US20050142173A1 - Pharmaceutical composition and method for treating hypogonadism - Google Patents

Pharmaceutical composition and method for treating hypogonadism Download PDF

Info

Publication number
US20050142173A1
US20050142173A1 US10/867,445 US86744504A US2005142173A1 US 20050142173 A1 US20050142173 A1 US 20050142173A1 US 86744504 A US86744504 A US 86744504A US 2005142173 A1 US2005142173 A1 US 2005142173A1
Authority
US
United States
Prior art keywords
testosterone
day
subject
serum
composition
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
US10/867,445
Inventor
Robert Dudley
Dominque Drouin
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.)
BESINS INTERNATIONAL-USA Inc
Laboratoires Besins International SAS
Unimed Pharmaceuticals LLC
Original Assignee
BESINS INTERNATIONAL-USA Inc
Unimed Pharmaceuticals LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=24614189&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20050142173(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to US10/867,445 priority Critical patent/US20050142173A1/en
Application filed by BESINS INTERNATIONAL-USA Inc, Unimed Pharmaceuticals LLC filed Critical BESINS INTERNATIONAL-USA Inc
Assigned to UNIMED PHARMACEUTICALS, INC. reassignment UNIMED PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DUDLEY, ROBERT E.
Assigned to BESINS INTERNATIONAL-USA, INC. reassignment BESINS INTERNATIONAL-USA, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: DROUIN, DOMINIQUE
Publication of US20050142173A1 publication Critical patent/US20050142173A1/en
Assigned to UNIMED PHARMACEUTICALS, LLC reassignment UNIMED PHARMACEUTICALS, LLC CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: UNIMED PHARMACEUTICALS, INC.
Assigned to LABORATOIRES BESINS INTERNATIONAL, SAS reassignment LABORATOIRES BESINS INTERNATIONAL, SAS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: UNIMED PHARMACEUTICALS, INC.
Priority to US13/071,276 priority patent/US20110172196A1/en
Priority to US13/071,264 priority patent/US20110201586A1/en
Priority to US13/902,035 priority patent/US20130261097A1/en
Priority to US13/942,245 priority patent/US9132089B2/en
Priority to US13/965,499 priority patent/US9125816B2/en
Priority to US14/800,949 priority patent/US20150313914A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds 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
    • 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
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • 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
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/14Esters of carboxylic acids, e.g. fatty acid monoglycerides, medium-chain triglycerides, parabens or PEG fatty acid esters
    • 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
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/32Macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. carbomers, poly(meth)acrylates, or polyvinyl pyrrolidone
    • 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
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/06Ointments; Bases therefor; Other semi-solid forms, e.g. creams, sticks, gels
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/24Drugs for disorders of the endocrine system of the sex hormones
    • A61P5/26Androgens
    • 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
    • A61K31/5685Compounds 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 having an oxo group in position 17, e.g. androsterone

Definitions

  • the present invention is directed to a pharmaceutical composition comprising testosterone in a gel formulation, and to methods of using the same.
  • Testosterone is the major circulating androgen in men. More than 95% of the 6-7 mg of testosterone produced per day is secreted by the approximately 500 million Leydig cells in the testes. Two hormones produced by the pituitary gland, luteinizing hormone (“LH”) and follicle stimulating hormone (“FSH”), are required for the development and maintenance of testicular function.
  • LH luteinizing hormone
  • FSH follicle stimulating hormone
  • LH The most important hormone for the regulation of Leydig cell number and function is LH.
  • LH secretion from the pituitary is inhibited through a negative-feedback pathway by increased concentrations of testosterone through the inhibition of the release of gonadotropin-releasing hormone (“GRH”) by the hypothalamus.
  • GSH gonadotropin-releasing hormone
  • FSH promotes spermatogenesis and is essential for the normal maturation of sperm.
  • FSH secretion from the pituitary normally is inhibited through a negative-feedback pathway by increased testosterone concentrations.
  • Testosterone circulates in the blood 98% bound to protein. In men, approximately 40% of the binding is to the high-affinity sex hormone binding globulin (“SHBG”). The remaining 60% is bound weakly to albumin. Thus, a number of measurements for testosterone are available from clinical laboratories.
  • the term “free” testosterone as used herein refers to the fraction of testosterone in the blood that is not bound to protein.
  • total testosterone or “testosterone” as used herein means the free testosterone plus protein-bound testosterone.
  • bioavailable testosterone refers to the non-SHBG bound testosterone and includes testosterone weakly bound to albumin.
  • DHT binds with greater affinity to SHBG than does testosterone.
  • the activity of testosterone depends on the reduction to DHT, which binds to cytosol receptor proteins. The steroid-receptor complex is then transported to the nucleus where it initiates transcription and cellular changes related to androgen action. DHT is also thought to lower prostate volume and inhibit tumor development in the prostate.
  • DHT+T total androgen
  • DHT/T ratio a ratio of DHT to testosterone
  • hypogonadism results from a variety of patho-physiological conditions in which testosterone concentration is diminished below the normal range.
  • the hypogonadic condition is sometimes linked with a number of physiological changes, such as diminished interest in sex, impotence, reduced lean body mass, decreased bone density, lowered mood, and energy levels.
  • Primary hypogonadism includes the testicular failure due to congenital or acquired anorchia, XYY Syndrome, XX males, Noonan's Syndrome, gonadal dysgenesis, Leydig cell tumors, maldescended testes, varicocele, Sertoli-Cell-Only Syndrome, cryptorchidism, bilateral torsion, vanishing testis syndrome, orchiectomy, Klinefelter's Syndrome, chemotherapy, toxic damage from alcohol or heavy metals, and general disease (renal failure, liver cirrhosis, diabetes, myotonia dystrophica). Patients with primary hypogonadism show an intact feedback mechanism in that the low serum testosterone concentrations are associated with high FSH and LH concentrations. However, because of testicular or other failures, the high LH concentrations are not effective at stimulating testosterone production.
  • hypogonadism involves an idiopathic gonadotropin or LH-releasing hormone deficiency.
  • This type of hypogonadism includes Kallman's Syndrome, Prader-Labhart-Willi's Syndrome, Laurence-Moon-Biedl's Syndrome, pituitary insufficiency/adenomas, Pasqualini's Syndrome, hemochromatosis, hyperprolactinemia, or pituitary-hypothalamic injury from tumors, trauma, radiation, or obesity. Because patients with secondary hypogonadism do not demonstrate an intact feedback pathway, the lower testosterone concentrations are not associated with increased LH or FSH levels. Thus, these men have low testosterone serum levels but have gonadotropins in the normal to low range.
  • hypogonadism may be age-related. Men experience a slow but continuous decline in average serum testosterone after approximately age 20 to 30 years. researchers estimate that the decline is about 1-2% per year. Cross-sectional studies in men have found that the mean testosterone value at age 80 years is approximately 75% of that at age 30 years. Because the serum concentration of SHBG increases as men age, the fall in bioavailable and free testosterone is even greater than the fall in total testosterone. researchers have estimated that approximately 50% of healthy men between the ages of 50 and 70 have levels of bioavailable testosterone that are below the lower normal limit. Moreover, as men age, the circadian rhythm of testosterone concentration is often muted, dampened, or completely lost. The major problem with aging appears to be within the hypothalamic-pituitary unit.
  • hypogonadism is the most common hormone deficiency in men, affecting 5 in every 1,000 men. At present, it is estimated that only five percent of the estimated four to five million American men of all ages with hypogonadism currently receive testosterone replacement therapy. Thus, for years, researchers have investigated methods of delivering testosterone to men.
  • the present invention is directed to a 1% testosterone hydroalcoholic gel that overcomes the problems associated with current testosterone replacement methods.
  • Subdermal implants have been used as a method of testosterone replacement since the 1940s.
  • the implant is produced by melting crystalline testosterone into a cylindrical form.
  • pellet implants are manufactured to contain either 100 mg (length 6 mm, surface area 1172) or 200 mg of testosterone (length 12 mm, surface area 202 mm2).
  • Patients receive dosages ranging from 100 to 1,200 mg, depending on the individual's requirements.
  • the implants are inserted subcutaneously either by using a trocar and cannula or by open surgery into an area where there is relatively little movement. Frequently, the implant is placed in the lower abdominal wall or the buttock. Insertion is made under local anesthesia, and the wound is closed with an adhesive dressing or a fine suture.
  • Implants have several major drawbacks.
  • subdermal testosterone implants produce supra-physiologically high serum testosterone levels which slowly decline so that before the next injection subnormally low levels of testosterone are reached. For example, in one recent pharmacokinetic study, hypogonadal patients who received six implants (1,200 mg testosterone) showed an initial short-lived burst release of testosterone within the first two days after application.
  • testosterone esters such as enanthate, cypionate
  • testosterone esters such as enanthate, cypionate
  • More recent studies have involved injection of testosterone buciclate or testosterone undecanoate in an oil-based vehicle.
  • Other researchers have injected testosterone microcapsule formulations.
  • Testosterone ester injection treatments suffer from many problems. Patients receiving injection therapy often complain that the delivery mechanism is painful and causes local skin reactions.
  • testosterone microcapsule treatment requires two simultaneous intramuscular injections of a relatively large volume, which may be difficult to administer due to the high viscosity of the solution and the tendency to block the needle.
  • Other men generally find testosterone injection therapy inconvenient because injection usually requires the patient to visit his physician every two to three weeks.
  • testosterone replacement treatments still create an undesirable pharmacokinetic profile.
  • the profile generally shows a supra-physiologic testosterone concentration during the first 24 to 48 hours followed by a gradual fall often to sub-physiologic levels over the next few weeks.
  • These high serum testosterone levels paralleled by increases in E2, are also considered the reason for acne and gynecomastia occurring in some patients, and for polycythaemia, occasionally encountered especially in older patients using injectable testosterone esters.
  • testosterone buciclate injections the treatment barely provides normal androgen serum levels and the maximal increase of serum testosterone over baseline does not exceed 172 ng/dL (6 nmol/dL) on average. Because libido, potency, mood, and energy are thought to fluctuate with the serum testosterone level, testosterone injections have largely been unsuccessful in influencing these variables. Thus, testosterone injection remains an undesirable testosterone replacement treatment method.
