15 100 Yes
     1-25 0-1 15-20  15-20  100 No
    c 85-99 1-4.5 0-1 0-10 0-10 100 Yes
    d 80-95  5-10 0-1 0-10 0-10 100 Yes
    e 70-90 10-20 0-1 0-10 0-10 100 Yes
    f 60-80 20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
    g 50-80 30-40 0-1 0-10 0-10 100 Yes
    20-30 0-1 0-10 0-10 100 Yes
    10-20 0-1 10-20  10-20  100 No
    Combination of solubilizers includes, not limited to oleic acid and glyceryl distearate, oleic acid and peppermint oil, or maisine 35-1 and glyceryl monocaprylate, but 2 or more solubilizers that are listed in Table 2.
  • Additional exemplary formulations are below. In a preferred embodiment, the formulations provide 440 mg to 490 mg of testosterone undecanoate per day to an individual when administered twice-a-day as one or two unit dosage forms per administration (e.g., 2-4 unit dosage forms per day). In another preferred embodiment, the formulations provide about 450 mg or about 474 mg of testosterone undecanoate per day to an individual when administered twice-a-day as one or two unit dosage forms per administration (e.g., 2-4 unit dosage forms per day). In the tables below the weight percent refers to the weight percent of the capsule fill composition.
  • % Testosterone
    Undecanoate
    (w/w) Solubilizer (w/w) Dispersant (w/w) Additive
    5 20-80% Monoglyceride 0-20% Polyoxyl Quantum
    Hydrogenated satis (q.s.)
    Vegetable Oil
    6 20-80% Monoglyceride 0-20% Polyoxyl q.s.
    Hydrogenated
    Vegetable Oil
    7 20-80% Monoglyceride 0-20% Polyoxyl q.s.
    Hydrogenated
    Vegetable Oil
    8 20-80% Monoglyceride 0-20% Polyoxyl q.s.
    Hydrogenated
    Vegetable Oil
    9 20-80% Monoglyceride 0-20% Polyoxyl q.s.
    Hydrogenated
    Vegetable Oil
    10 20-80% Monoglyceride 0-20% Polyoxyl q.s.
    Hydrogenated
    Vegetable Oil
    11 20-80% Monoglyceride 0-20% Polyoxyl q.s.
    Hydrogenated
    Vegetable Oil
    12 20-80% Monoglyceride 0-20% Polyoxyl q.s.
    Hydrogenated
    Vegetable Oil
    13 20-80% Monoglyceride 0-20% Polyoxyl q.s.
    Hydrogenated
    Vegetable Oil
    14 20-80% Monoglyceride 0-20% Polyoxyl q.s.
    Hydrogenated
    Vegetable Oil
    15 20-80% Monoglyceride Polyoxyl q.s.
    Hydrogenated
    Vegetable Oil
    16 20-80% Monoglyceride 0-20% Polyoxyl q.s.
    Hydrogenated
    Vegetable Oil
    17 20-80% Monoglyceride 0-20% Polyoxyl q.s.
    Hydrogenated
    Vegetable Oil
  • In one aspect, in the table above, the monoglyceride can be glyceryl monolinoleate. In another aspect, the monoglyceride can be glyceryl mono-oleate. In one aspect, the polyoxyl hydrogenated vegetable oil is a polyoxyl hydrogenated castor oil. In another aspect, polyoxyl hydrogenated vegetable oil is polyoxyl 40 hydrogenated castor oil. In another aspect, polyoxyl hydrogenated vegetable oil is polyoxyl 35 hydrogenated castor oil. In one aspect the additive can be chosen from adsorbing agents, anti-adherents, anticoagulants, antifoaming agents, anti-caking agents, anti-static agents, binders, bile acids, bufferants, bulking agents, chelating agents, coagulants, colorants, opaquants, coolants, cryoprotectants, diluents, dehumidifying agents, desiccants, desensitizers, disintegrants, dispersing agents, enzyme inhibitors, fillers, hydrating agent, super disintegrants, gums, mucilages, hydrogen bonding agents, enzymes, flavorants, humectants, humidifying agents, ion-exchange resins, lubricants, plasticizers, pH modifying agents, preservatives, organic solvents, spreading agent, stabilizers, suspending agent, thickeners, viscosity increasing agents, waxes, and mixtures thereof.
  • %
    Testosterone
    Undecanoate
    (w/w) Solubilizer (w/w) Dispersant (w/w) Additive
    5 20-80% 0-20% Polyoxyl q.s.
    Monoglyceride/Diglyceride Hydrogenated
    Vegetable Oil
    6 20-80% 0-20% Polyoxyl q.s.
    Monoglyceride/Diglyceride Hydrogenated
    Vegetable Oil
    7 20-80% 0-20% Polyoxyl q.s.
    Monoglyceride/Diglyceride Hydrogenated
    Vegetable Oil
    8 20-80% 0-20% Polyoxyl q.s.
    Monoglyceride/Diglyceride Hydrogenated
    Vegetable Oil
    9 20-80% 0-20% Polyoxyl q.s.
    Monoglyceride/Diglyceride Hydrogenated
    Vegetable Oil
    10 20-80% 0-20% Polyoxyl q.s.
    Monoglyceride/Diglyceride Hydrogenated
    Vegetable Oil
    11 20-80% 0-20% Polyoxyl q.s.
    Monoglyceride/Diglyceride Hydrogenated
    Vegetable Oil
    12 20-80% 0-20% Polyoxyl q.s.
    Monoglyceride/Diglyceride Hydrogenated
    Vegetable Oil
    13 20-80% 0-20% Polyoxyl q.s.
    Monoglyceride/Diglyceride Hydrogenated
    Vegetable Oil
    14 20-80% 0-20% Polyoxyl q.s.
    Monoglyceride/Diglyceride Hydrogenated
    Vegetable Oil
    15 20-80% 0-20% Polyoxyl q.s.
    Monoglyceride/Diglyceride Hydrogenated
    Vegetable Oil
    16 20-80% 0-20% Polyoxyl q.s.
    Monoglyceride/Diglyceride Hydrogenated
    Vegetable Oil
    17 20-80% 0-20% Polyoxyl q.s.
    Monoglyceride/Diglyceride Hydrogenated
    Vegetable Oil
  • In one aspect, in the table above, the solubilizer can be glyceryl monolinoleate. In another aspect, the solubilizer can be glyceryl mono-oleate. In another aspect, the solubilizer can be glyceryl palmitostearate. In one aspect, the solubilizer is a mono-diglyceride of medium chain fatty acids (e.g., caprylic and capric) In one aspect, the polyoxyl hydrogenated vegetable oil is a polyoxyl hydrogenated castor oil. In another aspect, the polyoxyl hydrogenated vegetable oil is polyoxyl 40 hydrogenated castor oil. In another aspect, the polyoxyl hydrogenated vegetable oil is polyoxyl 35 hydrogenated castor oil. In one aspect the additive can be chosen from adsorbing agents, anti-adherents, anticoagulants, antifoaming agents, anti-caking agents, anti-static agents, binders, bile acids, bufferants, bulking agents, chelating agents, coagulants, colorants, opaquants, coolants, cryoprotectants, diluents, dehumidifying agents, desiccants, desensitizers, disintegrants, dispersing agents, enzyme inhibitors, fillers, hydrating agent, super disintegrants, gums, mucilages, hydrogen bonding agents, enzymes, flavorants, humectants, humidifying agents, ion-exchange resins, lubricants, plasticizers, pH modifying agents, preservatives, organic solvents, spreading agent, stabilizers, suspending agent, thickeners, viscosity increasing agents, waxes, and mixtures thereof.
  • % Testosterone
    Undecanoate (w/w) Solubilizer (w/w) Dispersant (w/w) Additive
    5 20-80% 0-20% q.s.
    6 20-80% 0-20% q.s.
    7 20-80% 0-20% q.s.
    8 20-80% 0-20% q.s.
    9 20-80% 0-20% q.s.
    10 20-80% 0-20% q.s.
    11 20-80% 0-20% q.s.
    12 20-80% 0-20% q.s.
    13 20-80% 0-20% q.s.
    14 20-80% 0-20% q.s.
    15 20-80% 0-20% q.s.
    16 20-80% 0-20% q.s.
    17 20-80% 0-20% q.s.
  • In one aspect, in the table above, the solubilizer can be glyceryl monolinoleate. In another aspect, the solubilizer can be glyceryl mono-oleate. In one aspect, the solubilizer is a tocopherol. In another aspect, the solubilizer can be glyceryl palmitostearate. In one aspect, the solubilizer is chosen from any one or more of those described herein. In one aspect, the dispersant is chosen from any one of those described herein. In one aspect, the dispersant is a polyoxyl hydrogenated vegetable oil. In one aspect, the polyoxyl hydrogenated vegetable oil is a polyoxyl hydrogenated castor oil. In another aspect, the polyoxyl hydrogenated vegetable oil is polyoxyl 40 hydrogenated castor oil. In another aspect, the polyoxyl hydrogenated vegetable oil is polyoxyl 35 hydrogenated castor oil. In one aspect the additive can be chosen from adsorbing agents, anti-adherents, anticoagulants, antifoaming agents, anti-caking agents, anti-static agents, binders, bile acids, bufferants, bulking agents, chelating agents, coagulants, colorants, opaquants, coolants, cryoprotectants, diluents, dehumidifying agents, desiccants, desensitizers, disintegrants, dispersing agents, enzyme inhibitors, fillers, hydrating agent, super disintegrants, gums, mucilages, hydrogen bonding agents, enzymes, flavorants, humectants, humidifying agents, ion-exchange resins, lubricants, plasticizers, pH modifying agents, preservatives, organic solvents, spreading agent, stabilizers, suspending agent, thickeners, viscosity increasing agents, waxes, and mixtures thereof.
  • % Testosterone
    Undecanoate
    (w/w) Solubilizer (w/w) Dispersant (w/w) Additive
    5 20-80% Lipophilic 0-20% Hydrophilic q.s.
    Surfactant Surfactant
    6 20-80% Lipophilic 0-20% Hydrophilic q.s.
    Surfactant Surfactant
    7 20-80% Lipophilic 0-20% Hydrophilic q.s.
    Surfactant Surfactant
    8 20-80% Lipophilic 0-20% Hydrophilic q.s.
    Surfactant Surfactant
    9 20-80% Lipophilic 0-20% Hydrophilic q.s.
    Surfactant Surfactant
    10 20-80% Lipophilic 0-20% Hydrophilic q.s.
    Surfactant Surfactant
    11 20-80% Lipophilic 0-20% Hydrophilic q.s.
    Surfactant Surfactant
    12 20-80% Lipophilic 0-20% Hydrophilic q.s.
    Surfactant Surfactant
    13 20-80% Lipophilic 0-20% Hydrophilic q.s.
    Surfactant Surfactant
    14 20-80% Lipophilic 0-20% Polyoxyl q.s.
    Surfactant Hydrogenated
    Vegetable Oil
    15 20-80% Lipophilic 0-20% Hydrophilic q.s.
    Surfactant Surfactant
    16 20-80% Lipophilic 0-20% Hydrophilic q.s.
    Surfactant Surfactant
    17 20-80% Lipophilic 0-20% Hydrophilic q.s.
    Surfactant Surfactant
  • In one aspect, in the table above, the solubilizer can be glyceryl monolinoleate. In another aspect, the solubilizer can be glyceryl mono-oleate. In another aspect, the solubilizer can be glyceryl palmitostearate. In one aspect, the lipophilic surfactant is chosen from any one or more of those described therein. In one aspect, the hydrophilic surfactant is an ionic hydrophilic surfactant. In one aspect, the hydrophilic surfactant is a non-ionic hydrophilic surfactant. In one aspect, the hydrophilic surfactant is a polyoxyl hydrogenated vegetable oil. In one aspect, the polyoxyl hydrogenated vegetable oil is a polyoxyl hydrogenated castor oil. In another aspect, the polyoxyl hydrogenated vegetable oil is polyoxyl 40 hydrogenated castor oil. In another aspect, the polyoxyl hydrogenated vegetable oil is polyoxyl 35 hydrogenated castor oil. In one aspect, the hydrophilic surfactant is any one of those described herein. In one aspect the additive can be chosen from adsorbing agents, anti-adherents, anticoagulants, antifoaming agents, anti-caking agents, anti-static agents, binders, bile acids, bufferants, bulking agents, chelating agents, coagulants, colorants, opaquants, coolants, cryoprotectants, diluents, dehumidifying agents, desiccants, desensitizers, disintegrants, dispersing agents, enzyme inhibitors, fillers, hydrating agent, super disintegrants, gums, mucilages, hydrogen bonding agents, enzymes, flavorants, humectants, humidifying agents, ion-exchange resins, lubricants, plasticizers, pH modifying agents, preservatives, organic solvents, spreading agent, stabilizers, suspending agent, thickeners, viscosity increasing agents, waxes, and mixtures thereof.
  • In one aspect, the compositions in the Tables above releases (a) at least 80% or more at 1 or 2 hours; (b) less than 95% or 90% at 0.25 hours; (c) about 100% at 4, 3, 2, 1, 0.5, or 0.25 hours or (d) a combination of one, two, or three of (a)-(c) when tested using USP type 2 apparatus in about e.g., 1000 mL 8% Triton X100 solution in water at a specific temperature (e.g., 37° C.) at 100 rpm. In one aspect, the composition of this embodiment releases (a) at least 75% or more at 2 hours and (b) less than 95% or 90% at 0.25 hours. In one aspect, the compositions in the Tables above have carrier components chosen so as to have a release profile as described in this paragraph. In one aspect, the active pharmaceutical ingredient is fully solubilized in the composition. In another aspect, the active pharmaceutical ingredient is not fully solubilized as described herein.
  • EXAMPLES
  • The following examples are provided to promote a more clear understanding of certain embodiments of the present invention, and are in no way meant as a limitation thereon.
  • Example 1—Testosterone Undecanoate Composition
  • A testosterone undecanoate containing composition was prepared by using the components set forth in Table I. The composition is prepared by weighing all of the components, except the testosterone undecanoate, into a clean stainless steel container and mixed together at about 50° C. to about 70° C., using a stirrer. The testosterone undecanoate (TU) is added and stirred into the mixture of other components until the testosterone undecanoate dissolves. A predetermined quantity of this fill material is disposed into a capsule (for example, hard gelatin capsule) to get the required testosterone undecanoate dose per dosage unit. The capsules are allowed to cool at room temperature, banded (if required) and packaged in a HDPE bottle and tightly closed with an appropriate lid.
  • TABLE I
    Example 1 Composition mg/capsule
    Testosterone Undecanoate 200
    Solubilizer (e.g. Glycerides of coconut 725
    oil; Capmul ® MCM)
    Dispersant (e.g. lauroglycol) 300
    Dispersant (polyoxyl 40 hydrogenated 50
    castor oil or Cremophor ® RH40)
    TU-loading (wt %) of capsule fill = 15.7%
  • Example 2—Testosterone Undecanoate Composition
  • A testosterone undecanoate containing composition was prepared similarly to the method described in Example 1 using the components set forth in Table II.
  • TABLE II
    Example 2 Composition mg/capsule
    Testosterone Undecanoate 225
    Solubilizer (e.g. Maize oil glyceride) 260
    Dispersant (e.g. lauroglycol) 665
    TU-loading (wt %) of capsule fill ~19.6%
  • Examples 3 & 4—Testosterone Undecanoate Composition
  • Testosterone undecanoate containing composition were prepared similarly to the method described in Example 1 using the components set forth in Tables III and IV
  • TABLE III
    Example 3 Composition mg/capsule
    Testosterone Undecanoate 200
    Solubilizer (e.g. Glycerides of coconut 600
    oil; Capmul ® MCM)
    TU-loading (wt %) of capsule fill = 25%
  • TABLE IV
    Example 4 Composition mg/capsule
    Testosterone Undecanoate 180
    Solubilizer (Maize oil glyceride, Maisine 600
    35-1)
    TU-loading (wt %) of capsule fill = 23%
  • Example 5—Testosterone Undecanoate Composition
  • A testosterone undecanoate containing composition was prepared similarly to the method described in Example 1 using the components set forth in Table V
  • TABLE V
    Example 5 Composition mg/capsule
    Testosterone Undecanoate 240
    Solubilizer (e.g. Glycerides of 200
    coconut oil; Capmul ® MCM)
    Solubilizer (e.g. α-tocopherol) 490
    Dispersant (for e.g. polyoxyl castor 100
    oil or Cremophor ® EL)
    TU-Loading per caps TU-loading (wt %) of capsule fill = 23.3%
  • Example 6—Testosterone Undecanoate Composition