  • TESTODERM® (Alza Pharmaceuticals, Mountain View, Calif.) was the first testosterone-containing patch developed.
  • the TESTODERM® patch is currently available in two sizes (40 or 60 cm2).
  • the patch contains 10 or 15 mg of testosterone and delivers 4.0 mg or 6.0 mg of testosterone per day.
  • TESTODERM® is placed on shaved scrotal skin, aided by application of heat for a few seconds from a hair dryer.
  • FIG. 2 shows a typical pharmacokinetic testosterone profile for both the 40 cm2 and 60 cm2 patch. Studies have also shown that after two to four weeks of continuous daily use, the average plasma concentration of DHT and DHT/T increased four to five times above normal. The high serum DHT levels are presumably caused by the increased metabolism of 5 ⁇ -reductase in the scrotal skin.
  • the most recently developed non-scrotal patch is TESTODERM® TTS (Alza Pharmaceuticals, Mountain View, Calif.). It is an occlusive patch applied once daily to the arm, back, or upper buttocks.
  • the system is comprised of a flexible backing of transparent polyester/ethylene-vinyl acetate copolymer film, a drug reservoir of testosterone, and an ethylene-vinyl acetate copolymer membrane coated with a layer of polyisobutylene adhesive formulation.
  • a protective liner of silicone-coated polyester covers the adhesive surface.
  • the typical 24-hour steady state testosterone concentration achieved with TESTODERM® TTS patch is shown in FIG. 3 .
  • TESTODERM® patch Because of TESTODERM® patch is applied to the scrotal skin while the TESTODERM TTS® patch is applied to non-scrotal skin, the two patches provide different steady-state concentrations of the two major testosterone metabolites, DTH and E2: TABLE 4 Hormone Levels Using TESTODERM ® and TESTODERM ® TTS Hormone Placebo TESTODERM ® TESTODERM ® TTS DHT (ng/dL) 11 134 38 E 2 (pg/ml) 3.8 10 21.4
  • TESTODERM TTS® treatment creates a DHT/T ratio that is not different from that of a placebo treatment. Both systems, however, suffer from similar problems. In clinical studies, TESTODERM® TTS is associated with transient itching in 12% of patients, erythema in 3% of patients, and puritus in 2% of patients. Moreover, in one 14-day study, 42% of patients reported three or more detachments, 33% of which occurred during exercise.
  • ANDRODERM® (Watson Laboratories, Inc., Corona, Calif.) is a testosterone-containing patch applied to non-scrotal skin.
  • the circular patch has a total surface area of 37 cm.2
  • the patch consists of a liquid reservoir containing 12.2 mg of testosterone and a permeation-enhanced vehicle containing ethanol, water, monoglycerides, fatty acid esters, and gelling agents.
  • the suggested dose of two patches, applied each night in a rotating manner on the back, abdomen, upper arm, or thigh, delivers 4.1 to 6.8 mg of testosterone.
  • ANDRODERM® is associated with skin irritation in about a third of the patients, and 10% to 15% of subjects have been reported to discontinue the treatment because of chronic skin irritation.
  • Preapplication of corticosteroid cream at the site of application of ANDRODERM® has been reported to decrease the incidence and severity of the skin irritation.
  • Patches were variously described as noisy, visually indiscrete, embarrassing, unpleasant to apply and remove, and generally to be socially unacceptable. They fell off in swimming pools and showers, attracted ribald comments from sporting partners, and left bald red marks over trunk and limbs. Dogs, wife, and children were distracted by noise of the patches with body movements. Those with poor mobility or manual dexterity (and several were over 70 years of age) found it difficult to remove packaging an apply patches dorsally.
  • the transdermal patch generally offers an improved pharmacokinetic profile compared to other currently used testosterone delivery mechanisms.
  • the clinical and survey data shows that all of these patches suffer from significant drawbacks, such as buritus, burn-like blisters, and erythema.
  • the adverse effects associated with transdermal patch systems are “substantially higher” than reported in clinical trials. See Parker, supra.
  • the transdermal patch still remains an inadequate testosterone replacement therapy alternative for most men.
  • DHT-gel results in decreased serum testosterone, E2, LH, and FSH levels.
  • DHT gels are not effective at increasing testosterone levels in hypogonadal men. Accordingly, there is a definite need for a testosterone formulation that safely and effectively provides an optimal and predictable pharmacokinetic profile.
  • the present invention generally comprises a testosterone gel.
  • Daily transdermal application of the gel in hypogonadal men results in a unique pharmacokinetic steady-state profile for testosterone.
  • Long-term treatment further results in, for example, increased bone mineral density, enhanced libido, enhanced erectile frequency and satisfaction, increased positive mood, increased muscle strength, and improved body composition without significant skin irritation.
  • the present invention is also directed to a unique method of administering the testosterone gel employing a packet having a polyethylene liner compatible with the components of the gel.
  • FIG. 1 is a graph of testosterone concentrations, DHT concentrations, and the DHT/T ratio for patients receiving a subdermal testosterone pellet implant over a period of 300 days after implantation.
  • FIG. 2 shows a typical pharmacokinetic testosterone profile for both the 40 cm2 and 60 cm2 patch. Studies have also shown that after two to four weeks of continuous daily use, the average plasma concentration of DHT and DHT/T increased four to five times above normal. The high serum DHT levels are presumably caused by the increased metabolism of 5 ⁇ -reductase in the scrotal skin.
  • FIG. 3 is a 24-hour testosterone pharmacokinetic profile for patients receiving the TESTODERM® TTS patch.
  • FIG. 4 is a 24-hour testosterone pharmacokinetic profile for patients receiving the ANDRODERM® patch.
  • FIG. 5 ( a ) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men prior to receiving 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 5 ( b ) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men on the first day of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 5 ( c ) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men on day 30 of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel, or the testosterone patch (by initial treatment group).
  • FIG. 5 ( d ) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men on day 90 of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 5 ( e ) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men on day 180 of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by final treatment group).
  • FIG. 5 ( f ) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men on day 0, 1, 30, 90, and 180 of treatment with 5.0 g/day of AndroGel®.
  • FIG. 5 ( g ) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men on day 0, 1, 30, 90, and 180 of treatment with 10.0 g/day of AndroGel®.
  • FIG. 5 ( h ) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men on day 0, 1, 30, 90, and 180 of treatment with the testosterone patch.
  • FIG. 6 ( a ) is a graph showing the 24-hour free testosterone pharmacokinetic profile for hypogonadal men on day 1 of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 6 ( b ) is a graph showing the 24-hour free testosterone pharmacokinetic profile for hypogonadal men on day 30 of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 6 ( c ) is a graph showing the 24-hour free testosterone pharmacokinetic profile for hypogonadal men on day 90 of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 6 ( d ) is a graph showing the 24-hour free testosterone pharmacokinetic profile for hypogonadal men on day 180 of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by final treatment group).
  • FIG. 6 ( e ) is a graph showing the 24-hour free testosterone pharmacokinetic profile for hypogonadal men on day 0, 1, 30, 90, and 180 of treatment with 5.0 g/day of AndroGel®.
  • FIG. 6 ( f ) is a graph showing the 24-hour free testosterone pharmacokinetic profile for hypogonadal men on day 0, 1, 30, 90, and 180 of treatment with 10.0 g/day of AndroGel®.
  • FIG. 6 ( g ) is a graph showing the 24-hour free testosterone pharmacokinetic profile for hypogonadal men on day 0, 1, 30, 90, and 180 of treatment with the testosterone patch.
  • FIG. 7 is a graph showing the DHT concentrations on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 8 is a graph showing the DHT/T ratio on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 9 is a graph showing the total androgen concentrations (DHT+T) on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 10 is a graph showing the E2 concentrations on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 11 is a graph showing the SHBG concentrations on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 12 ( a ) is a graph showing the FSH concentrations on days 0 through 180 for men having primary hypogonadism and receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 12 ( b ) is a graph showing the FSH concentrations on days 0 through 180 for men having secondary hypogonadism and receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 12 ( c ) is a graph showing the FSH concentrations on days 0 through 180 for men having age-associated hypogonadism and receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 12 ( d ) is a graph showing the FSH concentrations on days 0 through 180 for men having hypogonadism of an unknown origin and receiving either 5.0 g/day of AndroGel®, 10.0of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 13 ( a ) is a graph showing the LH concentrations on days 0 through 180 for men having primary hypogonadism and receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 13 ( b ) is a graph showing the LH concentrations on days 0 through 180 for men having secondary hypogonadism and receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 13 ( c ) is a graph showing the LH concentrations on days 0 through 180 for men having age-associated hypogonadism and receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 13 ( d ) is a graph showing the LH concentrations on days 0 through 180 for men having hypogonadism of an unknown origin and receiving either 5.0 g/day of AndroGel®, 10.0 of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 14 ( a ) is a bar graph showing the change in hip BMD for hypogonadal men after 180 days of treatment with 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 14 ( b ) is a bar graph showing the change in spine BMD for hypogonadal men after 180 days of treatment with 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 15 is a graph showing PTH concentrations on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 16 is a graph showing SALP concentrations on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 17 is a graph showing the osteocalcin concentrations on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 18 is a graph showing the type I procollagen concentrations on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 19 is a graph showing the N-telopeptide/Cr ratio on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 20 is a graph showing the Ca/Cr ratio on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 21 ( a ) is a graph showing sexual motivation scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 21 ( b ) is a graph showing overall sexual desire scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 21 ( c ) is a graph showing sexual enjoyment (with a partner) scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 22 ( a ) is a graph showing sexual performance scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 22 ( b ) is a graph showing erection satisfaction performance scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 22 ( c ) is a graph showing percent erection scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 23 ( a ) is a graph showing positive mood scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 23 ( b ) is a graph showing negative mood scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 24 ( a ) is a bar graph showing the change in leg strength on days 90 and 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 24 ( b ) is a bar graph showing the change in arm strength on days 90 and 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 25 ( a ) is a bar graph showing the change in total body mass on days 90 and 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 25 ( b ) is a bar graph showing the change in lean body mass on days 90 and 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 25 ( c ) is a bar graph showing the change in fat mass on days 90 and 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 25 ( d ) is a bar graph showing the change in percent body fat on days 90 and 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • the present invention is directed to a pharmaceutical composition for percutaneous administration comprising at least one active pharmaceutical ingredient (e.g., testosterone) in a hydroalcoholic gel.