  • A testosterone undecanoate containing composition was prepared by using the components set forth in Table VI and a method similar to that described in Example 1.
  • TABLE VI
    Example 6 Composition mg/capsule
    Testosterone Undecanoate 200
    Solubilizer (e.g. Maize oil 490
    glycerides)
    Dispersant (e.g. polysorbate 80) 25
    Solidifying agent (e.g. polyethylene 45
    glycol, 8000 or PEG 8000)
    TU-Loading per capsule = 26.3%
  • Example 7—Testosterone Undecanoate Composition
  • A testosterone undecanoate containing composition was prepared by using the components set forth in Table VII and a method similar to that described in Example 1.
  • TABLE VII
    Example 7 Composition mg/capsule
    Testosterone Undecanoate 240
    Solubilizer (e.g. Maize oil glycerides) 325
    Solubilizer (e.g. oleic acid) 125
    Solubilizer (e.g. Benzyl Alcohol) 50
    Solubilizer (e.g. α-tocopherol) 75
    Solidifying agent (e.g. PEG 8000) 45
    TU- Loading per capsule = 28%
  • Example 8—Testosterone Undecanoate Composition
  • A testosterone undecanoate containing composition was prepared by using the components set forth in Table VIII and a method similar to that described in Example 1.
  • TABLE VIII
    Example 6 Composition mg/capsule
    Testosterone Undecanoate 240
    Solubilizer (e.g. oleic acid) 400
    Solidifying agent- (e.g. PEG 8000) 45
    TU- loading (wt %) of capsule fill = 35%
  • Example 9—Testosterone Undecanoate Composition
  • A testosterone undecanoate containing composition was prepared using the components set forth in Table IX and a method similar to that described in Example 1.
  • TABLE IX
    Example 7 Composition mg/capsule
    Testosterone Undecanoate 240
    Solubilizer (e.g. Maize oil glycerides) 400
    Solubilizer (e.g. α-tocopherol) 24
    Solidifying agent -(e.g. Glyceryl distearate; 25
    Percirol ® ATO 5)
    TU- loading (wt %) of capsule fill = 34.8%
  • Example 10—Testosterone Undecanoate Composition
  • A testosterone undecanoate containing composition can be prepared by using the components set forth in Table X by a method as follows: The required quantity of the glyceryl distearate or glyceryl monostearate and the PEG 8000 are placed in a stainless steel container and heated to about 50 to 70° C. to get a molten mixture. The testosterone undecanoate is added and stirred till it completely dissolves. A predetermined weight of the molten mixture is disposed into capsules and allowed to congeal at room temperature, banded and packed.
  • TABLE X
    Example 10 Composition mg/capsule
    Testosterone Undecanoate 100
    Glyceryl distearate (Percirol ® ATO 5) or 200
    glyceryl monostearate
    PEG 8000 50
  • The oral dosage capsules of Example 10, which contains non-dissolved TU, can provide, upon single administration along with food to a human male, a testosterone undecanoate AUC that is about 20% higher as compared to a composition that does not include the glyceryl distearate (Percirol® ATO 5) or glyceryl monostearate.
  • The composition of Example 10 can also be optionally modified so that a dispersant such as a disintegrating agent (e.g. Crospovidone at about 150 mg for every 100 mg TU dose) can be uniformly suspended under stirring in the molten testosterone undecanoate solution. This suspension can be further allowed to cooled and passed through ASTM 30 mesh get granulates or particulates which can be either filled in a capsule or compressed to a tablet.
  • Example 11—Testosterone Undecanoate Composition
  • A testosterone undecanoate containing composition wherein at least 50% of the testosterone undecanoate is dissolved is prepared by using the components set forth in Table XI and a method similar to that described under Example 1.
  • TABLE XI
    Example 11 Composition mg/capsule
    Testosterone Undecanoate 225
    Solubilizer (e.g. Castor Oil) 350
    Dispersant (e.g. lauroglycol); 180
    Solidifying agent (e.g. PEG 8000) 45
    TU- loading (wt %) of capsule fill = 28.1%
  • Examples 12-19—Testosterone Undecanoate Compositions
  • Testosterone undecanoate formulations of Examples 12 through 19 were prepared by using the components set forth in Table XII and by a method similar to that described under Example 1. Additionally, indicated amounts of the respective formulations were filled into hard gelatin capsules and the testosterone undecanoate release from capsules is measured using a USP Type-II apparatus at about 100 rpm in about 1000 mL of 8% w/w solution of Triton X100 in water, maintained at about 37° C. The results of the release testing are also shown in Table XII.
  • TABLE XII
    Composition mg/capsule
    Capsule Components/ Example Example Example Example Example Example Example Example Example
    Attributes 12 13 14 15 15A 16 17 18 19
    Testosterone 40  40  75 75 90 75 75 75 125
    Undecanoate
    Oleic Acid 227
    Castor oil 175 455 
    Lauroglycol 115
    Labrafil M2125CS 80
    Maize oil glycerides 455 455  315  316  316  515
    (Maisine 35-1)
    Polyoxyl 40 130 80 35 79 54 112
    Hydrogenated Castor
    Oil, (Cremophor
    RH40)
    Glyceryl distearate 50 60 25
    (Percirol ATO 5)
    Polyethylene Glycol 48 30 30  48
    8000,
    Total mg per capsule 267 330 660 660  500  660  500  500  800
    (mg)
    TU- loading (wt %) of  15%  12%  11.3% 11.3% 18% 11.3% 15% 15% 15.6%
    capsule fill
    TU released in 30 ~100% ~100% ~100% 30% 30%  ~78% 85% 32% 80%
    minutes (%)
    Time for 75% TU <120 <120  <120  <120  <120  <120  <120  <120  <120 
    release (minutes)
  • Examples 20-25—Testosterone Undecanoate Compositions
  • Testosterone undecanoate formulations of Examples 20 through 25 were prepared by using the components set forth in Table XIII and by a method similar to that described under Example 1. Additionally, indicated amounts of the respective formulations were filled into hard gelatin capsules and the testosterone undecanoate release from capsules was tested in about 1000 mL of 8% w/w solution of Triton X100 in water, maintained at about 37° C., using a USP Type-II apparatus at about 100 rpm. The results of the release testing are also shown in Table XIII.
  • TABLE XIII
    Composition mg/capsule
    Capsule Components/ Example Example Example Example Example Example
    Attributes 20 21 22 23 24 25
    Testosterone 200 200 240 240 240 240
    Undecanoate (TU)
    Maize oil glycerides 490 464 464 304
    (Maisine 35-1)
    Coconut oil glycerides 400
    (Capmul MCM)
    Alpha- tocopherol 510 50 50
    Benzyl alcohol 25 50
    Polyoxyl 40 25
    Hydrogenated Castor
    Oil,
    Polyoxyl 35 Castor Oil, 45
    Polyethylene Glycol 45 45 46 65 41
    8000, USP
    Total Fill wt. per 800 760 704 800 730 685
    capsule (mg)
    TU- loading (wt %) of 25 26.3 34.0 30.0 32.9 35.0
    capsule fill
    TU released in 30 43% 62% 38% 32% <75% <75%
    minutes
    Time for 75% TU <120 <120 <120 <120 <120 <120
    release (minutes)
  • Examples 26-29
  • Testosterone undecanoate formulations of Examples 26 through 29 were prepared by using the components set forth in Table XIV and a method similar to that described under Example 1. Additionally, indicated amounts of the respective formulations were filled into hard gelatin capsules and the testosterone undecanoate released from capsules was tested in about 1000 mL of 8% w/w solution of Triton X100 in water, maintained at about 37° C., using a USP Type-II apparatus at about 100 rpm. The results of the release testing are also shown in Table XIV.
  • TABLE XIV
    Composition mg/capsule
    Example Example Example Example
    Capsule Components/Attributes 26 27 28 29
    Testosterone Undecanoate 250 250 250 250
    Maize oil glycerides (Maisine 35-1) 486 937 410 939
    Polyoxyl 40 Hydrogenated Castor Oil 25 213 69 144
    (Cremophor RH40)
    Glyceryl distearate (Percirol ATO 5) 32 67
    Polyethylene Glycol 8000, 39 39
    Total mg per capsule 800 1400 800 1400
    TU- loading (wt %) of capsule fill 31.3 18 31.3 18
    TU released in 30 minutes <75% <75% <75% <75%
    Time for 75% TU release (minutes) <120 <120 <120 <120
    Note:
    Examples 27 and 29 can optionally be disposed in a delayed release capsule
  • Examples 30-35
  • Testosterone undecanoate formulations Examples 30 through 35 can be prepared by using the components set forth in Table XV and by a method similar to that described in Example 1. Additionally, indicated amounts of the respective formulations can be encapsulated in gelatin capsules and the testosterone undecanoate release from the capsules tested in about 1000 mL of 8% w/w solution of Triton X100 in water, maintained at about 37° C., using a USP Type-II apparatus at about 100 rpm. The results of the release testing are also shown in Table XV.
  • TABLE XV
    Composition mg/capsule
    Capsule Components/ Example Example Example Example Example Example
    Attributes 30 31 32 33 34 35
    Testosterone 368 320 490 240 40 300
    Undecanoate
    Maize oil glycerides 900 370 620 404
    (Maisine 35-1)
    Castor Oil 175 276
    Lauroglycol 115 184
    Tocopherol 102
    Benzyl alcohol 102
    Polyoxyl 40 46 25
    Hydrogenated
    Castor Oil,
    Polyethylene Glycol 86 45 86 41
    8000, USP
    TU- loading (wt %) of 26.3 36.3 35.0 35.0 12.0 39.5
    capsule fill
    Total Fill wt. per 1400 760 1400 685 330 760
    capsule
    TU released in 30 62% <62% <75% <75% ~100% <75%
    minutes
    Time for 75% TU <120 >120 <120 <120 <120 >120
    release (minutes)
  • Examples 30 through 35 demonstrate the importance of the choice of the solubilizers of the current invention and their levels to achieve greater testosterone undecanoate loading and yet maintain the solubilization of the testosterone undecanoate in the composition and/or the dosage form.
  • Examples 36 and 37—Testosterone Undecanoate Containing Compositions
  • The compositions of the current invention can be further adsorbed onto one or more substrate materials such as, for example, lactose, magnesium aluminosilicate, colloidal silicon dioxide, starch, microcrystalline cellulose, alkyl celluloses etc., whereby a free flowing powder/granule form is obtained which can be used as a granules, or disposed into capsule, or pressed into tablet. The amount of the substrate material can be from about 15% to about 40% of the weight of the composition. In one embodiment, the amount of the substrate material can be from about 20% to about 35% of the weight of the formed granule or powder. The method of making such adsorbed testosterone undecanoate compositions can include pouring the liquid compositions on the substrate material under and continuous mixing at room temperature or at about 50° C.-70° C., depending on the composition. After cooling, the adsorbed composition can be disposed into capsule or pressed into tablet. Table XVI illustrates examples of the freely flowable adsorbed solubilized testosterone undecanoate compositions.
  • TABLE XVI
    Composition (% w/w)
    Example Example
    Components/Attributes 36 37
    Testosterone Undecanoate 16 16
    Maize oil glycerides (Maisine 35-1) 50
    Castor Oil 45
    Sorbitan monolaurate (Span ® 20)  5
    Tocopherol  1
    Glycerylpalmito stearate  5
    Polyoxyl 40 Hydrogenated Castor Oil,  3
    Polyethylene Glycol 8000, USP  4
    Magnesium aluminosilicate (Neusilin ® US2) 25 30
  • Example 38—Stability of Testosterone Undecanoate Containing Compositions
  • A preliminary stability evaluation with respect to the change in potency and/or appearance of the potential primary degradation products was carried out with the compositions of Example 17 and Example 21, both filled in hard gelatin capsules at 200 mg/per capsule and 75 mg per capsules. The capsules were packed in HDPE bottles and staged for stability studies in the environmental chambers maintained at 25° C./60% RH. The primary degradation products were determined by a HPLC analysis method after about three months' storage and the results shown in Table XVII.
  • TABLE XVII
    TU composition Degradant
    Example 17 0.15%
    Example 21 0.06%
  • Examples 39, 39A, and 40 to 47—Testosterone Undecanoate Containing Compositions
  • Testosterone undecanoate-containing compositions (capsule fill material) were prepared by using the components set forth in Tables XVIII and XIX. It should be noted that Example 40 and 47 are expressed as wt % of Examples 17 and 12 respectively. The compositions are prepared by weighing all of the components, except the testosterone undecanoate, into a clean stainless steel container and mixed together at about 50° C. to about 70° C., using a stirrer. The testosterone undecanoate (TU) is added and stirred into the mixture of other components until the testosterone undecanoate dissolves. A predetermined quantity of the “capsule fill” was disposed into a capsule (for example, hard gelatin capsule) to get the required testosterone undecanoate dose per dosage unit. The capsules are allowed to cool at room temperature, banded (if required) and packaged in a HDPE bottle and tightly closed with an appropriate lid.
  • TABLE XVIII
    Example Example Example Example Example
    Capsule Components/Attributes 39 39A 40 41 42
    Testosterone Undecanoate, wt % 14 14 15 18 18
    Monoglycerides (e.g. maize oil monoglycerides, 64 75 63 68 68
    Maisine 35-1), wt %
    Hydrophilic surfactants (e.g. Cremophor RH40, 16  5 16  8 14
    Cremophor EL), wt %
    Fatty acids (e.g. linoleic acid, Linolenic acid,
    Oleic Acid), wt %
    Triglyceride (e.g. castor oil, maize oil, borage
    seed oil, lauroglycol, corn oil etc.), wt %
    Lipophilic surfactant (e.g. propylene glycol
    monolaurate), wt %
    Solidifying agent (e.g. polyethylene glycol  6  6  6  6
    8000), wt %
    % TU dissolved at RT 70-75 85-92 65-70 50-55 55-60
    % TU undissolved fraction at RT 25-30  8-15 30-35 45-50 40-45
    *It is noted that the compositions can also include one or more components such as anti-oxidants (e.g. BHA, BHT, Ascorbyl palmitate, vitamin E or combinations), flavorings etc.
  • TABLE XIX
    Example Example Example Example Example
    Capsule Components/Attributes 43 44 45 46 47
    Testosterone Undecanoate, wt % 25 18 20 12 12
    Monoglycerides (e.g. maize oil monoglycerides, 70
    Maisine 35-1), wt %
    Hydrophilic surfactants (e.g. Cremophor RH40,  5 17 16
    Cremophor EL), wt %
    Fatty acids (e.g. linoleic acid, Linolenic acid, 53 52 88
    Oleic Acid), wt %
    Triglyceride (e.g. castor oil, maize oil, borage 12 12 53
    seed oil, lauroglycol, corn oil etc.), wt %
    Lipophilic surfactant (e.g. propylene glycol 35
    monolaurate), wt %
    Solidifying agent (e.g. polyethylene glycol
    8000), wt %
    % TU dissolved at RT 40-45 100  100  100  100 
    % TU undissolved fraction at RT 55-60  0  0  0  0