  • the active ingredients employed in the composition may include anabolic steroids such as androisoxazole, bolasterone, clostebol, ethylestrenol, formyldienolone, 4-hydroxy-19-nortestosterone, methenolone, methyltrienolone, nandrolone, oxymesterone, quinbolone, stenbolone, trenbolone; androgenic steroids such as boldenone, fluoxymesterone, mestanolone, mesterolone, methandrostenolone, 17-methyltestosterone, 17 Alpha-methyl-testosterone 3-cyclopentyl enol ether, norethandrolone, normethandrone, oxandrolone, oxymetholone, prasterone, stanlolone, stanozol
  • the gel comprises one or more lower alcohols, such as ethanol or isopropanol; a penetration enhancing agent; a thickener; and water.
  • the present invention may optionally include salts, emollients, stabilizers, antimicrobials, fragrances, and propellants.
  • a “penetration enhancer” is an agent known to accelerate the delivery of the drug through the skin.
  • These agents also have been referred to as accelerants, adjuvants, and absorption promoters, and are collectively referred to herein as “enhancers.”
  • This class of agents includes those with diverse mechanisms of action including those which have the function of improving the solubility and diffusibility of the drug, and those which improve percutaneous absorption by changing the ability of the stratum corneum to retain moisture, softening the skin, improving the skin's permeability, acting as penetration assistants or hair-follicle openers or changing the state of the skin such as the boundary layer.
  • the penetration enhancer of the present invention is a functional derivative of a fatty acid, which includes isosteric modifications of fatty acids or non-acidic derivatives of the carboxylic functional group of a fatty acid or isosteric modifications thereof.
  • the functional derivative of a fatty acid is an unsaturated alkanoic acid in which the COOH group is substituted with a functional derivative thereof, such as alcohols, polyols, amides and substituted derivatives thereof.
  • fatty acid means a fatty acid that has four (4) to twenty-four (24) carbon atoms.
  • Non-limiting examples of penetration enhancers include C8-C22 fatty acids such as isostearic acid, octanoic acid, and oleic acid; C8-C22 fatty alcohols such as oleyl alcohol and lauryl alcohol; lower alkyl esters of C8-C22 fatty acids such as ethyl oleate, isopropyl myristate, butyl stearate, and methyl laurate; di(lower)alkyl esters of C6-C8 diacids such as diisopropyl adipate; monoglycerides of C8-C22 fatty acids such as glyceryl monolaurate; tetrahydrofurfiuryl alcohol polyethylene glycol ether; polyethylene glycol, propylene glycol; 2-(2-ethoxyethoxy) ethanol; diethylene glycol monomethyl ether; alkylaryl ethers of polyethylene oxide; polyethylene oxide monomethyl ethers; polyethylene oxide dimethyl
  • the thickeners used herein may include anionic polymers such as polyacrylic acid (CARBOPOL® by B.F. Goodrich Specialty Polymers and Chemicals Division of Cleveland, Ohio), carboxymethylcellulose and the like. Additional thickeners, enhancers and adjuvants may generally be found in United States Pharmacopeia/National Formulary (2000); Remington's The Science and Practice of Pharmacy, Meade Publishing Co.
  • anionic polymers such as polyacrylic acid (CARBOPOL® by B.F. Goodrich Specialty Polymers and Chemicals Division of Cleveland, Ohio), carboxymethylcellulose and the like. Additional thickeners, enhancers and adjuvants may generally be found in United States Pharmacopeia/National Formulary (2000); Remington's The Science and Practice of Pharmacy, Meade Publishing Co.
  • the amount of drug to be incorporated in the composition varies depending on the particular drug, the desired therapeutic effect, and the time span for which the gel is to provide a therapeutic effect.
  • the composition is used in a “pharmacologically effective amount.” This means that the concentration of the drug is such that in the composition it results in a therapeutic level of drug delivered over the term that the gel is to be used. Such delivery is dependent on a number of variables including the drug, the form of drug, the time period for which the individual dosage unit is to be used, the flux rate of the drug from the gel, surface area of application site, etc.
  • the amount of drug necessary can be experimentally determined based on the flux rate of the drug through the gel, and through the skin when used with and without enhancers.
  • the gel is comprised of the following substances in approximate amounts: TABLE 5 Composition of AndroGel ® AMOUNT (w/w) PER 100 g OF SUBSTANCE GEL Testosterone 1.0 g Carbopol 980 0.90 g Isopropyl myristate 0.50 g 0.1 N NaOH 4.72 g Ethanol (95% w/w) 72.5 g* Purified water (qsf) 100 g *Corresponding to 67 g of ethanol.
  • the constituents of this formulation may be varied in amounts yet continue to be within the spirit and scope of the present invention.
  • the composition may contain about 0.1 to about 10.0 g of testosterone, about 0.1 to about 5.0 g Carbopol, about 0.1 to about 5.0 g isopropyl myristate, and about 30.0 to about 98.0 g ethanol; or about 0.1% to about 10.0% of testosterone, about 0.1% to about 5.0% Carbopol, about 0.1% to about 5.0% isopropyl myristate, and about 30.0% to about 98.0% ethanol, on a weight to weight basis of the composition.
  • a therapeutically effective amount of the gel is rubbed onto a given area of skin by the user.
  • the combination of the lipophilic testosterone with the hydroalcoholic gel helps drive the testosterone in to the outer layers of the skin where it is absorbed and then slowly released into the blood stream.
  • the administration of the gel of the present invention has a sustained effect.
  • Toxicity and therapeutic efficacy of the active ingredients can be determined by standard pharmaceutical procedures, e.g., for determining LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50.
  • Compounds which exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • treatment refers to any treatment of a human condition or disease and includes: (1) preventing the disease or condition from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it, (2) inhibiting the disease or condition, i.e., arresting its development, (3) relieving the disease or condition, i.e., causing regression of the condition, or (4) relieving the conditions caused by the disease, i.e., stopping the symptoms of the disease.
  • composition and method of the present invention may be used to treat these disorders in humans and animals of any kind, such as dogs, pigs, sheep, horses, cows, cats, zoo animals, and other commercially bred farm animals.
  • One embodiment of the present invention involves the transdermal application of AndroGel® as a method of treating male hypogonadism.
  • application of the gel results in a unique pharmacokinetic profile for testosterone, as well as concomitant modulation of several other sex hormones.
  • Application of the testosterone gel to hypogonadal male subjects also results in (1) increased bone mineral density, (2) enhanced libido, (3) enhanced erectile capability and satisfaction, (4) increased positive mood, (5) increased muscle strength, and (6) better body composition, such increased total body lean mass and decreased total body fat mass.
  • the gel is not generally associated with significant skin irritation.
  • hypogonadal men were recruited and studied in 16 centers in the United States.
  • the patients were between 19 and 68 years and had single morning serum testosterone levels at screening of less than or equal to 300 ng/dL (10.4 nmol/L).
  • a total of 227 patients were enrolled: 73, 78, and 76 were randomized to receive 5.0 g/day of AndroGel® (delivering 50 mg/day of testosterone to the skin of which about 10% or 5 mg is absorbed), 10.0 g/day of AndroGel® (delivering 100 mg/day of testosterone to the skin of which about 10% or 10 mg is absorbed), or the ANDRODERM® testosterone patch (“T patch”) (delivering 50 mg/day of testosterone), respectively.
  • the randomized, multi-center, parallel study compared two doses of AndroGel® with the ANDRODERM® testosterone patch.
  • the study was double-blind with respect to the AndroGel® dose and open-labeled for the testosterone patch group.
  • the subjects were randomized to receive 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or two non-scrotal patches.
  • the subjects were administered one of the following treatments: 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, or two non-scrotal patches.
  • Patients who were applying AndroGel® had a single, pre-application serum testosterone measured on day 60 and, if the levels were within the normal range of 300 to 1,000 ng/dL (10.4 to 34.7 nmol/L), then they remained on their original dose.
  • AndroGel® groups based on the pre-application serum testosterone levels on day 60.
  • Twenty subjects in the 5.0 g/day AndroGel® group had the dose increased to 7.5 g/day.
  • Twenty patients in the 10.0 g/day AndroGel® group had the AndroGel® dose reduced to 7.5 g/day.
  • There were three patients in the testosterone patch group who were switched to 5.0 g/day AndroGel® because of patch intolerance.
  • One 10.0 g/day AndroGel® subject was adjusted to receive 5.0 g/day and one 5.0 AndroGel® subject had the dose adjusted to 2.5 g/day.
  • the number of subjects enrolled into day 91 to 180 of the study thus consisted of 51 receiving 5.0 g/day of AndroGel®, 40 receiving 7.5 g/day of AndroGel®, 52 receiving 10.0 g/day of AndroGel®, and 52 continuing on the ANDRODERM® patch.
  • the treatment groups in this example may thus be characterized in two ways, either by “initial” or by the “final” treatment group.
  • Fasting blood samples for calcium, inorganic phosphorus, parathyroid hormone (“PTH”), osteocalcin, type I procollagen, and skeletal specific alkaline phosphatase (“SALP”) were collected on days 0, 30, 90, 120, and 180.
  • PTH parathyroid hormone
  • SALP skeletal specific alkaline phosphatase
  • N-telopeptide a fasting two-hour timed urine collection for urine creatinine, calcium, and typecollagen cross-linked N-telopeptides
  • AndroGel® Approximately 250 g of AndroGel® was packaged in multidose glass bottles that delivered 2.25 g of the gel for each actuation of the pump. Patients assigned to apply 5.0 g/day of AndroGel® testosterone were given one bottle of AndroGel® and one bottle of placebo gel (containing vehicle but no testosterone), while those assigned to receive 10.0 g/day of AndroGel® were dispensed two bottles of the active AndroGel®. The patients were then instructed to apply the bottle contents to the right and left upper arms/shoulders and to the right and left sides of the abdomen on an alternate basis.
  • the 7.5 g/day AndroGel® group received their dose in an open-label fashion. After 90 days, for the subjects titrated to the AndroGel® 7.5 g/day dose, the patients were supplied with three bottles, one containing placebo and the other two AndroGel®. The subjects were instructed to apply one actuation from the placebo bottle and three actuations from a AndroGel® bottle to four different sites of the body as above. The sites were rotated each day taking the same sequence as described above.
  • ANDRODERM® testosterone patches each delivering 2.5 mg/day of testosterone were provided to about one-third of the patients in the study. These patients were instructed to apply two testosterone patches to a clean, dry area of skin on the back, abdomen, upper arms, or thighs once per day. Application sites were rotated with approximately seven days interval between applications to the same site.
  • the gel/patches were applied following pre-dose evaluations. On the remaining days, the testosterone gel or patches were applied at approximately 8:00 a.m. for 180 days.
  • the patients had multiple blood samples for testosterone and free testosterone measurements at 30, 15 and 0 minutes before and 2, 4, 8, 12, 16, and 24 hours after AndroGel® or patch application.
  • subjects returned on days 60, 120, and 150 for a single blood sampling prior to application of the gel or patch.
  • Serum DHT, E2, FSH, LH and SHBG were measured on samples collected before gel application on days 0, 30, 60, 90, 120, 150, and 180.
  • Sera for all hormones were stored frozen at 20° C. until assay. All samples for a patient for each hormone were measured in the same assay whenever possible. The hormone assays were then measured at the Endocrine Research Laboratory of the UCLA-Harbor Medical Center.
  • Serum testosterone levels were measured after extraction with ethylacetate and hexane by a specific radioimmunoassay (“RIA”) using reagents from ICN (Costa Mesa, Calif.).
  • the cross reactivities of the antiserum used in the testosterone RIA were 2.0% for DHT, 2.3% for androstenedione, 0.8% for 3-Beta-androstanediol, 0.6% for etiocholanolone and less than 0.01% for all other steroids tested.
  • the lower limit of quantitation (“LLQ”) for serum testosterone measured by this assay was 25 ng/dL (0.87 nmol/L).
  • the mean accuracy of the testosterone assay determined by spiking steroid free serum with varying amounts of testosterone (0.9to 52 nmol/L), was 104% and ranged from 92% to 117%.
  • the intra-assay and inter-assay coefficients of the testosterone assay were 7.3 and 11.1%, respectively, at the normal adult male range. In normal adult men, testosterone concentrations range from 298 to 1,043 ng/dL (10.33 to 36.17 nmol/L) as determined at the UCLA-Harbor Medical Center.
  • FIG. 5 ( a ) shows that the mean testosterone levels had a maximum level between 8 to 10 a.m. (i.e., at 0 to 2 hours) and the minimum 8 to 12 hours later, demonstrating a mild diurnal variation of serum testosterone.
  • FIG. 5 ( b ) and Tables 8(c)-(d) show the pharmacokinetic profile for all three initial treatment groups after the first application of transdermal testosterone.
  • treatment with AndroGel® and the testosterone patch produced increases in testosterone concentrations sufficiently large to bring the patients into the normal range in just a few hours.
  • the pharmacokinetic profiles were markedly different in the AndroGel® and patch groups. Serum testosterone rose most rapidly in the testosterone patch group reaching a maximum concentration (Cmax) at about 12 hours (Tmax).
  • FIGS. 5 ( c ) and 5 ( d ) show the unique 24-hour pharmacokinetic profile of AndroGel®-treated patients on days 30 and 90.
  • serum testosterone levels showed small and variable increases shortly after dosing. The levels then returned to a relatively constant level.
  • the testosterone patch group patients exhibited a rise over the first 8 to 12 hours, a plateau for another 8 hours, and then a decline to the baseline of the prior day.
  • the Cavg in the 10.0 g/day AndroGel® group was 1.4 fold higher than in the 5.0 g/day AndroGel® group and 1.9 fold higher than the testosterone patch group.
  • the testosterone patch group also had a Cmin substantially below the lower limit of the normal range.
  • the accumulation ratio was 0.94 for testosterone patch group, showing no accumulation.
  • the accumulation ratios at 1.54 and 1.9 were significantly higher in the 5.0 g/day AndroGel® group and 10.0 g/day AndroGel® group, respectively.
  • the differences in accumulation ratio among the groups persisted on day 90. This data indicates that the AndroGel® preparations had a longer effective half-life than testosterone patch.
  • FIG. 5 ( e ) shows the 24-hour pharmacokinetic profile for the treatment groups on day 180.
  • Table 8(e) shows, the serum testosterone concentrations achieved and the pharmacokinetic parameters were similar to those on days 30 and 90 in those patients who continued on their initial randomized treatment groups.
  • Table 8(f) shows that the patients titrated to the 7.5 g/day AndroGel® group were not homogeneous. The patients that were previously in the 10.0 g/day group tended to have higher serum testosterone levels than those previously receiving 5.0 g/day.
  • the Cavg in the patients in the 10.0 g/day group who converted to 7.5 g/day on day 90 was 744 ng/dL, which was 1.7 fold higher than the Cavg of 450in the patients titrated to 7.5 g/day from 5.0 g/day.
  • the Cavg remained lower than those remaining in the 5.0 group.
  • the Cavg became similar to those achieved by patients remaining in the 10.0 g/day group without dose titration.
  • FIGS. 5 ( f )-( h ) compare the pharmacokinetic profiles for the 5.0 g/day AndroGel® group, the 10.0 g/day AndroGel® g/day group, and the testosterone patch group at days 0, 1, 30, 90, and 180, respectively.
  • the mean serum testosterone levels in the testosterone patch group remained at the lower limit of the normal range throughout the treatment period.
  • the mean serum testosterone levels remained at about 490-570 ng/dL for the 5.0 g/day AndroGel® group and about 630-860 ng/dL AndroGel® for the 10.0 g/day group.
  • Table 8(g) shows the increase in AUC0-24 on days 30, 90, and 180 from the pretreatment baseline (net AUC0-24).
  • the bioequivalence assessment was performed on the log-transformed AUCs using “treatment” as the only factor.
  • the AUCs were compared after subtracting away the AUC contribution from the endogenous secretion of testosterone (the AUC on day 0) and adjusting for the two-fold difference in applied doses.
  • the AUC ratio on day 30 was 0.95 (90% C.I.: 0.75-1.19) and on day 90 was 0.92 (90% C.I.: 0.73-1.17). When the day 30 and day 90 data was combined, the AUC ratio was 0.93 (90% C.I.: 0.79-1.10).
  • the data shows dose proportionality for AndroGel® treatment.
  • the geometric mean for the increase in AUC0-24 from day 0 to day 30 or day 90 was twice as great for the 10.0 g/day group as for the 5.0 g/day group.
  • a 125 ng/dL mean increase in serum testosterone Cavg level was produced by each 2.5 g/day of AndroGel®.
  • the data shows that 0.1 g/day of AndroGel® produced, on the average, a 5 ng/dL increase in serum testosterone concentration. This dose proportionality aids dosing adjustment by the physician.
  • each 2.5 g packet will produce, on average, a 125 ng/dL increase in the Cavg for serum total testosterone.
  • Serum free testosterone was measured by RIA of the dialysate, after an overnight equilibrium dialysis, using the same RIA reagents as the testosterone assay.
  • the LLQ of serum free testosterone, using the equilibrium dialysis method, was estimated to be 22 pmol/L.
  • steroid free serum was spiked with increasing doses of testosterone in the adult male range, increasing amounts of free testosterone were recovered with a coefficient of variation that ranged from 11.0-18.5%.
  • the intra- and interassay coefficients of free testosterone were 15% and 16.8% for adult normal male values, respectively.
  • free testosterone concentrations range from 3.48-17.9 ng/dL (121-620 pmol/L) in normal adult men.
  • FIG. 6 ( a ) shows the 24-hour pharmacokinetic profiles for the three treatment groups on day 1.
  • the serum free testosterone levels peaked at 12 about 4 hours earlier than those achieved by the AndroGel® groups
  • the serum free testosterone levels then declined in the testosterone patch group whereas in the AndroGel® groups, the serum free testosterone levels continued to rise.
  • FIGS. 6 ( b ) and 6 ( c ) show the pharmacokinetic profiles of free testosterone in the AndroGel®-treated groups resembled the unique testosterone profiles on days 30 and 90.
  • the mean serum free testosterone levels in the three groups were within normal range. Similar to the total testosterone results, the free testosterone Cavg achieved by the 10.0 g/day group was 1.4 fold higher than the 5.0 g/day group and 1.7 fold higher than the testosterone patch group.
  • the accumulation ratio for the testosterone patch was significantly less than that of the 5.0 g/day AndroGel® group and the 10.0 g/day AndroGel® group.
  • FIG. 6 ( d ) shows the free testosterone concentrations by final treatment groups on day 180.
  • the free testosterone concentrations exhibited a similar pattern as serum testosterone.
  • the 24-hour pharmacokinetic parameters were similar to those on days 30 and 90 in those subjects who remained in the three original randomized groups. Again, in the subjects titrated to receive 7.5 g/day of AndroGel®, the group was not homogenous.
  • the free testosterone Cavg in the patients with doses adjusted upwards from 5.0 to 7.5 g/day remained 29% lower than those of subjects remaining in the 5.0 g/day group.
  • the free testosterone Cavg in the patients whose doses were decreased from 10.0 to 7.5 g/day was 11% higher than those in remaining in the 10.0 g/day group.
  • FIGS. 6 ( e )-( g ) show the free testosterone concentrations in the three groups of subjects throughout the 180-day treatment period. Again, the free testosterone levels followed that of testosterone. The mean free testosterone levels in all three groups were within the normal range with the 10.0 g/day group maintaining higher free testosterone levels than both the 5.0 g/day and the testosterone patch groups.
  • Serum DHT was measured by RIA after potassium permanganate treatment of the sample followed by extraction.
  • the methods and reagents of the DHT assay were provided by DSL (Webster, Tex.).
  • the cross reactivities of the antiserum used in the RIA for DHT were 6.5% for 3- ⁇ -androstanediol, 1.2% for 3- ⁇ -androstanediol, 0.4% for 3- ⁇ -androstanediol glucuronide, and 0.4% for testosterone (after potassium permanganate treatment and extraction), and less than 0.01% for other steroids tested.
  • the intra-assay and inter-assay coefficients of variation for the DHT assay were 7.8 and 16.6%, respectively, for the normal adult male range.