    *It is noted that the compositions can also include one or more components such as anti-oxidants (e.g. BHA, BHT, Ascorbyl palmitate, vitamin E or combinations), flavorings etc.
  • The compositions of tables XVIII and XIX include descriptions of the amounts of testosterone undecanoate that is dissolved at room temperature (RT). The percent (%) of the undissolved testosterone undecanoate in the composition at RT can range from about 5 wt % to about 60 wt %. Examples 44 to 47 do not include a solidifying agent and that about 100 wt % of the testosterone undecanoate is dissolved in these composition. All Examples, except Examples 44 to 47, are free of ionizable fatty acids such as oleic acid.
  • Example 48
  • The clinical testing for the select inventive composition examples were conducted in a randomized study in human subjects including hypogonadal males having a pre-test screening serum testosterone concentrations less than 3 ng/mL (˜10.5 nmol/L). The study compositions were administered orally with at least 240 mL of water about 30 minutes after starting a standardized meal (at least 30-35% fat) which was preceded by a fast of about 10 hours. The testing methodology allows for determining single dose pharmacokinetic profiles. Dose amounts as well as mean pharmacokinetic parameters from the testing are shown in the Table XX.
  • TABLE XX
    Example Example Example Example Example
    Attribute 15A 40 41 45 47
    Total daily TU dose 360-460 430-460 430-460 600-650 240
    administered, mg
    Mean Cmax to mean 1.54-2.69 1.54-2.69 1.54-2.69 1.93 >2.84
    Cave ratio
    Dose-normalized 3 × 10−6 to 3 × 10−6 to 3 × 10−6 to 3 × 10−6 to 1.6 × 10−6
    Cmax, dL−1 7 × 10−6 7 × 10−6 7 × 10−6 7 × 10−6
  • As can be seen from the data presented in Table XX, Examples 40, 41, and 45 provide a higher dose normalized Cmax and a lower Cmax to Cave ratio after single dose administration (one half of the daily dose) as compared to Example 47, which is a lower loading, fully dissolved composition.
  • Example 49
  • Examples 40, 41, 44, and 45 were clinically tested in a randomized study in human male subjects, including hypogonadal males having pre-test serum testosterone concentrations less than 3 ng/mL (˜10.5 nmol/L). The example compositions were administered orally with at least 240 mL of water about 30 minutes after starting a standardized meal (at least 30-35% fat) which was preceded by a fast of about 10 hours. The testing methodology allows for determining single dose PK profiles. Dose amounts as well as mean pharmacokinetic parameters from the testing are shown in Table XXI.
  • TABLE XXI
    Example Example Example Example
    Attribute 40 41 44 45
    Total daily TU dose 430-460 430-460 632 632
    administered, mg
    Dose-normalized >1.9 × 10−6 >2.7 × 10−6 ≤1.63 × 10−6 ≤1.63 × 10−6
    Cave, dL−1
  • As can be seen in Table XXI, the compositions of Example 40 and 41 provide a desirable higher dose normalized Cave (bioavailability) after single dose administration (one half of the daily dose) as compared to Examples 44 and 45. It is noteworthy that the testosterone undecanoate in Examples 44 and 45 is fully dissolved in the composition at room temperature. Further, each of Examples 44 and 45 contain oleic acid, an ionizable fatty acid, and each is devoid of solids (i.e. a solidifying agent and/or undissolved testosterone undecanoate).
  • Example 50
  • Based on test results from Examples 40, 41 and 45 pharmacokinetic (PK) performance parameters related to target pharmacokinetic criteria in a group of hypogonadal men with no dose titration (i.e. the maintenance dose is the same as the initial TU daily dose) were estimated. The data shown is based on normal patient distribution and a variability of about 50%, although similar results are expected if the variability changes up to an additional 15%. The simulated PK performance parameters allow for the Cave and Cmax to be measured on different days after day 84. Table XXII shows the estimated parameters for Examples 40, 41 and 45, including various daily dose quantities for the compositions and capsules Example 40.
  • TABLE XXII
    Example Example Example
    Attribute/ 40A 40B 40C 40D 41 45
    Total daily TU dose, mg 150 360-420 430 1000 340-400 632
    Target PK Criteria Group PK Performance Results
    Cavg in the range of 300-1100 ng/dL <75% >75% <75% <75% >75% <75%
    Cmax is less than 1500 ng/dL >85% >85% >85% <85% >85% <85%
    Cmax in the range between 1800 and  <5%  <5%  <5%  >5%  <5%  >5%
    2500 ng/dL
    Cmax greater than 2500 ng/dL  <1%  <1%  <1%  >1%  <1%  >1%
  • Based on the results shown in Table XXII, it is evident that dosing based on Example 40-B and Example 41 provide PK performance criteria which may not require individualized titrations of the daily dose.
  • Example 51
  • Based on test results from Examples 40 and 45, pharmacokinetic (PK) performance parameters were estimated related to target PK criteria in a group of hypogonadal men using a dose titration metric of Cave with a single dose titration. The estimated data is based on normal patient distribution and a variability of about 50%. Similar results are expected if the variability in PK parameters and/or maintenance dose changes by up to 15% or the selection of another relevant dose-sensitive metric, such as Ct, Cmax, Cmin, or the like. The simulated PK performance parameters allow for the Cave and Cmax to be measured on different days after day 90. Table XXIII shows the estimated parameters for Examples 40 and 45, including various daily dose quantities for the compositions and capsules of Example 40.
  • TABLE XXIII
    Example
    40A 40E 40D Example 45
    Total TU initial Daily Dose 150 360-460 1000  580-650
    (IDD), mg
    Maintenance Daily Dose (% ±40 ±40 ±40 ±40
    of IDD)
    PK Performance Criteria Group PK Performance Results
    Cavg in the range of 300-1100 <75% >75% <75% <75%
    ng/dL
    Cmax is less than 1500 ng/dL >85% >85% <85% <85%
    Cmax in the range between  <5%  <5%  >5%  >5%
    1800 and 2500 ng/dL
    Cmax greater than 2500 ng/dL  <1%  <1%  >1%  >1%
  • Based on the results shown in Table XXIII, it is evident that high loading testosterone undecanoate compositions, such as Example 40-E, meet target PK performance criteria with a single individualized titration of the daily dose into a maintenance dose in responders to the exogenous TU and who are in need of a dose change based on titration metric results.
  • Example 52
  • Based on test results from Examples 40 and 45, pharmacokinetic performance parameters were estimated related to target PK criteria in a group of hypogonadal men using a dose titration metric of Cave and two dose titrations. The estimated data is based on normal patient distribution and a variability of about 50%. Similar results are expected if the variability in PK parameters and/or maintenance dose changes by up to 15% or the selection of another relevant dose-sensitive metric, such as Ct, Cmax, Cmin, or the like. The simulated PK performance parameters allow for the Cave and Cmax to be measured on different days after day 90. Table XXIV shows the estimated parameters for Examples 40 and 45, including various daily dose quantities for the compositions and capsules Example 40.
  • TABLE XXIV
    Example
    40A 40E 40C Example 45
    Total TU Initial Daily (IDD) 150 360-460 1000  580-650
    Maintenance Daily Dose (% ±40 ±40 ±40 ±40
    of IDD)
    PK Performance Criteria Group PK Performance Results
    Cavg in the range of 300-1100 <75% >75% <75% >75%
    ng/dL
    Cmax is less than 1500 ng/dL >85% >85% <85% >85%
    Cmax in the range between  <5%  <5%  >5%  <5%
    1800 and 2500 ng/dL
    Cmax greater than 2500 ng/dL  <1%  <1%  >1%  >1%
  • Based on the estimated values shown in Table XXIV, it is evident that high loading testosterone undecanoate compositions, such as Example 40-E, meet target PK performance criteria with a two individualized titration of the daily dose into a maintenance dose in responders to the exogenous TU and who are in need of a dose change based on titration metric results.
  • Example 53
  • Based on test results from Examples 40 41, 44 and 45, pharmacokinetic performance parameters were estimated related to target pharmacokinetic criteria in a group of hypogonadal men using a dose titration metric of Cave and three dose titrations. The estimated data is based on normal patient distribution and a variability of about 50%. Similar results are expected if the variability in PK parameters and/or maintenance dose changes by up to 15% or the selection of another relevant dose-sensitive metric, such as Ct, Cmax, Cmin, or the like. The simulated PK performance parameters allow for the Cave and Cmax to be measured on different days after day 90. Table XXV shows the estimated parameters for Examples 40, 41, 44 and 45, including various daily dose quantities for the compositions and capsules Example 40.
  • TABLE XXV
    Example Example Example Example
    40A 40E 40D 41 44 45
    Total TU Initial Daily (IDD) dose 120 360-460 1000  350-440 580-650 580-650
    Maintenance Daily Dose (% of IDD) ±40 ±40 ±40 ±40 ±40 ±40
    PK Performance Criteria Group PK Performance Results
    Cavg in the range of 300-1100 ng/dL <75% >75% <75% >75% >75% >75%
    Cmax is less than 1500 ng/dL >85% >85% <85% >85% >85% >85%
    Cmax in the range between 1800 and  <5%  <5%  >5%  <5%  <5%  <5%
    2500 ng/dL
    Cmax greater than 2500 ng/dL  <1%  <1%  >1%  <1%  <1%  <1%
  • Based on the results shown in Table XXV, it is evident that high loading testosterone undecanoate compositions, such as Examples 40-E, 41, 44, and 45 meet target PK performance criteria with a up to three individualized titrations of the daily dose into a subsequent maintenance doses in responders to the exogenous TU and who are in need of a dose changes based on titration metric results. As evident from the results of the Examples in Tables XXII to XXV, upon each subsequent titration, the compositions disclosed herein offer higher patient dose tolerance and increased chances of success to achieve serum testosterone in the eugonadal range.
  • Example 54—Clinical Practice Titration Metrics
  • Patient and physician-friendly clinical practice titrations metrics were derived from the titration node day PK measurements (Cave) following steady state (>15 days) administration of the high loading testosterone undecanoate compositions disclosed herein. It should be noted that as the Cave and Cmax were observed to have a strong correlation, the Cmax data could also be used to arrive at a similar clinical practice titration metric. As noted above in the application, Ct is the serum total testosterone concentration upon single testosterone administration, after “t” hours post-dose of a typical single daily dose of a BID daily dose regimen on the titration node day. Table XXVI shows a titration table based on the titration metric Ct for various time points following administration.
  • TABLE XXVI
    Sampling Time “Up-titration” Ct limit “Down-titration” Ct limit
    “t”(hours post-dose) (ng/ml) (ng/ml)
    t = 2 1.96 9.39
    t = 4 4.12 18.08
    t = 6 5.46 16.37
    t = 8 2.97 7.83
    t = 12 1.35 2.93
  • For example, those subjects in a group of hypogonadal men, whose C2 (i.e. Ct at t=2) is <1.96 ng/mL would have to be administered with a higher (for e.g. from about 125% to 155% of the dose prior to the titration) maintenance TU dose going forward (“up-titration) for effective testosterone therapy. Whereas those subjects in a group of hypogonadal men, whose C2 (i.e. Ct at t=2) is >9.39 ng/mL, would have to be administered with a lower (for example, from about 45 to 75% of the dose prior to the titration) maintenance TU dose going forward (“down-titration”) for effective testosterone therapy. It should also be noted that the “t” can be within about 1 hour.
  • Example 55—Dosing Regimens for Effective Testosterone Therapy in Hypogonadal Males
  • Dosing regimens were prepared based on simulated data from observed steady state PK data in hypogonadal men assuming a normal patient distribution and with variability of about 50% and a dose titration metric of Cave. Similar results can be expected if the variability in PK parameters changes by up to 15% using titration metric Ct relationship given in Table-XXVI. Safety assessment can be done any day the Cmax is measured for a testosterone product (typically, 84 days from initial dosing). Similarly, efficacy assessments can be done any day the Cave is measured for a testosterone product (typically, 84 days from initial dosing). The regimens are prepared based on the understanding that the Cave and Cmax can be measured on different days.
  • TABLE XXVII
    Oral Total TU Maintenance Metric for Cave on efficacy Cmax on safety
    Initial Daily No. of TU Daily Dose (as serum assessment assessment
    Dosing Dose (IDD), Dose % change from total T, day, day,
    Methodology mg Titrations IDD) (ng/mL) (ng/dL) (ng/dL)
    A 350-650 3 ±25-55% Ct >300 <1500
    B 580-650 2 ±25-55% Ct >300 <1500
    C 360-480 1 ±25-55% Ct >300 <1500
    D 350-420 0 ±25-55% Ct >300 <1500
    E 650-900 3 ±25-55% Ct >300 >1500
  • Unlike the recommended imprecise oral TU dosing regimen for the commercially available product Andriol®, the dosing regimen methodology disclosed herein for subjects in need of dose adjustments (high or low responders, such as illustrated in Example dosing methods, A to D) includes appropriate initial daily dose, maintenance dose and titration metric result in effective testosterone therapy (Cave>300 ng/dL) for optimal efficacy, while maintaining the Cmax<1500 ng/dL in order to provide a better safety profile. It should be noted that dosing method E for a compositions of this invention would require more than three titrations which would generally be inconvenient, impractical, and may not allow for high levels of patient compliance.
  • It is understood that the above-described various types of compositions, dosage forms and/or modes of applications are only illustrative of preferred embodiments of the present invention. Numerous modifications and alternative arrangements may be devised by those skilled in the art without departing from the spirit and scope of the present invention and the appended claims are intended to cover such modifications and arrangements. Thus, while the present invention has been described above with particularity and detail in connection with what is presently deemed to be the most practical and preferred embodiments of the invention, it will be apparent to those of ordinary skill in the art that variations including, but not limited to, variations in size, materials, shape, form, function and manner of operation, assembly and use may be made without departing from the principles and concepts set forth herein.

Claims (42)

1. A pharmaceutical capsule for oral delivery, comprising:
a capsule shell
a capsule fill, comprising
a solubilizer; and
about 14 wt % to about 35 wt % testosterone undecanoate based on the total capsule fill;
wherein a single oral administration to a male subject of one or more capsules with a total testosterone undecanoate daily dose of about 350 mg to about 650 mg provides a ratio of serum testosterone Cmax to serum testosterone Cave of about 2.7 or less.
2. The pharmaceutical capsule of claim 1, wherein upon a single dose administration of the capsule provides a dose-normalized serum testosterone Cmax of about 3×10−6 dL−1 or higher.
3. The pharmaceutical capsule of claim 1, wherein upon a single dose administration of the capsule provides a dose-normalized serum testosterone Cave of about 1.9×10−6 dL−1 or higher.
4. The pharmaceutical capsule of claim 1, wherein the capsule is a hard capsule with having a fill volume of about 0.3 mL to about 1.1 mL.
5. The pharmaceutical capsule of claim 1, wherein the testosterone undecanoate comprises about 14 wt % to about 18 wt % of the total capsule fill.
6. The pharmaceutical capsule of claim 1, wherein the solubilizer is selected from the group consisting of C8 to C22 fatty acid glycerides, C8 to C22 fatty acids, omega fatty acids, and mixtures thereof.
7. The pharmaceutical capsule of claim 6, wherein the C8 to C22 fatty acid glyceride comprises at least about 40 wt % as monoglycerides.
8. The pharmaceutical capsule of claim 6, wherein the C8 to C22 fatty acid glycerides are selected from the group consisting of monoglycerides of linoleic acid, diglycerides of linoleic acid, monoglycerides of oleic acid, diglycerides of oleic acid, and combinations thereof.
9. The pharmaceutical capsule of claim 6, wherein the C8-C22 fatty acid glycerides are selected from the group consisting of monoglycerides of capric acid, diglycerides of capric acid, monoglycerides of caprylic acid, diglycerides of caprylic acid, and combinations thereof.
10. The pharmaceutical capsule of claim 6, wherein the C8 to C22 fatty acid glycerides are derived from sources selected from the group consisting of maize oil, poppy seed oil, safflower oil, sunflower oil, borage seed oil, coconut oil, palm kernel oil, castor oil, and mixtures thereof.
11. The pharmaceutical capsule of claim 1, wherein the solubilizer comprises about 50 wt % to about 86 wt % of the capsule fill.
12. The pharmaceutical capsule of claim 1, wherein the capsule fill further includes a dispersant.
13. The pharmaceutical capsule of claim 12, wherein the dispersant includes a lipophilic surfactant selected from the group consisting of propylene glycol mono caprylate, propylene glycol oleate, propylene glycol monostearate, propylene glycol monolaurate, propylene glycol monooleate, propylene glycol dicaprylate/dicaprate, sorbitan monooleate, PEG-5 hydrogenated castor oil, PEG-7 hydrogenated castor oil, PEG-9 hydrogenated castor oil, PEG-6 corn oil, PEG-6 almond oil, PEG-6 apricot kernel oil, PEG-6 olive oil, PEG-6 peanut oil, PEG-6 hydrogenated palm kernel oil, sorbitan monolaurate, sorbitan monopalmitate, sorbitan monooleate, sorbitan monostearate, sorbitan tristearate, sorbitan monolaurate, sorbitan monopalmitate, sorbitan monooleate, sorbitan trioleate, and combinations thereof.
14. The pharmaceutical capsule of claim 12, wherein the dispersant includes a hydrophilic surfactant.
15. The pharmaceutical capsule of claim 14, wherein the hydrophilic surfactant is present in an amount such that it does not appreciably solubilize the testosterone undecanoate in the capsule fill.
16. The pharmaceutical capsule of claim 13, wherein the capsule fill includes a lipophilic component and a hydrophilic surfactant.
17. The pharmaceutical capsule of claim 1, wherein the capsule fill further comprises a solidifying agent selected from the group consisting of: polyethylene glycols, sorbitol, gelatin, stearic acid, cetyl alcohol, cetosterayl alcohol, paraffin wax, polyvinyl alcohol, glyceryl stearates, glyceryl distearate, glyceryl monostearate, glyceryl palmitostearate, glyceryl behenate, waxes, hydrogenated castor oil, hydrogenated vegetable oil, bees wax, microcrystalline wax, sterols, phytosterols, cholesterol, and mixtures thereof.
18. The pharmaceutical capsule of claim 17, wherein the solidifying agent comprises about 3 wt % to about 15 wt % of the total capsule fill.
19. The pharmaceutical capsule of claim 1, wherein upon administration of one or more capsules daily to each subject in a group of at least 24 hypogonadal males for a period of at least 84 days, the capsule provides a serum testosterone Cavg of 300 ng/dL to 1100 ng/dL in at least 75% of the hypogonadal males in the group, and at least one of the following:
a) a serum testosterone Cmax of less than 1500 ng/dL in at least 85% of the hypogonadal males in the group;
b) a serum testosterone Cmax of about 1800 ng/dL to about 2500 ng/dL in 5% or less of the hypogonadal males in the group; or
c) a serum testosterone Cmax greater than 2500 ng/dL in about 1% or less of the hypogonadal males in the group.
20. The pharmaceutical capsule of claim 19, wherein the administration over the at least 84 days can be divided into two dosing regimens including an initial regimen in which an daily dose of the initial regimen is administered and a maintenance regimen in which a maintenance daily dose is administered.
21. The pharmaceutical capsule of claim 20, wherein the daily dose of the maintenance regimen of testosterone undecanoate is about 45% to about 155% of the initial daily dose.
22. The pharmaceutical capsule of claim 20, wherein the amount of the daily dose of the maintenance regimen is based on at least one testosterone undecanoate dose titration metric derived from the measurement of the serum testosterone concentration on at least one titration node day.
23. The pharmaceutical capsule of claim 22, wherein the testosterone undecanoate dose titration can be such that:
i) if at time (t) of 1 to less than 3 hours following administration the serum testosterone concentration is less than 2.0 ng/mL there is an increase is dose and if the serum testosterone concentration is more than 9.4 there is a decrease in dose;
ii) if at time (t) of 3 to less than 8 hours following administration the serum testosterone concentration is less than 4.1 ng/mL there is an increase is dose and if the serum testosterone concentration is more than 18.1 there is a decrease in dose;
iii) if at time (t) of 8 to less than 12 hours following administration the serum testosterone concentration is less than 3.0 ng/mL there is an increase is dose and if the serum testosterone concentration is more than 7.8 there is a decrease in dose; or
iv) if at time (t) of 12 to less than 14 hours following administration the serum testosterone concentration is less than 1.4 ng/mL there is an increase is dose and if the serum testosterone concentration is more than 2.9 there is a decrease in dose.
24. The pharmaceutical capsule of claim 1, wherein the capsule fill is substantially free of ionizable fatty acids
25. The pharmaceutical capsule of claim 1, wherein at least 5 wt % of the testosterone undecanoate in the capsule fill is present in undissolved form.
26. The pharmaceutical capsule of claim 1, wherein the capsule fill includes 25 wt % or less total triglycerides.
27. The pharmaceutical capsule of claim 1, wherein the capsule fill is substantially free of oleic acid.
28. The pharmaceutical capsule of claim 1, wherein the male subject is a hypogonadal male subject.
29. A method for providing a serum concentration of testosterone within a target serum testosterone concentration Cave range for a male subject, comprising the steps of,
1) orally administering to the male subject an initial regimen including a daily dose of a testosterone undecanoate-containing composition, wherein the testosterone undecanoate comprises about 14 wt % to about 35 wt % of the testosterone undecanoate-containing composition and wherein the daily dose provides about 350 mg to about 650 mg of testosterone undecanoate to the male subject;
2) determining a dose titration metric based on a measurement of serum testosterone concentration for the male subject on at least one titration node day within the initial regimen; and
3) orally administering to the male subject a maintenance regimen including a daily dose of testosterone undecanoate-containing composition, wherein the testosterone undecanoate-containing composition comprises about 14 wt % to about 35 wt % of the testosterone undecanoate-containing composition and wherein the maintenance regimen provides an daily dose of testosterone undecanoate to the subject based on the titration metric determined on the at least one titration node day of the initial regimen sufficient to provide a serum testosterone plasma concentration within the target range
wherein the method provides a steady state ratio of serum testosterone Cmax to Cave of 2.7 or less.
30. The method of claim 29, further comprising the steps of:
4) determining a dose titration metric based on a measurement of serum testosterone concentration for the male subject on at least one titration node day within the maintenance regimen;
5) orally administering to the male subject a second maintenance regimen including a daily dose of testosterone undecanoate-containing composition, wherein the testosterone undecanoate-containing composition comprises about 14 wt % to about 35 wt % of the testosterone undecanoate-containing composition and wherein the second maintenance regimen provides an daily dose of testosterone undecanoate to the subject based on the titration metric determined on the at least one titration node day of the maintenance regimen sufficient to provide a serum testosterone plasma concentration within the target range; and optionally
6) repeating steps 4 and 5, if needed.
31. The method of claim 29, wherein the daily dose administered in the initial regimen is the same dosage amount as the daily dose administered in the maintenance regimen.
32. The method of claim 29, wherein the titration node day is any single day from day 15 to day 84 following the start of the initial regimen.
33. The method of claim 29, wherein the testosterone undecanoate dose titration can be such that:
i) if at time (t) of 1 to less than 3 hours following administration the serum testosterone concentration is less than 2.0 ng/mL there is an increase is dose and if the serum testosterone concentration is more than 9.4 there is a decrease in dose;
ii) if at time (t) of 3 to less than 8 hours following administration the serum testosterone concentration is less than 4.1 ng/mL there is an increase is dose and if the serum testosterone concentration is more than 18.1 there is a decrease in dose;
iii) if at time (t) of 8 to less than 12 hours following administration the serum testosterone concentration is less than 3.0 ng/mL there is an increase is dose and if the serum testosterone concentration is more than 7.8 there is a decrease in dose; or
v) if at time (t) of 12 to less than 14 hours following administration the serum testosterone concentration is less than 1.4 ng/mL there is an increase is dose and if the serum testosterone concentration is more than 2.9 there is a decrease in dose.
34. The method of claim 29, wherein the daily dose of the maintenance regimen provides an amount of testosterone undecanoate that is about 45% to about 155% of that of the initial daily dose.
35. The method of claim 29, wherein the target serum testosterone Cave range is about 300 ng/dL to 1100 ng/dL and is achieved on or after day 84 following the start of the initial regimen.
36. The method of claim 29, wherein the daily dose during the initial and maintenance regimens includes twice-a-day administration of the testosterone undecanoate-containing composition in conjunction with meals.
37. The method of claim 29, wherein the method provides a serum testosterone Cavg in the range of 300 ng/dL to 1100 ng/dL in 75% or more of hypogonadal males after 84 days from the start of the initial regimen based on a minimum group size of 24 hypogonadal males.
38. The method of claim 29, wherein the method provides a serum testosterone Cmax of 1500 ng/dL or less in less than or equal to 85% of hypogonadal males after 84 days from the start of the initial regimen based on a minimum group size of 24 hypogonadal males.
39. The method of claim 29, wherein the method provides a serum testosterone Cmax in the range of 1800 ng/dL to 2500 ng/dL in about 5% or less of hypogonadal males after 84 days from the start of the initial regimen based on a minimum group size of 24 hypogonadal males.
40. The method of claim 29, wherein the method provides a serum testosterone Cmax of 2500 ng/dL in about 1% or less of hypogonadal males after 84 days from the start of the initial regimen based on a minimum group size of 24 hypogonadal males.
41. The method of claim 29, wherein testosterone undecanoate comprises about 14 wt % to about 18 wt % of the testosterone undecanoate-containing composition.
42. The method of claim 29, wherein the male subject is a hypogonadal male.
US15/803,691 2010-11-30 2017-11-03 High-strength testosterone undecanoate compositions Abandoned US20180153904A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US15/803,691 US20180153904A1 (en) 2010-11-30 2017-11-03 High-strength testosterone undecanoate compositions
US16/412,360 US11433083B2 (en) 2010-11-30 2019-05-14 High-strength testosterone undecanoate compositions