  • the normal adult male range of DHT was 30.7-193.2 ng/dL (1.06 to 6.66 nmol/L) as determined by the UCLA-Harbor Medical Center.
  • the pretreatment mean serum DHT concentrations were between 36 and 42 ng/dL, which were near the lower limit of the normal range in all three initial treatment groups. None of the patients had DHT concentrations above the upper limit of the normal range on the pretreatment day, although almost half (103 patients) had concentrations less than the lower limit.
  • FIG. 7 shows that after treatment, the differences between the mean DHT concentrations associated with the different treatment groups were statistically significant, with patients receiving AndroGel® having a higher mean DHT concentration than the patients using the patch and showing dose-dependence in the mean serum DHT concentrations. Specifically, after testosterone patch application mean serum DHT levels rose to about 1.3 fold above the baseline. In contrast, serum DHT increased to 3.6 and 4.8 fold above baseline after application of 5.0 and 10.0 g/day of AndroGel®, respectively.
  • the increase in DHT concentrations are likely attributed to the concentration and location of 5 ⁇ -reductase in the skin.
  • the large amounts of 5 ⁇ -reductase in the scrotal skin presumably causes an increase in DHT concentrations in the TESTODERM® patch.
  • the ANDRODERM® and TESTODERM TTS® patches create little change in DTH levels because the surface area of the patch is small and little 5 ⁇ -reductase is located in nonscrotal skin.
  • AndroGel® presumably causes an increase in DHT levels because the gel is applied to a relatively large skin area and thus exposes testosterone to greater amounts of the enzyme.
  • the UCLA-Harbor Medical Center reports a DHT/T ratio of 0.052-0.328 for normal adult men.
  • the mean ratios for all three treatments were within the normal range on day 0.
  • the AndroGel® treatment groups showed the largest increase in DHT/T ratio.
  • the mean ratios for all of the treatment groups remained within the normal range on all observation days.
  • the UCLA-Harbor Medical Center has determined that the normal total androgen concentration is 372 to 1,350 ng/dL. As shown in FIG. 9 and Table 12, the mean pre-dose total androgen concentrations for all three treatments were below the lower limit of the normal range on pretreatment day 0. The total androgen concentrations for both AndroGel® groups were within the normal range on all treatment observation days. In contrast, the mean concentrations for patients receiving the testosterone patch was barely within the normal range on day 60 and 120, but were below the lower normal limit on days 30, 90, 150, and 180.
  • Serum E2 levels were measured by a direct assay without extraction with reagents from ICN (Costa Mesa, Calif.). The intra-assay and inter-assay coefficients of variation of E2 were 6.5 and 7.1% respectively.
  • the UCLA-Harbor Medical Center reported an average E2 concentration ranging from 7.1 to 46.1 pg/mL (63 to 169 pmol/L) for normal adult male range. The LLQ of the E2 was 18 pmol/L.
  • the cross reactivities of the E2 antibody were 6.9% for estrone, 0.4% for equilenin, and less than 0.01% for all other steroids tested.
  • the accuracy of the E2 assay was assessed by spiking steroid free serum with increasing amount of E2 (18 to 275 pmol/L). The mean recovery of E2 compared to the amount added was 99.1 % and ranged from 95 to 101 %.
  • FIG. 10 depicts the E2 concentrations throughout the 180-day study.
  • the pretreatment mean E2 concentrations for all three treatment groups were 23-24 pg/mL.
  • the E2 levels increased by an average 9.2% in the testosterone patch during the treatment period, 30.9% in the 5.0 g/day AndroGel® group, and 45.5% in the 10.0 g/day AndroGel® group. All of the mean concentrations fell within the normal range.
  • E2 is believed to be important for the maintenance of normal bone. In addition, E2 has a positive effect on serum lipid profiles.
  • Serum SHBG levels were measured with a fluoroimmunometric assay (“FIA”) obtained from Delfia (Wallac, Gaithersberg, Md.). The intra- and interassay coefficients were 5% and 12% respectively. The LLQ was 0.5 nmol/L. The UCLA-Harbor Medical Center determined that the adult normal male range for the SHBG assay is 0.8 to 46.6 nmol/L.
  • FAA fluoroimmunometric assay
  • the serum SHBG levels were similar and within the normal adult male range in the three treatment groups at baseline. None of the treatment groups showed major changes from the baseline on any of the treatment visit days. After testosterone replacement, serum SHBG levels showed a small decrease in all three groups. The most marked change occurred in the 10.0 g/day AndroGel® group.
  • Serum FSH and LH were measured by highly sensitive and specific solid-phase FIA assays with reagents provided by Delfia (Wallac, Gaithersburg, Md.).
  • the intra-assay coefficient of variations for LH and FSH fluroimmunometric assays were 4.3 and 5.2%, respectively; and the interassay variations for LH and FSH were 11.0% and 12.0%, respectively.
  • the LLQ was determined to be 0.2 IU/L. All samples obtained from the same subject were measured in the same assay.
  • the UCLA-Harbor Medical Center reports that the adult normal male range for LH is 1.0-8.1 U/L and for FSH is 1.0-6.9U/L.
  • Table 15(a)-(d) shows the concentrations of FSH throughout the 180-day treatment depending on the cause of hypogonadism: (1) primary, (2) secondary, (3) age-associated, or (4) unknown.
  • Treatment with the 10.0 g/day AndroGel® group required approximately 120 days to reach steady state.
  • the mean FSH concentration in patients applying 5.0 g/day of AndroGel® showed an initial decline that was completed by day 30 and another declining phase at day 120 and continuing until the end of treatment.
  • Mean FSH concentrations in the patients receiving the testosterone patch appeared to reach steady state after 30 days but were significantly higher than the normal range.
  • Patients with secondary hypogonadism have a deficient testosterone negative feedback system.
  • the mean FSH concentrations decreased during treatment, although the decrease over time was not statistically significant for the testosterone patch.
  • the patients in the 5.0 g/day AndroGel® group showed a decrease in the mean FSH concentration by about 35% by day 30, with no further decrease evident by day 60. Beyond day 90, the mean FSH concentration in the patients appeared to slowly return toward the pretreatment value. By day 30, all of the 10.0 g/day AndroGel® group had FSH concentrations less than the lower limit.
  • the 5.0 g/day AndroGel® group had a mean pretreatment FSH concentration above the normal range.
  • the mean concentration for this group was within the normal range by day 30 and had decreased more than 50% on days 90 and 180.
  • the decrease in FSH mean concentration in the 10.0 g/day AndroGel® group showed a more rapid response.
  • the concentrations in all six patients decreased to below the lower normal limit by day 30 and remained there for the duration of the study.
  • the six patients who received the testosterone patch exhibited no consistent pattern in the mean FSH level; however, there was an overall trend towards lower FHS levels with continued treatment.
  • the LH concentrations prior to treatment were about 175% of the upper limit of the normal range in primary hypogonadal patients.
  • the mean LH concentrations decreased during treatment in all groups.
  • only the AndroGel® groups decreased the mean LH concentrations enough to fall within the normal range.
  • the primary hypogonadal men receiving AndroGel® showed dose-dependence in both the rate and extent of the LH response.
  • the secondary hypogonadal men were less sensitive to exogenous testosterone.
  • the pretreatment mean concentrations were all within the lower limit normal range.
  • the mean LH concentrations decreased during treatment with all three regimens as shown in FIG. 13 ( b ) and Table 16(b).
  • BMD Bone Mineral Density
  • BMD was assessed by dual energy X-ray absorptiometry (“DEXA”) using Hologic QDR 2000 or 4500 A (Hologic, Waltham, Mass.) on days 0 and 180 in the lumbar spine and left hip regions.
  • BMD of spine was calculated as the average of BMD at L1 to L4.
  • BMD of the left hip which included Ward's triangle, was calculated by the average of BMD from neck, trochanter, and intertrochanter regions.
  • the scans were centrally analyzed and processed at Hologic.
  • BMD assessments were performed at 13 out of the 16 centers (206 out of 227 subjects) because of the lack of the specific DEXA equipment at certain sites.
  • FIGS. 14 ( a )- 14 ( b ) show that before treatment, the BMD of the hip or the spine was not different among the three treatment groups.
  • Significant increases in BMD occurred only in subjects in the AndroGel® 10.0 g/day group and those who switched from AndroGel® 10.0 to 7.5 g/day groups.
  • the increases in BMD were about 1% in the hip and 2% in the spine during the six-month period. Average increases in BMD of 0.6% and 1% in the hip and spine were seen in those receiving 5.0 g/day of AndroGel® but no increase was observed in the testosterone patch group.
  • the baseline hip and spine BMD and the change in BMD on day 180 were not significantly correlated with the average serum testosterone concentration on day 0.
  • the changes in BMD in the hip or spine after testosterone replacement were not significantly different in subjects with hypogonadism due to primary, secondary, aging, or unclassified causes; nor were they different between naive and previously testosterone replaced subjects.
  • the change in BMD in the spine was negatively correlated with baseline BMD values, indicating that the greatest increase in BMD occurred in subjects with the lowest initial BMD.
  • the increase in BMD in the hip (but not in the spine) after testosterone treatment was correlated with the change in serum testosterone levels.
  • the results described above are supported by measurements of a number of serum and urine markers of bone formation. Specifically, the mean concentrations of the serum markers (PTH, SALP, osteocalcin, type I procollagen) generally increase during treatment in all treatment groups. In addition, the ratios of two urine markers of bone formation (N-telopeptide/creatinine ratio and calcium/creatinine ratio) suggests a decrease in bone resorption.
  • PTH Paraathyroid or Calciotropic Hormone
  • Serum intact PTH was measured by two site immunoradiometric assay (“IRMA”) kits from Nichol's Institute (San Juan Capistrano, Calif.).
  • the LLC for the PTH assay was 12.5 ng/L.
  • the intra- and inter-assay coefficients of variation were 6.9 and 9.6%, respectively.
  • the UCLA-Harbor Medical Center has reported previously that the normal male adult range of PTH is 6.8 to 66.4 ng/L.
  • Table 18 provides the PTH concentrations over the 180-day study.
  • FIG. 15 shows that the mean serum PTH levels were within the normal male range in all treatment groups at baseline. Statistically significant increases in serum PTH were observed in all subjects as a group at day 90 without inter-group differences. These increases in serum PTH were maintained at day 180 in all three groups.