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US12/957,206 US20120135074A1 (en) 2010-11-30 2010-11-30 High-Strength Testosterone Undecanoate Compositions
PCT/US2011/062538 WO2012075081A2 (en) 2010-11-30 2011-11-30 High-strength testosterone undecanoate compositions
US13/485,807 US9034858B2 (en) 2010-11-30 2012-05-31 High-strength testosterone undecanoate compositions
US14/691,229 US9205057B2 (en) 2010-11-30 2015-04-20 High-strength testosterone undecanoate compositions
US14/801,674 US20150320765A1 (en) 2010-11-30 2015-07-16 High-strength testosterone undecanoate compositions
US14/952,796 US9480690B2 (en) 2010-11-30 2015-11-25 High-strength testosterone undecanoate compositions
US15/270,357 US20170007622A1 (en) 2010-11-30 2016-09-20 High-strength testosterone undecanoate compositions
US15/803,691 US20180153904A1 (en) 2010-11-30 2017-11-03 High-strength testosterone undecanoate compositions

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/270,357 Continuation-In-Part US20170007622A1 (en) 2010-11-30 2016-09-20 High-strength testosterone undecanoate compositions

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/412,360 Continuation US11433083B2 (en) 2010-11-30 2019-05-14 High-strength testosterone undecanoate compositions

Publications (1)

Publication Number Publication Date
US20180153904A1 true US20180153904A1 (en) 2018-06-07

Family

ID=62240184

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/803,691 Abandoned US20180153904A1 (en) 2010-11-30 2017-11-03 High-strength testosterone undecanoate compositions
US16/412,360 Active US11433083B2 (en) 2010-11-30 2019-05-14 High-strength testosterone undecanoate compositions

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/412,360 Active US11433083B2 (en) 2010-11-30 2019-05-14 High-strength testosterone undecanoate compositions

Country Status (1)

Country Link
US (2) US20180153904A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11826460B2 (en) 2018-07-23 2023-11-28 Chong Kun Dang Pharmaceutical Corp. Stable pharmaceutical composition comprising testosterone undecanoate

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9034858B2 (en) 2010-11-30 2015-05-19 Lipocine Inc. High-strength testosterone undecanoate compositions

Family Cites Families (322)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2680749A (en) 1951-12-01 1954-06-08 Eastman Kodak Co Water-soluble tocopherol derivatives
US2742487A (en) 1952-05-02 1956-04-17 Coconut Processes Inc Method of extracting oil from mature, fresh coconut meats
US3097139A (en) 1960-03-10 1963-07-09 Ici Ltd Hypocholesterolaemia compositions
US3097144A (en) 1960-10-14 1963-07-09 Upjohn Co Heat-cured, polymeric, medicinal dosage film coatings containing a polyvinylpyrrolidone copolymer, polyethenoid acid, and polyethylene glycol
CH399447A (en) 1961-04-14 1965-09-30 Ciba Geigy Process for the production of a new steroid hormone ester
US3164520A (en) 1962-10-29 1965-01-05 Olin Mathieson Injectable steroid compositions containing at least 75% benzyl benzoate
US3510561A (en) 1965-05-20 1970-05-05 Canada Packers Ltd Sulfone-enhanced heparin absorption through mucous membranes
FR1578974A (en) 1968-05-16 1969-08-22
US4098802A (en) 1975-02-18 1978-07-04 Akzona Incorporated Oral pharmaceutical preparation having androgenic activity
NL189235C (en) 1974-02-28 1993-02-16 Akzo Nv METHOD FOR THE PREPARATION OF AN ORAL PHARMACEUTICAL PREPARATION WITH ANDROGENIC ACTION
US4147783A (en) 1974-02-28 1979-04-03 Akzona Incorporated Oral pharmaceutical preparation
JPS5266616A (en) 1975-11-29 1977-06-02 Sawai Seiyaku Kk Manufacturing of solidified oily liquid substance
GB1567515A (en) 1976-05-28 1980-05-14 Akzo Nv Esters of testo-sterone and 5 -dihydro-testosterone
FR2374910A1 (en) 1976-10-23 1978-07-21 Choay Sa PREPARATION BASED ON HEPARIN, INCLUDING LIPOSOMES, PROCESS FOR OBTAINING IT AND MEDICINAL PRODUCTS CONTAINING SUCH PREPARATIONS
FR2408345A1 (en) 1976-11-30 1979-06-08 Besins Jean Louis NEW COMPOSITION WITH ANTI-CONCEPTIONAL ACTION
DK25877A (en) 1977-01-21 1978-08-15 Nordisk Insulinlab PROCEDURE FOR EXTRACTING PURE ALBUMIN FROM BLOOD PLASMA
JPS53107408A (en) 1977-02-28 1978-09-19 Yamanouchi Pharmaceut Co Ltd Micellar preparation for rectal infusion
FI65914C (en) 1978-03-07 1984-08-10 Sandoz Ag FRAMEWORK FOR PHARMACEUTICAL COMPOSITION OF CYCLOSPORINE A
NZ196349A (en) 1980-03-07 1984-08-24 Interx Research Corp Enhancement of absorption rate of orally administered polar bioactive agents
JPS5770824A (en) 1980-10-20 1982-05-01 Nippon Saafuakutanto Kogyo Kk Vehicle for medicine
FR2494112B1 (en) 1980-11-19 1986-01-10 Laruelle Claude
FR2502951B1 (en) 1981-04-06 1985-12-06 Sandoz Sa TOPICAL PHARMACEUTICAL COMPOSITIONS IN THE FORM OF A MICRO-EMULSION
US4439432A (en) 1982-03-22 1984-03-27 Peat Raymond F Treatment of progesterone deficiency and related conditions with a stable composition of progesterone and tocopherols
US4654327A (en) 1982-04-21 1987-03-31 Research Corp. Quaternary ammonium complexes of heparin
DE3237814A1 (en) 1982-10-12 1984-04-12 Warner-Lambert Co., 07950 Morris Plains, N.J. WATER-FREE EMULSIONS AND USE THEREOF
SE8206744D0 (en) 1982-11-26 1982-11-26 Fluidcarbon International Ab PREPARATION FOR CONTROLLED RELEASE OF SUBSTANCES
IL68769A (en) 1983-05-23 1986-02-28 Hadassah Med Org Pharmaceutical compositions containing insulin for oral administration
US4731384A (en) 1983-07-01 1988-03-15 Troponwerke Gmbh & Co, Kg Etofenamate formulation
US4832952A (en) 1983-07-07 1989-05-23 American Home Products Corporation Pharmaceutical composition containing a liquid lubricant
DE3331009A1 (en) 1983-08-27 1985-03-14 Basf Ag, 6700 Ludwigshafen METHOD FOR INCREASING THE ENTERAL RESORBABILITY OF HEPARIN OR. HEPARINOIDS AND THE SO AVAILABLE HEPARIN OR HEPARINOID PREPARATION
DE3406497A1 (en) 1984-02-23 1985-09-05 Mueller Bernhard Willi Werner HIGHLY DISPERSAL PHARMACEUTICAL MULTI-COMPONENT SYSTEMS AND METHOD FOR THEIR PRODUCTION
US4713246A (en) 1984-03-19 1987-12-15 Bristol-Myers Company Etoposide oral dosage form
US4795327A (en) 1984-03-26 1989-01-03 Forest Laboratories, Inc. Controlled release solid drug dosage forms based on mixtures of water soluble nonionic cellulose ethers and anionic surfactants
US4572915A (en) 1984-05-01 1986-02-25 Bioglan Laboratories Clear micellized solutions of fat soluble essential nutrients
US4703042A (en) 1984-05-21 1987-10-27 Bodor Nicholas S Orally active heparin salts containing multivalent cationic units
GB8414221D0 (en) 1984-06-04 1984-07-11 Sterwin Ag Unit dosage form
DE3421468A1 (en) 1984-06-08 1985-12-19 Dr. Rentschler Arzneimittel Gmbh & Co, 7958 Laupheim LIPID NANOPELLETS AS A CARRIER SYSTEM FOR MEDICINAL PRODUCTS FOR PERORAL USE
GB8903804D0 (en) 1989-02-20 1989-04-05 Sandoz Ltd Improvements in or relating to organic compounds
US5639724A (en) 1984-07-24 1997-06-17 Sandoz Ltd. Cyclosporin galenic forms
JPS6150978A (en) 1984-08-16 1986-03-13 Takeda Chem Ind Ltd Pyridine derivative and preparation thereof
US4897269A (en) 1984-09-24 1990-01-30 Mezei Associates Limited Administration of drugs with multiphase liposomal delivery system
US4867984A (en) 1984-11-06 1989-09-19 Nagin K. Patel Drug in bead form and process for preparing same
CA1282326C (en) 1984-12-14 1991-04-02 Paul J. Jarosz Pharmaceutical composition containing 13-cis vitamin a acid as the active ingredient
DE3500103A1 (en) 1985-01-04 1986-07-10 R.P. Scherer GmbH, 6930 Eberbach PHARMACEUTICAL PREPARATION WITH AN INTENSIVE SOLUTION IN WATER AND DIGESTIVE JUICES
US4874795A (en) 1985-04-02 1989-10-17 Yesair David W Composition for delivery of orally administered drugs and other substances
US4628052A (en) 1985-05-28 1986-12-09 Peat Raymond F Pharmaceutical compositions containing dehydroepiandrosterone and other anesthetic steroids in the treatment of arthritis and other joint disabilities
FR2585246A1 (en) 1985-07-26 1987-01-30 Cortial PROCESS FOR OBTAINING SOLID PHARMACEUTICAL FORMS WITH PROLONGED RELEASE
US4717596A (en) 1985-10-30 1988-01-05 International Business Machines Corporation Method for vacuum vapor deposition with improved mass flow control
US5023108A (en) 1986-01-13 1991-06-11 Research Corporation Aqueous dispersions of waxes and lipids for pharmaceutical coating
US5433959A (en) 1986-02-13 1995-07-18 Takeda Chemical Industries, Ltd. Stabilized pharmaceutical composition
CA1327010C (en) 1986-02-13 1994-02-15 Tadashi Makino Stabilized solid pharmaceutical composition containing antiulcer benzimidazole compound and its production
US4849227A (en) 1986-03-21 1989-07-18 Eurasiam Laboratories, Inc. Pharmaceutical compositions
US4963540A (en) 1986-04-16 1990-10-16 Maxson Wayne S Method for treatment of premenstrual syndrome
US5140021A (en) 1986-04-16 1992-08-18 Genesis Systems Corporation Method and dosage form for treatment of premenstrual syndrome
SE457693B (en) 1986-07-01 1989-01-23 Drilletten Ab COMPOSITION WITH REGULATED RELEASE WAS A BIOLOGICAL MATERIAL LOST OR DISPERSED IN AN L2 PHASE
FR2602423B1 (en) 1986-08-08 1989-05-05 Ethypharm Sa PROCESS FOR THE PREPARATION OF A FENOFIBRATE-BASED MEDICINAL PRODUCT, OBTAINED BY THIS PROCESS
NZ221411A (en) 1986-08-11 1989-10-27 Innovata Biomed Ltd Pharmaceutical compositions containing microcapsules and a surfactant
US5071643A (en) 1986-10-17 1991-12-10 R. P. Scherer Corporation Solvent system enhancing the solubility of pharmaceuticals for encapsulation
GB8630273D0 (en) 1986-12-18 1987-01-28 Til Medical Ltd Pharmaceutical delivery systems
HU205861B (en) 1986-12-19 1992-07-28 Sandoz Ag Process for producing hydrosole of pharmaceutically effective material
JPH0662402B2 (en) 1987-01-14 1994-08-17 アライアンス ファーマシューチカル コーポレイション Brominated perfluorocarbon emulsion and method for producing the same
US5026560A (en) 1987-01-29 1991-06-25 Takeda Chemical Industries, Ltd. Spherical granules having core and their production
US4900734A (en) 1987-08-27 1990-02-13 Maxson Wayne S Novel pharmaceutical composition containing estradiol and progesterone for oral administration
US5756450A (en) 1987-09-15 1998-05-26 Novartis Corporation Water soluble monoesters as solubilisers for pharmacologically active compounds and pharmaceutical excipients and novel cyclosporin galenic forms
US5035891A (en) 1987-10-05 1991-07-30 Syntex (U.S.A.) Inc. Controlled release subcutaneous implant
US5244925A (en) 1987-12-18 1993-09-14 Kabi Pharmacia Aktiebolag Emulsion for parenteral administration
GB8730011D0 (en) 1987-12-23 1988-02-03 Smithkline Dauelsberg Pharmaceutical compositions
FR2627696B1 (en) 1988-02-26 1991-09-13 Fournier Innovation Synergie NEW GALENIC FORM OF FENOFIBRATE
DE3807895A1 (en) 1988-03-10 1989-09-21 Knoll Ag PRODUCTS CONTAINING A CALCIUM ANTAGONIST AND A LIPID DOWNER
US5350741A (en) 1988-07-30 1994-09-27 Kanji Takada Enteric formulations of physiologically active peptides and proteins
US5342625A (en) 1988-09-16 1994-08-30 Sandoz Ltd. Pharmaceutical compositions comprising cyclosporins
KR0148748B1 (en) 1988-09-16 1998-08-17 장 크라메르, 한스 루돌프 하우스 A multiphase cyclosporin composition
US6007840A (en) 1988-09-16 1999-12-28 Novartis Ag Pharmaceutical compositions comprising cyclosporins
GB8822857D0 (en) 1988-09-29 1988-11-02 Patralan Ltd Pharmaceutical formulations
DE3838094A1 (en) 1988-11-10 1990-05-17 Nordmark Arzneimittel Gmbh SOLID PHARMACEUTICAL RETARD FORM
US4994439A (en) 1989-01-19 1991-02-19 California Biotechnology Inc. Transmembrane formulations for drug administration
US5364632A (en) 1989-04-05 1994-11-15 Yissum Research Development Company Of The Hebrew University Of Jerusalem Medicinal emulsions
FR2647346B1 (en) 1989-05-29 1991-09-06 Besins Iscovesco Laboratoires STABILIZED PROGESTERONE COMPOUND, PROCESS FOR PREPARING THE SAME, AND USE OF THE COMPOUND FOR OBTAINING A MEDICAMENT
US5104656A (en) 1989-06-16 1992-04-14 Seth Pyare L Percutaneous treatment with a high potency non-steroidal anti-inflammatory agent
DE3919982A1 (en) 1989-06-19 1990-12-20 Liedtke Pharmed Gmbh ORAL LIPID MEDICINE FORM
US5532002A (en) 1989-08-17 1996-07-02 Cortecs Limited Gelatin pharmaceutical formulations
US5091187A (en) 1990-04-26 1992-02-25 Haynes Duncan H Phospholipid-coated microcrystals: injectable formulations of water-insoluble drugs
US5091188A (en) 1990-04-26 1992-02-25 Haynes Duncan H Phospholipid-coated microcrystals: injectable formulations of water-insoluble drugs
US5298497A (en) 1990-05-15 1994-03-29 E. R. Squibb & Sons, Inc. Method for preventing onset of hypertension employing a cholesterol lowering drug
ATE121618T1 (en) 1990-08-13 1995-05-15 David W Yesair MIXED LIPID-BICARBONATE COLLOIDAL PARTICLES FOR DELIVERING MEDICINAL AND CALORIES.
US5270005A (en) 1990-09-07 1993-12-14 Baxter International Inc. Extracorporeal blood oxygenation system incorporating integrated reservoir-membrane oxygenerator-heat exchanger and pump assembly
US5665379A (en) 1990-09-28 1997-09-09 Pharmacia & Upjohn Aktiebolag Lipid particle forming matrix, preparation and use thereof
DE69124459T3 (en) 1990-11-02 2001-05-31 Novartis Ag Cyclosporine
US5152997A (en) 1990-12-11 1992-10-06 Theratech, Inc. Method and device for transdermally administering testosterone across nonscrotal skin at therapeutically effective levels
US5145684A (en) 1991-01-25 1992-09-08 Sterling Drug Inc. Surface modified drug nanoparticles
IT1245761B (en) 1991-01-30 1994-10-14 Alfa Wassermann Spa PHARMACEUTICAL FORMULATIONS CONTAINING GLYCOSAMINOGLICANS ABSORBABLE ORALLY.
US5300529A (en) 1991-02-12 1994-04-05 Isp Investments Inc. Stable, clear, efficacious aqueous microemulsion compositions containing a high loading of a water-insoluble, agriculturally active chemical
US5403593A (en) 1991-03-04 1995-04-04 Sandoz Ltd. Melt granulated compositions for preparing sustained release dosage forms
TW212139B (en) 1991-04-15 1993-09-01 Yamanouchi Pharma Co Ltd
AU668509B2 (en) 1991-04-19 1996-05-09 Affinity Biotech, Inc. Convertible microemulsion formulations
US5688761A (en) 1991-04-19 1997-11-18 Lds Technologies, Inc. Convertible microemulsion formulations
US5223268A (en) 1991-05-16 1993-06-29 Sterling Drug, Inc. Low solubility drug-coated bead compositions
US5380535A (en) 1991-05-28 1995-01-10 Geyer; Robert P. Chewable drug-delivery compositions and methods for preparing the same
US5698155A (en) 1991-05-31 1997-12-16 Gs Technologies, Inc. Method for the manufacture of pharmaceutical cellulose capsules
IL102633A0 (en) 1991-07-26 1993-01-14 Smithkline Beecham Corp Compositions
WO1993006921A1 (en) 1991-10-04 1993-04-15 Gs Biochem Ab Particles, method of preparing said particles and uses thereof
JPH0597672A (en) 1991-10-08 1993-04-20 Terumo Corp Amide derivative-containing solid preparation and its production
RU2120798C1 (en) 1991-11-22 1998-10-27 Проктер энд Гэмбл Фармасьютикалз, Инк. Solid pharmaceutical composition for oral administration
US5206219A (en) 1991-11-25 1993-04-27 Applied Analytical Industries, Inc. Oral compositions of proteinaceous medicaments
HUT63579A (en) 1991-12-20 1993-09-28 Chinoin Gyogyszer Es Vegyeszet Process for producing double-phase pharmaceutical compositions suitable for treating diseases occurring during neurodegenerative processes
JP3852621B2 (en) 1992-01-21 2006-12-06 あすか製薬株式会社 Vascular endothelial cell function improving agent
GB9201857D0 (en) 1992-01-29 1992-03-18 Smithkline Beecham Plc Novel compound
US5571533A (en) 1992-02-07 1996-11-05 Recordati, S.A., Chemical And Pharmaceutical Company Controlled-release mucoadhesive pharmaceutical composition for the oral administration of furosemide
SE9200951D0 (en) 1992-03-27 1992-03-27 Kabi Pharmacia Ab PHARMACEUTICAL COMPOSITION CONTAINING A DEFINED LIPID SYSTEM
GB9212511D0 (en) 1992-06-12 1992-07-22 Cortecs Ltd Pharmaceutical compositions
IT1255449B (en) 1992-06-30 1995-10-31 Fabio Berlati USE OF NOR- AND HOMO-DERIVATIVES OF BILE ACIDS AS DRUGS ABSORPTION PROMOTERS.
PH30929A (en) 1992-09-03 1997-12-23 Janssen Pharmaceutica Nv Beads having a core coated with an antifungal and a polymer.
JPH08505367A (en) 1992-10-16 1996-06-11 スミスクライン・ビーチャム・コーポレイション Pharmaceutical emulsion composition
US5376688A (en) 1992-12-18 1994-12-27 R. P. Scherer Corporation Enhanced solubility pharmaceutical solutions