  • SALP was quantitated by IRMA using reagents supplied by Hybritech (San Diego, Calif.).
  • the LLQ for the SALP assay was 3.8 ⁇ g/L.; and the intra- and inter-assay precision coefficients were 2.9 and 6.5%, respectively.
  • the UCLA-Harbor Medical Center reported that the adult normal male concentration of SALP ranges from 2.4 to 16.6 ⁇ g/L.
  • Serum osteocalcin was measured by an IRMA from Immutopics (San Clemente, Calif.). The LLQ was 0.45 ⁇ g/L. The intra- and inter-assay coefficients were 5.6 and 4.4%, respectively. The UCLA-Harbor Medical Center reports that the normal male adult range for the osteocalcin assay ranges from 2.9 to 12.7 ⁇ g/L.
  • the baseline mean serum osteocalcin levels were within the normal range in all treatment groups.
  • mean serum osteocalcin increased with testosterone replacement in all subjects as a group without significant differences between the groups.
  • serum osteocalcin either plateaued or showed a decrease by day 180.
  • Serum type I procollagen was measured using a RIA kit from Incstar Corp (Stillwater, Minn.). The LLQ of the procollagen assay was 5 ⁇ g/L, and the intra- and inter-assay precisions were 6.6 and 3.6%, respectively. The UCLA-Harbor Medical Center reports that the normal adult male concentration of type I procollagen ranges from 56 to 310 ⁇ g/L.
  • FIG. 18 and Table 21 show that serum procollagen generally followed the same pattern as serum osteocalcin.
  • the mean levels were similar and within the normal range in all treatment groups.
  • serum procollagen increased significantly in all subjects as a group without treatment group differences.
  • the increase in procollagen was highest on day 30 and then plateaued until day 120.
  • the serum procollagen levels returned to baseline levels.
  • Urine Bone Turnover Markers N-telopeptide/Cr and Ca/Cr Ratios.
  • Urine calcium and creatinine were estimated using standard clinical chemistry procedures by an autoanalyzer operated by the UCLA-Harbor Pathology Laboratory. The procedures were performed using the COBAS MIRA automated chemistry analyzer system manufactured by Roche Diagnostics Systems. The sensitivity of the assay for creatinine was 8.9 mg/dL and the LLQ was 8.9 mg/dL. According to the UCLA-Harbor Medical Center, creatinine levels in normal adult men range from 2.1 mM to 45.1 mM. The sensitivity of the assay for calcium was 0.7 mg/dL and the LLQ was 0.7 mg/dL.
  • the normal range for urine calcium is 0.21 mM to 7.91N-telopeptides were measured by an enzyme-linked immunosorbant assay (“ELISA”) from Ostex (Seattle, Wash.).
  • the LLQ for the N-telopeptide assay was 5 nM bone collagen equivalent (“BCE”).
  • BCE bone collagen equivalent
  • the intra- and inter-assay had a precision of 4.6 and 8.9%, respectively.
  • the normal range for the N-telopeptide assay was 48-2529 nM BCE. Samples containing low or high serum/urine bone marker levels were reassayed after adjusting sample volume or dilution to ensure all samples would be assayed within acceptable precision and accuracy.
  • the normal adult male range for the N-telopeptide/Cr ratio is 13 to 119 nM BCE/nM Cr.
  • urinary N-telopeptide/Cr ratios were similar in all three treatment groups at baseline but decreased significantly in the AndroGel® 10.0 g/day group but not in the AndroGel® 5.0 g/day or testosterone patch group during the first 90 days of treatment. The decrease was maintained such that urinary N-telopeptide/Cr ratio remained lower than baseline in AndroGel® 10.0 g/day and in those subjects adjusted to 7.5 g/day from 10.0 g/day group at day 180. This ratio also decreased in the testosterone patch treatment group by day 180.
  • FIG. 20 shows no significant difference in baseline urinary Ca/Cr ratios in the three groups.
  • urinary Ca/Cr ratios did not show a significant decrease in any treatment group at day 90.
  • urinary Ca/Cr showed marked variation without significant changes in any treatment groups.
  • Serum calcium showed no significant inter-group differences at baseline, nor significant changes after testosterone replacement. Serum calcium levels showed insignificant changes during testosterone replacement.
  • Sexual function and mood were assessed by questionnaires the patients answered daily for seven consecutive days before clinic visits on day 0 and on days 30, 60, 90, 120, 150, and 180 days during gel and patch application.
  • the subjects recorded whether they had sexual day dreams, anticipation of sex, flirting, sexual interaction (e.g., sexual motivation parameters) and orgasm, erection, masturbation, ejaculation, intercourse (e.g., sexual performance parameters) on each of the seven days.
  • the value was recorded as 0 (none) or 1 (any) for analyses and the number of days the subjects noted a parameter was summed for the seven-day period.
  • the average of the four sexual motivation parameters was taken as the sexual motivation score and that of the five sexual motivation parameters as the sexual motivation mean score (0 to 7).
  • the subjects also assessed their level of sexual desire, sexual enjoyment, and satisfaction of erection using a seven-point Likert-type scale (0 to 7) and the percent of full erection from 0 to 100%.
  • the subjects rated their mood using a 0 to 7 score.
  • This questionnaire had been described previously and are fully incorporated by reference. See Wang et al., “Testosterone Replacement Therapy Improves Mood in Hypogonadal Men A Clinical Research Center Study,” 81 J. Clinical Endocrinology & Metabolism 3578-3583 (1996).
  • Libido was assessed from responses on a linear scale of: (1) overall sexual desire, (2) of sexual activity without a partner, and (3) enjoyment of sexual activity with a partner. As shown in FIG. 21 ( b ) and Table 24, as a group, overall sexual desire increased after transdermal testosterone treatment without inter-group difference. Sexual enjoyment with and without a partner ( FIG. 21 ( c ) and Tables 25 and 26) also increased as a group.
  • FIG. 22 ( a ) shows that while all treatment groups had the same baseline sexual performance rating, the rating improved with transdermal testosterone treatment in all groups.
  • the subjects' self-assessment of satisfaction of erection ( FIG. 22 ( b ) and Table 27) and percent full erection ( FIG. 22 ( c ) and Table 28) were also increased with testosterone replacement without significant differences between groups.
  • the improvement in sexual function was not related to the dose or the delivery method of testosterone. Nor was the improvement related to the serum testosterone levels achieved by the various testosterone preparations. The data suggest that once a threshold (serum testosterone level probably at the low normal range) is achieved, normalization of sexual function occurs. Increasing serum testosterone levels higher to the upper normal range does not further improve sexual motivation or performance.
  • the positive and negative mood summary responses to testosterone replacement therapy are shown in FIGS. 23 ( a ) and 23 ( b ). All three treatment groups had similar scores at baseline and all subjects as a group showed improvement in positive mood. Similarly, the negative mood summary scores were similar in the three groups at baseline and as a group the responses to transdermal testosterone applications showed significant decreases without showing between group differences. Specifically, positive mood parameters, such as sense of well being and energy level, improved and negative mood parameters, such as sadness and irritability, decreased. The improvement in mood was observed at day 30 and was maintained with continued treatment. The improvement in mood parameters was not dependent on the magnitude of increase in the serum testosterone levels. Once the serum testosterone increased into the low normal range, maximal improvement in mood parameters occurred. Thus, the responsiveness in sexual function and mood in hypogonadal men in response to testosterone therapy appeared to be dependent on reaching a threshold of serum testosterone at the low normal range.
  • Muscle strength was assessed on days 0, 90, and 180.
  • the one-repetitive maximum (“1-RM”) technique was used to measure muscle mass in bench press and seated leg press exercises. The muscle groups tested included those in the hips, legs, shoulders, arms, and chest.
  • the 1-RM technique assesses the maximal force generating capacity of the muscles used to perform the test. After a 5-10 minute walking and stretching period, the test began with a weight believed likely to represent the patient's maximum strength. The test was repeated using increments of about 2-10 pounds until the patient was unable to lift additional weight with acceptable form. Muscle strength was assessed in 167 out of the 227 patients. Four out of 16 centers did not participate in the muscle strength testing because of lack of the required equipment.
  • FIG. 24 ( a ) and 24 ( b ) and Table 29 The responses of muscle strength testing by the arm/chest and leg press tests are shown in FIG. 24 ( a ) and 24 ( b ) and Table 29. There were no statistical significant differences in arm/chest or leg muscle strength among the three groups at baseline. In general, muscle strength improved in both the arms and legs in all three treatment groups without inter-group differences at both day 90 and 180. The results showed an improvement in muscle strength at 90 and 180 days, more in the legs than the arms, which was not different across treatment groups nor on the different days of assessment. Adjustment of the dose at day 90 did not significantly affect the muscle strength responses to transdermal testosterone preparations.
  • Body composition was measured by DEXA with Hologic 2000 or 4500A series on days 0, 90, and 180. These assessments were done in 168 out of 227 subjects because the Hologic DEXA equipment was not available at 3 out of 16 study centers. All body composition measurements were centrally analyzed and processed by Hologic (Waltham, Mass.).
  • TBM total body mass
  • TBN total body lean mass
  • PFT percent fat
  • TFT total body fat mass
  • FIGS. 25 ( c ) and ( d ) show that the TFT and the PFT decreased in all transdermal AndroGel® treatment groups.
  • TFT was significantly reduced in the 5.0 g/day and 10.0 g/day AndroGel® groups, but was not changed in the testosterone patch group. This decrease was maintained at day 180.
  • the decrease in PFT remained significantly lower in all AndroGel® treated groups but not significantly reduced in the testosterone patch group.
  • the increase in TLN and the decrease in TFT associated with testosterone replacement therapy showed significant correlations with the serum testosterone level attained by the testosterone patch and the different doses of AndroGel®.
  • Testosterone gel administered at 10.0 increased lean mass more than the testosterone patch and the 5.0 g/day AndroGel® groups.
  • the changes were apparent on day 90 after treatment and were maintained or enhanced at day 180.
  • Such changes in body composition was significant even though the subjects were withdrawn from prior testosterone therapy for six weeks.
  • the decrease in TFT and PFT was also related to the serum testosterone achieved and were different across the treatment groups.
  • the testosterone patch group did not show a decrease in PFT or TFT after 180 days of treatment.