GB9300875D0 (en) 1993-01-18 1993-03-10 Ucb Sa Nanocapsule containing pharmaceutical compositions
US5686105A (en) 1993-10-19 1997-11-11 The Procter & Gamble Company Pharmaceutical dosage form with multiple enteric polymer coatings for colonic delivery
DE4312034A1 (en) 1993-04-13 1994-10-20 Hesch Rolf Dieter Prof Dr Med Novel androgens and anabolic steroids
AU686149B2 (en) 1993-04-19 1998-02-05 Institute For Advanced Skin Research Inc. Microemulsion preparation containing difficultly absorbable substance
BE1006990A5 (en) 1993-04-22 1995-02-07 Univ Gent METHOD AND COMPOSITION TO MAKE AN ACTIVE INGREDIENT IN A solid dosage form.
DE69425453T2 (en) 1993-04-23 2001-04-12 Novartis Ag Drug delivery device with controlled release
NZ247516A (en) 1993-04-28 1995-02-24 Bernard Charles Sherman Water dispersible pharmaceutical compositions comprising drug dissolved in solvent system comprising at least one alcohol and at least one surfactant
SE9302135D0 (en) 1993-06-18 1993-06-18 Kabi Pharmacia Ab NEW PHARMACEUTICAL COMPOSITION
US5639474A (en) 1993-07-01 1997-06-17 Hanmi Pharm. Ind., Ltd. Cyclosporin soft capsule composition
DE4322826A1 (en) 1993-07-08 1995-01-12 Galenik Labor Freiburg Gmbh Pharmaceutical preparation
ES2068762B1 (en) 1993-07-21 1995-12-01 Lipotec Sa A NEW PHARMACEUTICAL PREPARATION TO IMPROVE THE BIOAVAILABILITY OF DRUGS OF DIFFICULT ABSORPTION AND PROCEDURE FOR THEIR OBTAINING.
JPH0741422A (en) 1993-07-30 1995-02-10 Nissui Pharm Co Ltd Method for solubilizing gamma-oryzanol in water
TW359614B (en) 1993-08-31 1999-06-01 Takeda Chemical Industries Ltd Composition containing benzimidazole compounds for rectal administration
DE69424487T2 (en) 1993-09-09 2001-01-18 Takeda Chemical Industries Ltd Formulation containing an antibacterial and an antiulcus active ingredient
US6022852A (en) 1993-10-22 2000-02-08 Hexal Ag Pharmaceutical composition containing cyclosporin A
WO1995014037A1 (en) 1993-11-17 1995-05-26 Ibah, Inc. Transparent liquid for encapsulated drug delivery
DE4340781C3 (en) 1993-11-30 2000-01-27 Novartis Ag Liquid preparations containing cyclosporin and process for their preparation
US5374446A (en) 1993-12-10 1994-12-20 Arco Chemical Technology, L.P. Linked esterified alkoxylated polyols useful as reduced calorie fat substitutes
KR0146671B1 (en) 1994-02-25 1998-08-17 김충환 Cyclosporin-containing powder composition
US5811120A (en) 1994-03-02 1998-09-22 Eli Lilly And Company Solid orally administerable raloxifene hydrochloride pharmaceutical formulation
GB9405304D0 (en) 1994-03-16 1994-04-27 Scherer Ltd R P Delivery systems for hydrophobic drugs
US5731355A (en) 1994-03-22 1998-03-24 Zeneca Limited Pharmaceutical compositions of propofol and edetate
GB9409778D0 (en) 1994-05-16 1994-07-06 Dumex Ltd As Compositions
SE518578C2 (en) 1994-06-15 2002-10-29 Gs Dev Ab Lipid-based composition
US6692766B1 (en) 1994-06-15 2004-02-17 Yissum Research Development Company Of The Hebrew University Of Jerusalem Controlled release oral drug delivery system
US5817320A (en) 1994-06-20 1998-10-06 The United States Of America As Represented By The Secretary Of The Agriculture In ovo immunization of avian embryos with oil-emulsion vaccines
US5616330A (en) 1994-07-19 1997-04-01 Hemagen/Pfc Stable oil-in-water emulsions incorporating a taxine (taxol) and method of making same
FR2722984B1 (en) 1994-07-26 1996-10-18 Effik Lab PROCESS FOR THE PREPARATION OF DRY PHARMACEUTICAL FORMS AND THE PHARMACEUTICAL COMPOSITIONS THUS PRODUCED
GB9417524D0 (en) 1994-08-31 1994-10-19 Cortecs Ltd Pharmaceutical compositions
SE518619C2 (en) 1994-12-09 2002-10-29 Gs Dev Ab Controlled release composition containing monocaproin
US5858398A (en) 1994-11-03 1999-01-12 Isomed Inc. Microparticular pharmaceutical compositions
US5965161A (en) 1994-11-04 1999-10-12 Euro-Celtique, S.A. Extruded multi-particulates
KR0167613B1 (en) 1994-12-28 1999-01-15 한스 루돌프 하우스, 니콜 케르커 Cyclosporin-containing soft capsule compositions
US5545628A (en) 1995-01-10 1996-08-13 Galephar P.R. Inc. Pharmaceutical composition containing fenofibrate
US5629021A (en) 1995-01-31 1997-05-13 Novavax, Inc. Micellar nanoparticles
FR2730231B1 (en) 1995-02-02 1997-04-04 Fournier Sca Lab COMBINATION OF FENOFIBRATE AND VITAMIN E, USE IN THERAPEUTICS
US5560931A (en) 1995-02-14 1996-10-01 Nawosystems L.L.C. Formulations of compounds as nanoparticulate dispersions in digestible oils or fatty acids
US5571536A (en) 1995-02-06 1996-11-05 Nano Systems L.L.C. Formulations of compounds as nanoparticulate dispersions in digestible oils or fatty acids
US5573783A (en) 1995-02-13 1996-11-12 Nano Systems L.L.C. Redispersible nanoparticulate film matrices with protective overcoats
JP2740153B2 (en) 1995-03-07 1998-04-15 エフ・ホフマン−ラ ロシユ アーゲー Mixed micelle
US5653987A (en) 1995-05-16 1997-08-05 Modi; Pankaj Liquid formulations for proteinic pharmaceuticals
SI9500173B (en) 1995-05-19 2002-02-28 Lek, Three-phase pharmaceutical form with constant and controlled release of amorphous active ingredient for single daily application
US5726181A (en) 1995-06-05 1998-03-10 Bionumerik Pharmaceuticals, Inc. Formulations and compositions of poorly water soluble camptothecin derivatives
FR2737121B1 (en) 1995-07-27 1997-10-03 Cl Pharma NEW GALENIC FORMULATIONS OF FENOFIBRATE AND THEIR APPLICATIONS
US5766629A (en) 1995-08-25 1998-06-16 Sangstat Medical Corporation Oral cyclosporin formulations
US6645988B2 (en) 1996-01-04 2003-11-11 Curators Of The University Of Missouri Substituted benzimidazole dosage forms and method of using same
US5858401A (en) 1996-01-22 1999-01-12 Sidmak Laboratories, Inc. Pharmaceutical composition for cyclosporines
JPH09241152A (en) 1996-03-01 1997-09-16 Sunstar Inc Oil-in-water emulsion
IL117773A (en) 1996-04-02 2000-10-31 Pharmos Ltd Solid lipid compositions of coenzyme Q10 for enhanced oral bioavailability
GB9608719D0 (en) 1996-04-26 1996-07-03 Scherer Ltd R P Pharmaceutical compositions
DE19619045C1 (en) 1996-05-02 1997-11-13 Jenapharm Gmbh Use of combination products for the treatment of hypogonadal men and men with pituitary disorders
US6057339A (en) 1996-05-09 2000-05-02 Bristol-Myers Squibb Company Method of inhibiting or treating phytosterolemia with an MTP inhibitor
WO1997048382A2 (en) 1996-06-18 1997-12-24 Otsuka Pharmaceutical Co., Ltd. Multiple-unit type prolonged action drug preparation
US5846971A (en) 1996-06-28 1998-12-08 Schering Corporation Oral antifungal composition
EP0914100A1 (en) 1996-06-28 1999-05-12 Schering Corporation Oral composition comprising a triazole antifungal compound
US5883109A (en) 1996-07-24 1999-03-16 Bristol-Myers Squibb Company Method for lowering serum lipid levels employing an MTP inhibitor in combination with another cholesterol lowering drug
EP1208847B8 (en) 1996-07-30 2007-02-14 Novartis AG Pharmaceutical compositions for the treatment of transplant rejection, as well as autoimmune or inflammatory conditions
US6465016B2 (en) 1996-08-22 2002-10-15 Research Triangle Pharmaceuticals Cyclosporiine particles
IL128632A (en) 1996-08-22 2003-03-12 Rtp Pharma Corp Compositions comprising microparticles of water-insoluble substances and method for preparing same
SE9603077D0 (en) 1996-08-29 1996-08-29 Tetra Laval Holdings & Finance An apparatus for and method of performing an animal-related action regarding at least a portion of the body of an animal
US5891469A (en) 1997-04-02 1999-04-06 Pharmos Corporation Solid Coprecipitates for enhanced bioavailability of lipophilic substances
US5798333A (en) 1996-09-17 1998-08-25 Sherman; Bernard C. Water-soluble concentrates containing cyclosporins
US6361796B1 (en) 1996-10-25 2002-03-26 Shire Laboratories, Inc. Soluble form osmotic dose delivery system
US6077543A (en) 1996-12-31 2000-06-20 Inhale Therapeutic Systems Systems and processes for spray drying hydrophobic drugs with hydrophilic excipients
US6458373B1 (en) 1997-01-07 2002-10-01 Sonus Pharmaceuticals, Inc. Emulsion vehicle for poorly soluble drugs
GB9700878D0 (en) 1997-01-17 1997-03-05 Scherer Ltd R P Dosage forms and method for ameliorating male erectile dysfunction
FR2758459B1 (en) 1997-01-17 1999-05-07 Pharma Pass FENOFIBRATE PHARMACEUTICAL COMPOSITION HAVING HIGH BIODAVAILABILITY AND PROCESS FOR PREPARING THE SAME
US6066653A (en) 1997-01-17 2000-05-23 Bristol-Myers Squibb Co. Method of treating acid lipase deficiency diseases with an MTP inhibitor and cholesterol lowering drugs
KR20000069900A (en) 1997-01-30 2000-11-25 한스 루돌프 하우스, 헨리테 브룬너, 베아트리체 귄터 Oil-Free Pharmaceutical Compositions Containing Cyclosporin A
ES2384551T3 (en) 1997-03-05 2012-07-06 Sugen, Inc. Formulations for hydrophobic pharmaceutical agents
ATE286403T1 (en) 1997-03-12 2005-01-15 Abbott Lab HYDROPHILIC BINARY SYSTEMS FOR ADMINISTRATION OF CYCLOSPORINE
WO1998047486A1 (en) 1997-04-18 1998-10-29 Taisho Pharmaceutical Co., Ltd. Microemulsion
JPH1149664A (en) 1997-04-18 1999-02-23 Taisho Pharmaceut Co Ltd Microemulsion
US6046177A (en) 1997-05-05 2000-04-04 Cydex, Inc. Sulfoalkyl ether cyclodextrin based controlled release solid pharmaceutical formulations
US5874418A (en) 1997-05-05 1999-02-23 Cydex, Inc. Sulfoalkyl ether cyclodextrin based solid pharmaceutical formulations and their use
US5981586A (en) 1997-05-23 1999-11-09 Pershadsingh; Harrihar A. Methods for treating proliferative and inflammatory skin diseases
TW580397B (en) 1997-05-27 2004-03-21 Takeda Chemical Industries Ltd Solid preparation
SK282427B6 (en) 1997-06-11 2002-01-07 Abbott Laboratories Solid pharmaceutical composition with controlled release
JP2002510311A (en) 1997-06-27 2002-04-02 アストラ・アクチエボラーグ Proliposomal powder for inhalation stabilized by tocopherol
KR100509130B1 (en) 1997-07-29 2005-08-18 파마시아 앤드 업존 캄파니 엘엘씨 Self-Emulsifying Formulation for Lipophilic Compounds
IT1294760B1 (en) 1997-09-03 1999-04-12 Jagotec Ag PROCEDURE FOR THE PREPARATION OF PHARMACEUTICAL TABLETS ABLE TO RELEASE, ACCORDING TO PREDETERMINABLE SCHEMES, LITTLE ACTIVE INGREDIENTS
KR100222918B1 (en) 1997-09-04 1999-10-01 윤덕용 Absorbent comprising of alkali salt and copper oxide deposited ñ†-alumina
CA2214895C (en) 1997-09-19 1999-04-20 Bernard Charles Sherman Improved pharmaceutical composition comprising fenofibrate
US6296876B1 (en) 1997-10-06 2001-10-02 Isa Odidi Pharmaceutical formulations for acid labile substances
US20050070516A1 (en) 1997-10-28 2005-03-31 Vivus Inc. As-needed administration of an androgenic agent to enhance female desire and responsiveness
US20020013304A1 (en) 1997-10-28 2002-01-31 Wilson Leland F. As-needed administration of an androgenic agent to enhance female sexual desire and responsiveness
US6027747A (en) 1997-11-11 2000-02-22 Terracol; Didier Process for the production of dry pharmaceutical forms and the thus obtained pharmaceutical compositions
US5891845A (en) 1997-11-21 1999-04-06 Fuisz Technologies Ltd. Drug delivery systems utilizing liquid crystal structures
HUP0100043A2 (en) 1997-12-08 2001-08-28 Byk Gulden Lomberg Chemische Fabrik Gmbh. Novel administration form comprising an acid-sensitive active compound
WO1999029300A1 (en) 1997-12-10 1999-06-17 Rtp Pharma Inc. Self-emulsifying fenofibrate formulations
US6013665A (en) 1997-12-16 2000-01-11 Abbott Laboratories Method for enhancing the absorption and transport of lipid soluble compounds using structured glycerides
US6287594B1 (en) 1998-01-20 2001-09-11 Edward S. Wilson Oral liquid compositions
US6086376A (en) 1998-01-30 2000-07-11 Rtp Pharma Inc. Dry aerosol suspension of phospholipid-stabilized drug microparticles in a hydrofluoroalkane propellant
FR2774591B1 (en) 1998-02-12 2000-05-05 Lipha PHARMACEUTICAL COMPOSITION COMPRISING THE ASSOCIATION OF METFORMIN AND FIBRATE AND THE USE THEREOF FOR THE PREPARATION OF MEDICINES FOR REDUCING HYPERGLYCEMIA
ID25908A (en) 1998-03-06 2000-11-09 Novartis Ag EMULSION PRACTONCENTRATES CONTAINING CYCLOSPORINE OR MACROLIDES
DK173431B1 (en) 1998-03-20 2000-10-23 Gea Farmaceutisk Fabrik As Pharmaceutical formulation comprising a 2 - [[(2-pyridinyl) methyl] sulfinyl] benzimidazole with anti-ulcer activity and progress
CA2268211A1 (en) 1998-04-13 1999-10-13 Medical College Of Hampton Roads Control of selective estrogen receptor modulators
DK1736144T3 (en) 1998-05-18 2015-12-07 Takeda Pharmaceutical Orally disintegrating tablets.
EP1015046A2 (en) 1998-07-14 2000-07-05 Em Industries, Inc. Microdisperse drug delivery systems
ES2157731B1 (en) 1998-07-21 2002-05-01 Liconsa Liberacion Controlada ORAL PHARMACEUTICAL PREPARATION OF AN ANTIFUNGIC ACTIVITY COMPOUND AND PROCEDURE FOR PREPARATION.
US6174547B1 (en) 1999-07-14 2001-01-16 Alza Corporation Dosage form comprising liquid formulation
US5993880A (en) 1998-10-01 1999-11-30 Kraft Foods Inc. Non-staining, acid-stable, cold-water-soluble, edible green color and compositions for preparing acidic foods and beverages
US6977083B1 (en) 1998-10-02 2005-12-20 Jenapharm Gmbh & Co. Kg Bioadhesive tablet containing testosterone/testosterone ester mixtures and method for producing a predetermined testosterone time-release profile with same
US6383517B1 (en) 1999-01-29 2002-05-07 Abbott Laboratories Process for preparing solid formulations of lipid-regulating agents with enhanced dissolution and absorption
US6180138B1 (en) 1999-01-29 2001-01-30 Abbott Laboratories Process for preparing solid formulations of lipid-regulating agents with enhanced dissolution and absorption
US6447806B1 (en) 1999-02-25 2002-09-10 Novartis Ag Pharmaceutical compositions comprised of stabilized peptide particles
US7374779B2 (en) 1999-02-26 2008-05-20 Lipocine, Inc. Pharmaceutical formulations and systems for improved absorption and multistage release of active agents
US6294192B1 (en) 1999-02-26 2001-09-25 Lipocine, Inc. Triglyceride-free compositions and methods for improved delivery of hydrophobic therapeutic agents
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US20030104048A1 (en) 1999-02-26 2003-06-05 Lipocine, Inc. Pharmaceutical dosage forms for highly hydrophilic materials