  • AndroGel® Treatment with AndroGel® (5.0 to 10.0 g/day) for 90 days reduced PFT and TFT. This decrease was maintained in the 5.0 and 7.5 g/day groups at 180 days but were further lowered with continued treatment with the higher dose of the AndroGel®.
  • the mean compliance was 93.1% and 96.0% for the 5.0 g/day and 10.0 g/day AndroGel® groups during days 1-90, respectively. Compliance remained at over 93% for the three AndroGel® groups from days 91-180.
  • the testosterone patch compliance was 65% during days 1-90 and 74% during days 91-180. The lower compliance in the testosterone patch group was mostly due to skin reactions from the subjects' records.
  • the present invention is also directed to a method for dispensing and packaging the gel.
  • the invention comprises a hand-held pump capable of delivering about 2.5 g of testosterone gel with each actuation.
  • the gel is packaged in foil packets comprising a polyethylene liner. Each packet holds about 2.5 g of testosterone gel. The patient simply tears the packet along a perforated edge to remove the gel.
  • isopropyl myristate binds to the polyethylene liner, additional isopropyl myristate is added to the gel in order to obtain a pharmaceutically effective gel when using this delivery embodiment.
  • about 41% more isopropyl myristate is used in the gel composition (i.e., about 0.705 g instead of about 0.5 g in Table 5), to compensate for this phenomenon.
  • composition can also be dispensed from a rigid multi-dose container (e.g., with a hand pump) having a larger foil packet of the composition inside the container.
  • a rigid multi-dose container e.g., with a hand pump
  • larger packets also comprise a polyethylene liner as above.
  • Both embodiments permit a patient to deliver accurate but incremental amounts of gel (e.g., either 2.5 g, 5.0 g, 7.5 g, etc.) to the body. These delivery mechanisms thus permit the gel to be administered in unit dose form depending on the particular needs and characteristics of the patient.
  • gel e.g., either 2.5 g, 5.0 g, 7.5 g, etc.

Abstract

A pharmaceutical composition useful for treating hypogonadism is disclosed. The composition comprises an androgenic or anabolic steroid, a C1-C4 alcohol, a penetration enhancer such as isopropyl myristate, and water. Also disclosed is a method for treating hypogonadism utilizing the composition.

Description

    RELATED APPLICATIONS
  • This application is a continuation of U.S. patent application Ser. No. 10/248,267 filed on Jan. 3, 2003, which is a continuation of U.S. patent application Ser. No. 09/651,777 filed Aug. 30, 2000, now U.S. Pat. No. 6,503,894, the disclosures of which are incorporated herein by reference in their entirety to the extent permitted by law.
  • FIELD OF THE INVENTION
  • The present invention is directed to a pharmaceutical composition comprising testosterone in a gel formulation, and to methods of using the same.
  • BACKGROUND OF THE INVENTION
  • A. Testosterone Metabolism in Men.
  • Testosterone is the major circulating androgen in men. More than 95% of the 6-7 mg of testosterone produced per day is secreted by the approximately 500 million Leydig cells in the testes. Two hormones produced by the pituitary gland, luteinizing hormone (“LH”) and follicle stimulating hormone (“FSH”), are required for the development and maintenance of testicular function.
  • The most important hormone for the regulation of Leydig cell number and function is LH. In eugonadal men, LH secretion from the pituitary is inhibited through a negative-feedback pathway by increased concentrations of testosterone through the inhibition of the release of gonadotropin-releasing hormone (“GRH”) by the hypothalamus. FSH promotes spermatogenesis and is essential for the normal maturation of sperm. FSH secretion from the pituitary normally is inhibited through a negative-feedback pathway by increased testosterone concentrations.
  • Testosterone is responsible primarily for the development and maintenance of secondary sex characteristics in men. In the body, circulating testosterone is metabolized to various 17-keto steroids through two different pathways. Testosterone can be metabolized to dihydrotestosterone (“DHT”) by the enzyme 5α-reductase. There are two forms of 5α-reductase in the body: one form is found predominately in the liver and non-genital skin while another form is found in the urogenital tract of the male and the genital skin of both sexes. Testosterone can also be metabolized to estradiol (“E2”) by an aromatase enzyme complex found in the liver, fat, and the testes.
  • Testosterone circulates in the blood 98% bound to protein. In men, approximately 40% of the binding is to the high-affinity sex hormone binding globulin (“SHBG”). The remaining 60% is bound weakly to albumin. Thus, a number of measurements for testosterone are available from clinical laboratories. The term “free” testosterone as used herein refers to the fraction of testosterone in the blood that is not bound to protein. The term “total testosterone” or “testosterone” as used herein means the free testosterone plus protein-bound testosterone. The term “bioavailable testosterone” as used herein refers to the non-SHBG bound testosterone and includes testosterone weakly bound to albumin.
  • The conversion of testosterone to DHT is important in many respects. For example, DHT binds with greater affinity to SHBG than does testosterone. In addition, in many tissues, the activity of testosterone depends on the reduction to DHT, which binds to cytosol receptor proteins. The steroid-receptor complex is then transported to the nucleus where it initiates transcription and cellular changes related to androgen action. DHT is also thought to lower prostate volume and inhibit tumor development in the prostate. Thus, given the importance of DHT and testosterone in normal body functioning, researchers frequently assess and report androgen concentrations in patients as total androgen (“DHT+T”) or as a ratio of DHT to testosterone (“DHT/T ratio”).
  • The following table from the UCLA-Harbor Medical Center summarizes the hormone concentrations in normal adult men range:
    TABLE 1
    Hormone Levels in Normal Men
    Hormone Normal Range
    Testosterone 298 to 1043 ng/dL
    Free testosterone 3.5 to 17.9 ng/dL
    DHT 31 to 193 ng/dL
    DHT/T Ratio 0.052 to 0.33
    DHT + T 372 to 1349 ng/dL
    SHBG 10.8 to 46.6 nmol/L
    FSH 1.0 to 6.9 mlU/mL
    LH 1.0 to 8.1 mlU/mL
    E2 17.1 to 46.1 pg/mL
  • There is considerable variation in the half-life of testosterone reported in the literature, ranging from 10 to 100 minutes. Researchers do agree, however, that circulating testosterone has a diurnal variation in normal young men. Maximum levels occur at approximately 6:00 to 8:00 a.m. with levels declining throughout the day. Characteristic profiles have a maximum testosterone level of 720 ng/dL and a minimum level of 430 ng/dL. The physiological significance of this diurnal cycle, if any, however, is not clear.
  • B. Hypogonadal Men and Current Treatments for Hypogonadism.
  • Male hypogonadism results from a variety of patho-physiological conditions in which testosterone concentration is diminished below the normal range. The hypogonadic condition is sometimes linked with a number of physiological changes, such as diminished interest in sex, impotence, reduced lean body mass, decreased bone density, lowered mood, and energy levels.
  • Researchers generally classify hypogonadism into one of three types. Primary hypogonadism includes the testicular failure due to congenital or acquired anorchia, XYY Syndrome, XX males, Noonan's Syndrome, gonadal dysgenesis, Leydig cell tumors, maldescended testes, varicocele, Sertoli-Cell-Only Syndrome, cryptorchidism, bilateral torsion, vanishing testis syndrome, orchiectomy, Klinefelter's Syndrome, chemotherapy, toxic damage from alcohol or heavy metals, and general disease (renal failure, liver cirrhosis, diabetes, myotonia dystrophica). Patients with primary hypogonadism show an intact feedback mechanism in that the low serum testosterone concentrations are associated with high FSH and LH concentrations. However, because of testicular or other failures, the high LH concentrations are not effective at stimulating testosterone production.
  • Secondary hypogonadism involves an idiopathic gonadotropin or LH-releasing hormone deficiency. This type of hypogonadism includes Kallman's Syndrome, Prader-Labhart-Willi's Syndrome, Laurence-Moon-Biedl's Syndrome, pituitary insufficiency/adenomas, Pasqualini's Syndrome, hemochromatosis, hyperprolactinemia, or pituitary-hypothalamic injury from tumors, trauma, radiation, or obesity. Because patients with secondary hypogonadism do not demonstrate an intact feedback pathway, the lower testosterone concentrations are not associated with increased LH or FSH levels. Thus, these men have low testosterone serum levels but have gonadotropins in the normal to low range.
  • Third, hypogonadism may be age-related. Men experience a slow but continuous decline in average serum testosterone after approximately age 20 to 30 years. Researchers estimate that the decline is about 1-2% per year. Cross-sectional studies in men have found that the mean testosterone value at age 80 years is approximately 75% of that at age 30 years. Because the serum concentration of SHBG increases as men age, the fall in bioavailable and free testosterone is even greater than the fall in total testosterone. Researchers have estimated that approximately 50% of healthy men between the ages of 50 and 70 have levels of bioavailable testosterone that are below the lower normal limit. Moreover, as men age, the circadian rhythm of testosterone concentration is often muted, dampened, or completely lost. The major problem with aging appears to be within the hypothalamic-pituitary unit. For example, researchers have found that with aging, LH levels do not increase despite the low testosterone levels. Regardless of the cause, these untreated testosterone deficiencies in older men may lead to a variety of physiological changes, including sexual dysfunction, decreased libido, loss of muscle mass, decreased bone density, depressed mood, and decreased cognitive function. The net result is geriatric hypogonadism, or what is commonly referred to as “male menopause.” Today, hypogonadism is the most common hormone deficiency in men, affecting 5 in every 1,000 men. At present, it is estimated that only five percent of the estimated four to five million American men of all ages with hypogonadism currently receive testosterone replacement therapy. Thus, for years, researchers have investigated methods of delivering testosterone to men. These methods include intramuscular injections (43%), oral replacement (24%), pellet implants (23%), and transdermal patches (10%). A summary of these methods is shown in Table 2.