US6761903B2 (en) 1999-06-30 2004-07-13 Lipocine, Inc. Clear oil-containing pharmaceutical compositions containing a therapeutic agent
US6267985B1 (en) 1999-06-30 2001-07-31 Lipocine Inc. Clear oil-containing pharmaceutical compositions
WO2000059512A1 (en) 1999-04-01 2000-10-12 Akzo Nobel N.V. Formulation comprising testosteron undecanoate and castor oil
US6652880B1 (en) 1999-04-01 2003-11-25 R.P. Scherer Technologies, Inc. Oral pharmaceutical compositions containing long-chain triglycerides and liphophilic surfactants
US6383471B1 (en) 1999-04-06 2002-05-07 Lipocine, Inc. Compositions and methods for improved delivery of ionizable hydrophobic therapeutic agents
MXPA01011981A (en) 1999-05-24 2003-09-04 Sonus Pharma Inc Emulsion vehicle for poorly soluble drugs.
GB9912476D0 (en) 1999-05-28 1999-07-28 Novartis Ag Organic compounds
US20030235595A1 (en) 1999-06-30 2003-12-25 Feng-Jing Chen Oil-containing, orally administrable pharmaceutical composition for improved delivery of a therapeutic agent
US6458383B2 (en) 1999-08-17 2002-10-01 Lipocine, Inc. Pharmaceutical dosage form for oral administration of hydrophilic drugs, particularly low molecular weight heparin
US6982281B1 (en) 2000-11-17 2006-01-03 Lipocine Inc Pharmaceutical compositions and dosage forms for administration of hydrophobic drugs
US6309663B1 (en) 1999-08-17 2001-10-30 Lipocine Inc. Triglyceride-free compositions and methods for enhanced absorption of hydrophilic therapeutic agents
US20030236236A1 (en) 1999-06-30 2003-12-25 Feng-Jing Chen Pharmaceutical compositions and dosage forms for administration of hydrophobic drugs
AR024462A1 (en) 1999-07-01 2002-10-02 Merck & Co Inc PHARMACEUTICAL TABLETS
US6228400B1 (en) 1999-09-28 2001-05-08 Carlsbad Technology, Inc. Orally administered pharmaceutical formulations of benzimidazole derivatives and the method of preparing the same
US6720001B2 (en) 1999-10-18 2004-04-13 Lipocine, Inc. Emulsion compositions for polyfunctional active ingredients
US20030180352A1 (en) 1999-11-23 2003-09-25 Patel Mahesh V. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US20060034937A1 (en) 1999-11-23 2006-02-16 Mahesh Patel Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
PT1108425E (en) 1999-12-16 2005-10-31 Medinfar Produtos Farmaceutico NEW MULTI-MUNICIPAL PHARMACEUTICAL PREPARATIONS CONTAINING SUBSTITUTED BENZIMIDAZOES
HUP0204372A3 (en) 1999-12-23 2004-06-28 Pfizer Prod Inc Pharmaceutical compositions providing enhanced drug concentrations
EP2415462A1 (en) 1999-12-23 2012-02-08 Mayne Pharma International Pty Ltd. Improved pharmaceutical compositions for poorly soluble drugs
US6340471B1 (en) 1999-12-30 2002-01-22 Alvin Kershman Method for preparing solid delivery system for encapsulated and non-encapsulated pharmaceuticals
FR2803203B1 (en) 1999-12-31 2002-05-10 Fournier Ind & Sante NEW GALENIC FORMULATIONS OF FENOFIBRATE
US20020102301A1 (en) 2000-01-13 2002-08-01 Joseph Schwarz Pharmaceutical solid self-emulsifying composition for sustained delivery of biologically active compounds and the process for preparation thereof
US7025979B2 (en) 2000-02-15 2006-04-11 Schering Ag Male contraceptive formulation comprising norethisterone
DE10016544A1 (en) 2000-04-03 2001-10-11 Bayer Ag New phenyl-substituted cyclic keto-enol compounds useful e.g. as insecticides, acaricides, nematocides, acaricides, herbicides, ectoparasiticides, antifouling agents or intermediates
US6468559B1 (en) 2000-04-28 2002-10-22 Lipocine, Inc. Enteric coated formulation of bishosphonic acid compounds and associated therapeutic methods
WO2002000261A2 (en) 2000-06-26 2002-01-03 Monsanto Technology Llc Surfactant-containing formulations for extended release of somatotropin
CA2382371C (en) 2000-07-07 2011-09-20 Baxter International Inc. Medical system, method and apparatus employing mems
WO2002015938A2 (en) 2000-08-23 2002-02-28 Akzo Nobel N.V. Testosterone ester formulation for human use
US20040002482A1 (en) 2000-08-30 2004-01-01 Dudley Robert E. Androgen pharmaceutical composition and method for treating depression
US6503894B1 (en) 2000-08-30 2003-01-07 Unimed Pharmaceuticals, Inc. Pharmaceutical composition and method for treating hypogonadism
JP4637338B2 (en) 2000-09-22 2011-02-23 大塚製薬株式会社 Cilostazol dry coated tablets
US6887493B2 (en) 2000-10-25 2005-05-03 Adi Shefer Multi component controlled release system for oral care, food products, nutraceutical, and beverages
US6589562B1 (en) 2000-10-25 2003-07-08 Salvona L.L.C. Multicomponent biodegradable bioadhesive controlled release system for oral care products
US20020103139A1 (en) 2000-12-01 2002-08-01 M. Weisspapir Solid self-emulsifying controlled release drug delivery system composition for enhanced delivery of water insoluble phytosterols and other hydrophobic natural compounds for body weight and cholestrol level control
US20060142257A1 (en) 2001-01-19 2006-06-29 Eberhard Nieschlag Male contraceptive formulation comprising norethisterone
DE10164844B4 (en) 2001-02-22 2005-05-25 Aquanova German Solubilisate Technologies (Agt) Gmbh Tocopherol concentrate
AUPR573001A0 (en) 2001-06-15 2001-07-12 Glaxo Wellcome Australia Ltd Lymphatic drug delivery system
US20030022875A1 (en) 2001-07-27 2003-01-30 Wilson Leland F. As-needed administration of orally active androgenic agents to enhance female sexual desire and responsiveness
WO2003030894A1 (en) 2001-10-11 2003-04-17 Alcon, Inc. Methods for treating dry eye
US6665880B2 (en) 2001-11-01 2003-12-23 Kimberly-Clark Worldwide, Inc. Protective garments with glove flaps
US6630134B1 (en) 2002-01-08 2003-10-07 Zenitech Llc Guerbet wax esters in personal care applications
ATE395044T1 (en) 2002-02-01 2008-05-15 Pfizer Prod Inc PHARMACEUTICAL COMPOSITIONS OF AMORPHIC DISPERSIONS OF ACTIVE INGREDIENTS AND LIPOPHILIC MICROPHASE-FORMING MATERIALS
US20030190343A1 (en) 2002-03-05 2003-10-09 Pfizer Inc. Palatable pharmaceutical compositions for companion animals
US20040002445A1 (en) 2002-03-28 2004-01-01 Rajneesh Taneja Enhancement of endogenous gonadotropin production
US20030186892A1 (en) 2002-03-28 2003-10-02 Rajneesh Taneja Enhancement of endogenous gonadotropin production
DE10218107A1 (en) 2002-04-23 2003-11-20 Jenapharm Gmbh Process for the production of crystals of steroids, crystals available thereafter and their use in pharmaceutical formulations
IL164870A0 (en) 2002-04-30 2005-12-18 Britch Ford Steel John A Security and property management system
US20050287203A1 (en) 2002-05-08 2005-12-29 Nijs De H Formulation comprising testosteron undecanoate and castor oil
US20030209508A1 (en) 2002-05-10 2003-11-13 Gayle Rosenberg Expandable mug organizer
EP1562548A1 (en) 2002-11-14 2005-08-17 Shear/Kershman Laboratories, Inc. Oral testosterone delivery system with improved sustained release
US20040115287A1 (en) 2002-12-17 2004-06-17 Lipocine, Inc. Hydrophobic active agent compositions and methods
US20050100608A1 (en) 2003-02-21 2005-05-12 Watson Pharmaceuticals, Inc. Testosterone oral dosage formulations and associated methods
JO2505B1 (en) 2003-03-14 2009-10-05 باير شيرنغ فارما اكتنجيسيلشافت method and pharmaceutical compositions for reliable achievements of acceptable serum testosterone levels
US6913244B1 (en) 2003-05-02 2005-07-05 Gordon Edgar Atkinson Urinary slide valve
WO2005011635A2 (en) 2003-08-04 2005-02-10 Pfizer Products Inc. Pharmaceutical compositions of adsorbates of amorphous drugs and lipophilic microphase-forming materials
DE602004017198D1 (en) 2003-08-04 2008-11-27 Camurus Ab METHOD FOR LOADING AMPHIPHILIC PARTICLES WITH ACTIVE SUBSTANCES
AR047938A1 (en) 2003-08-25 2006-03-15 Combinatorx Inc FORMULATIONS, CONJUGATES AND COMBINATIONS OF PHARMACOS FOR THE TREATMENT OF NEOPLASMS
CA2540984C (en) 2003-10-10 2011-02-08 Lifecycle Pharma A/S A solid dosage form comprising a fibrate
US20050096365A1 (en) 2003-11-03 2005-05-05 David Fikstad Pharmaceutical compositions with synchronized solubilizer release
US20060003002A1 (en) 2003-11-03 2006-01-05 Lipocine, Inc. Pharmaceutical compositions with synchronized solubilizer release
US7138389B2 (en) 2004-02-09 2006-11-21 University Of Washington Oral androgen therapy using modulators of testosterone bioavailability
US20050220825A1 (en) 2004-03-10 2005-10-06 Adrian Funke Molecular dispersions of drospirenone
WO2006007354A2 (en) 2004-06-28 2006-01-19 Alza Corporation A drug/polymer complex, preferably ciprofloxacin/hpmc, its method of manufacturing using lyophilization and its use in an osmotic device
WO2006012502A2 (en) 2004-07-23 2006-02-02 Rigel Pharmaceuticals, Inc. Formulation of insoluble small molecule therapeutics in lipid-based carriers
US20060106004A1 (en) 2004-11-12 2006-05-18 Brody Steven A Unique methods and formulations of bio-identical sex steroids for the treatment of pathophysiologic aberrations of menopause
US20060134210A1 (en) 2004-12-22 2006-06-22 Astrazeneca Ab Solid dosage form comprising proton pump inhibitor and suspension made thereof
CN101227892B (en) 2005-04-08 2013-06-05 舌交付有限公司 Buccal delivery system
WO2006113242A2 (en) 2005-04-13 2006-10-26 Unimed Pharmaceuticals, Inc. Method of increasing testosterone and related steroid concentrations in women
US8492369B2 (en) 2010-04-12 2013-07-23 Clarus Therapeutics Inc Oral testosterone ester formulations and methods of treating testosterone deficiency comprising same
KR20080009201A (en) 2005-04-15 2008-01-25 클라루스 쎄러퓨틱스, 아이엔씨. Pharmaceutical delivery systems for hydrophobic drugs and compositions comprising same
CN101217963A (en) 2005-04-15 2008-07-09 克劳拉斯医疗有限公司 Pharmaceutical delivery systems for hydrophobic drugs and compositions comprising same
KR20080038317A (en) 2005-06-30 2008-05-06 코닌클리즈케 필립스 일렉트로닉스 엔.브이. Method and system for controlling the output of a luminaire
US7400031B2 (en) 2005-09-19 2008-07-15 International Business Machines Corporation Asymmetrically stressed CMOS FinFET
FR2895679B1 (en) 2005-12-29 2012-06-08 Pf Medicament STABILIZATION OF TESTOSTERONE IN TRANSDERMAL DEVICES
WO2007100614A2 (en) 2006-02-24 2007-09-07 Scidose, Llc STABLE NON-CRYSTALLINE FORMULATION COMPRISING HMG-CoA REDUCTASE INHIBITOR
GB0807605D0 (en) 2008-04-28 2008-06-04 Diurnal Ltd Lipid composition
US11304960B2 (en) 2009-01-08 2022-04-19 Chandrashekar Giliyar Steroidal compositions
US20130225544A1 (en) 2009-01-08 2013-08-29 Lipocine Inc. Lipobalanced long chain testosterone esters for oral delivery
EP2229936B1 (en) 2009-03-09 2015-05-06 PharmaSol GmbH Nanonized testosterone formulations for improved bioavailability
US20110263552A1 (en) 2009-12-31 2011-10-27 Sov Therapeutics Modulation of side effect profile of 5-alpha reductase inhibitor therapy
WO2011082384A2 (en) 2009-12-31 2011-07-07 Differential Drug Development Associates, Llc Modulation of solubility, stability, absorption, metabolism, and pharmacokinetic profile of lipophilic drugs by sterols
WO2011112747A1 (en) 2010-03-09 2011-09-15 Catholic Healthcare West Methods for inhibiting preterm labor and uterine contractility disorders and preventing cervical ripening
ES2525520T3 (en) 2010-04-12 2014-12-26 Clarus Therapeutics, Inc. Oral testosterone ester formulations and methods to treat testosterone deficiency with them
RU2482847C2 (en) 2010-11-17 2013-05-27 Леонид Леонидович Клопотенко Pharmaceutical composition, containing sildenafil or alprostadil, minoxidil or euphyllin, testosteron or yohimbine and liposomes for local application
US20120135074A1 (en) 2010-11-30 2012-05-31 Chandrashekar Giliyar High-Strength Testosterone Undecanoate Compositions
US20140309202A1 (en) 2010-11-30 2014-10-16 Lipocine Inc. High-strength testosterone undecanoate compositions
US9358241B2 (en) 2010-11-30 2016-06-07 Lipocine Inc. High-strength testosterone undecanoate compositions
US9034858B2 (en) 2010-11-30 2015-05-19 Lipocine Inc. High-strength testosterone undecanoate compositions
US20120148675A1 (en) 2010-12-10 2012-06-14 Basawaraj Chickmath Testosterone undecanoate compositions
JO3755B1 (en) 2011-01-26 2021-01-31 Ferring Bv Testosterone formulations
US20130178454A1 (en) 2011-11-17 2013-07-11 Shalender Bhasin Combination of testosterone and ornithine decarboxylase (odc) inhibitors
WO2016033556A1 (en) 2014-08-28 2016-03-03 Lipocine Inc. BIOAVAILABLE SOLID STATE (17-β)-HYDROXY-4-ANDROSTEN-3-ONE ESTERS
RU2017111503A (en) 2014-08-29 2018-10-05 Липосин Инк. COMPOSITIONS (17-β) -3-OXOANDROST-4-EN-17-IL UNDECANOATE AND METHODS FOR PRODUCING AND USING THEM
US20160184321A1 (en) * 2014-09-24 2016-06-30 Lipocine Inc. Compositions and their use in oral dosing regimens
WO2016205423A2 (en) 2015-06-15 2016-12-22 Lipocine Inc. Composition and method for oral delivery of androgen prodrugs
US20170319596A1 (en) * 2015-11-10 2017-11-09 Lipocine Inc. Methods for improving quality of life or sexual domain function and composition useful in the methods
CA3078723A1 (en) * 2016-11-28 2018-05-31 Nachiappan Chidambaram Oral testosterone undecanoate therapy

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11826460B2 (en) 2018-07-23 2023-11-28 Chong Kun Dang Pharmaceutical Corp. Stable pharmaceutical composition comprising testosterone undecanoate

Also Published As

Publication number Publication date
US11433083B2 (en) 2022-09-06
US20190365780A1 (en) 2019-12-05

Similar Documents

Publication Publication Date Title
US10881671B2 (en) High-strength testosterone undecanoate compositions
US9358241B2 (en) High-strength testosterone undecanoate compositions
US20120135074A1 (en) High-Strength Testosterone Undecanoate Compositions
US20140303132A1 (en) Lipobalanced long chain testosterone esters for oral delivery
US20140309202A1 (en) High-strength testosterone undecanoate compositions
US11433083B2 (en) High-strength testosterone undecanoate compositions
CA2942005A1 (en) Lipobalanced long chain testosterone esters for oral delivery
US11931367B2 (en) Oral (17-ß)-3-oxoandrost-4-en-17-yl tridecanoate therapy

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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