    TABLE 2
    Mode of Application and Dosage of
    Various Testosterone Preparations
    Preparation Route Of Application Full Substitution Dose
    In Clinical Use
    Testosterone Intramuscular injection 200-25.0 g every
    enanthate 2-3 weeks
    Testosterone Intramuscular injection 200 mg every 2 weeks
    cypionate
    Testosterone Oral 2-4 capsules at
    undecanoate 40 mg per day
    Transdermal Scrotal skin 1 membrane per day
    testosterone
    patch
    Transdermal Non-scrotal skin 1 or 2 systems per day
    testosterone
    patch
    Testosterone Implantation under the 3-6 implants of
    implants abdominal skin 200 mg every 6 months
    Under Development
    Testosterone Sublingual 2.5-5.0 mg
    cyclodextrin twice daily
    Testosterone Intramuscular injection 1000 mg every
    undecanoate 8-10 weeks
    Testosterone Intramuscular injection 1000 mg every
    buciclate 12-16 weeks
    Testosterone Intramuscular injection 315 mg for 11 weeks
    microspheres
    Obsolete
    17α-Methyl- Oral 25-5.0 g per day
    testosterone
    Fluoxymesterone Sublingual 10-25 mg per day
    Oral 10-20 mg per day
  • As discussed below, all of the testosterone replacement methods currently employed suffer from one or more drawbacks, such as undesirable pharmacokinetic profiles or skin irritation. Thus, although the need for an effective testosterone replacement methodology has existed for decades, an alternative replacement therapy that overcomes these problems has never been developed. The present invention is directed to a 1% testosterone hydroalcoholic gel that overcomes the problems associated with current testosterone replacement methods.
  • 1. Subdermal Pellet Implants.
  • Subdermal implants have been used as a method of testosterone replacement since the 1940s. The implant is produced by melting crystalline testosterone into a cylindrical form. Today, pellet implants are manufactured to contain either 100 mg (length 6 mm, surface area 1172) or 200 mg of testosterone (length 12 mm, surface area 202 mm2). Patients receive dosages ranging from 100 to 1,200 mg, depending on the individual's requirements. The implants are inserted subcutaneously either by using a trocar and cannula or by open surgery into an area where there is relatively little movement. Frequently, the implant is placed in the lower abdominal wall or the buttock. Insertion is made under local anesthesia, and the wound is closed with an adhesive dressing or a fine suture.
  • Implants have several major drawbacks. First, implants require a surgical procedure which many hypogonadal men simply do not wish to endure. Second, implant therapy includes a risk of extrusion (8.5%), bleeding (2.3%), or infection (0.6%). Scarring is also a risk. Perhaps most important, the pharmacokinetic profile of testosterone pellet implant therapy fails to provide men with a suitable consistent testosterone level. In general, subdermal testosterone implants produce supra-physiologically high serum testosterone levels which slowly decline so that before the next injection subnormally low levels of testosterone are reached. For example, in one recent pharmacokinetic study, hypogonadal patients who received six implants (1,200 mg testosterone) showed an initial short-lived burst release of testosterone within the first two days after application. A stable plateau was then maintained over then next two months (day 2: 1,015 ng/dL; day 63: 990 ng/dL). Thereafter, the testosterone levels declined to baseline by day 300. DHT serum concentrations also rose significantly above the baseline, peaking at about 63 days after implementation and greatly exceeding the upper limit of the normal range. From day 21 to day 189, the DHT/T ratio was significantly increased. The pharmacokinetic profiles for testosterone, DHT, and DHT/T in this study are shown in FIG. 1. See “Jockenhovel et al., Pharmacokinetics and Pharmacodynamics of Subcutaneous Testosterone Implants in Hypogonadal Men,” 45 Clinical Endocrinology 61-71 (1996). Other studies involving implants have reported similar undesirable pharmacokinetic profiles.
  • 2. Injection of Testosterone Esters.
  • Since the 1950s, researchers have experimented with the intermuscular depot injection of testosterone esters (such as enanthate, cypionate) to increase testosterone serum levels in hypogonadal men. More recent studies have involved injection of testosterone buciclate or testosterone undecanoate in an oil-based vehicle. Other researchers have injected testosterone microcapsule formulations.
  • Testosterone ester injection treatments suffer from many problems. Patients receiving injection therapy often complain that the delivery mechanism is painful and causes local skin reactions. In addition, testosterone microcapsule treatment requires two simultaneous intramuscular injections of a relatively large volume, which may be difficult to administer due to the high viscosity of the solution and the tendency to block the needle. Other men generally find testosterone injection therapy inconvenient because injection usually requires the patient to visit his physician every two to three weeks.
  • Equally important, injection-based testosterone replacement treatments still create an undesirable pharmacokinetic profile. The profile generally shows a supra-physiologic testosterone concentration during the first 24 to 48 hours followed by a gradual fall often to sub-physiologic levels over the next few weeks. These high serum testosterone levels, paralleled by increases in E2, are also considered the reason for acne and gynecomastia occurring in some patients, and for polycythaemia, occasionally encountered especially in older patients using injectable testosterone esters. In the case of testosterone buciclate injections, the treatment barely provides normal androgen serum levels and the maximal increase of serum testosterone over baseline does not exceed 172 ng/dL (6 nmol/dL) on average. Because libido, potency, mood, and energy are thought to fluctuate with the serum testosterone level, testosterone injections have largely been unsuccessful in influencing these variables. Thus, testosterone injection remains an undesirable testosterone replacement treatment method.
  • 3. Oral/Sublingual/Buccal Preparations of Androgens.
  • In the 1 970s, researchers began using oral, sublingual, or buccal preparations of androgens (such as fluoxymesterone, 17α-methyl-testosterone or testosterone undecanoate) as a means for testosterone replacement. More recently, researchers have experimented with the sublingual administration of testosterone-hydroxypropyl-beta-cyclodextrin inclusion complexes. Predictably, both fluoxymesterone and methyl testosterone are 17-alkylated and thus associated with liver toxicity. Because these substances must first pass through the liver, they also produce an unfavorable effect on serum lipid profile, increasing LDL and decreasing HDL, and carbohydrate metabolism. While testosterone undecanoate has preferential absorption through the intestinal lymphatics, it has not been approved in the United States.
  • The pharmacokinetic profiles for oral, sublingual, and buccal delivery mechanisms are also undesirable because patients are subjected to super-physiologic testosterone levels followed by a quick return to the baseline. For example, one recent testing of a buccal preparation showed that patients obtained a peak serum hormone levels within 30 minutes after administration, with a mean serum testosterone concentration of 2,688±147 ng/dL and a return to baseline in 4 to 6 hours. See Dobs et al., Pharmacokinetic Characteristics, Efficacy and Safety of Buccal Testosterone in Hypogonadal Males: A Pilot Study, 83 J. Clinical Endocrinology & Metabolism 33-39 (1998). To date, the ability of these testosterone delivery mechanisms to alter physiological parameters (such as muscle mass, muscle strength, bone resorption, urinary calcium excretion, or bone formation) is inconclusive. Likewise, researchers have postulated that super-physiologic testosterone levels may not have any extra beneficial impact on mood parameters such as anger, nervousness, and irritability.
  • 4. Testosterone Transdermal Patches.
  • The most recent testosterone delivery systems have involved transdermal patches. Currently, there are three patches used in the market: TESTODERM®, TESTODERM® TTS, and ANDRODERM®.
  • a. TESTODERM®.
  • TESTODERM® (Alza Pharmaceuticals, Mountain View, Calif.) was the first testosterone-containing patch developed. The TESTODERM® patch is currently available in two sizes (40 or 60 cm2). The patch contains 10 or 15 mg of testosterone and delivers 4.0 mg or 6.0 mg of testosterone per day. TESTODERM® is placed on shaved scrotal skin, aided by application of heat for a few seconds from a hair dryer.
  • FIG. 2 shows a typical pharmacokinetic testosterone profile for both the 40 cm2 and 60 cm2 patch. Studies have also shown that after two to four weeks of continuous daily use, the average plasma concentration of DHT and DHT/T increased four to five times above normal. The high serum DHT levels are presumably caused by the increased metabolism of 5α-reductase in the scrotal skin.
  • Several problems are associated with the TESTODERM® patch. Not surprisingly, many men simply do not like the unpleasant experience of dry-shaving the scrotal hair for optimal contact. In addition, patients may not be able to wear close-fitting underwear when undergoing treatment. Men frequently experience dislodgment of the patch, usually with exercise or hot weather. In many instances, men experience itching and/or swelling in the scrotal area. Finally, in a number of patients, there is an inability to achieve adequate serum hormone levels.
  • b. TESTODERM® TTS.
  • The most recently developed non-scrotal patch is TESTODERM® TTS (Alza Pharmaceuticals, Mountain View, Calif.). It is an occlusive patch applied once daily to the arm, back, or upper buttocks. The system is comprised of a flexible backing of transparent polyester/ethylene-vinyl acetate copolymer film, a drug reservoir of testosterone, and an ethylene-vinyl acetate copolymer membrane coated with a layer of polyisobutylene adhesive formulation. A protective liner of silicone-coated polyester covers the adhesive surface.
  • Upon application, serum testosterone concentrations rise to a maximum at two to four hours and return toward baseline within two hours after system removal. Many men, however, are unable to obtain and/or sustain testosterone levels within the normal range. The pharmacokinetic parameters for testosterone concentrations are shown as follows:
    TABLE 3
    TESTODERM ® TTS Testosterone Parameters
    Parameters Day
    1 Day 5
    Cmax (ng/dL) 482 ± 149 473 ± 148
    Tmax (h) 3.9 3.0
    Cmin (ng/dL) 164 ± 104 189 ± 86 
    Tmin (h) 0 0
  • The typical 24-hour steady state testosterone concentration achieved with TESTODERM® TTS patch is shown in FIG. 3.
  • Because of TESTODERM® patch is applied to the scrotal skin while the TESTODERM TTS® patch is applied to non-scrotal skin, the two patches provide different steady-state concentrations of the two major testosterone metabolites, DTH and E2:
    TABLE 4
    Hormone Levels Using TESTODERM ® and
    TESTODERM ® TTS
    Hormone Placebo TESTODERM ® TESTODERM ® TTS
    DHT (ng/dL) 11 134 38
    E2 (pg/ml) 3.8 10 21.4
  • Likewise, in contrast to the scrotal patch, TESTODERM TTS® treatment creates a DHT/T ratio that is not different from that of a placebo treatment. Both systems, however, suffer from similar problems. In clinical studies, TESTODERM® TTS is associated with transient itching in 12% of patients, erythema in 3% of patients, and puritus in 2% of patients. Moreover, in one 14-day study, 42% of patients reported three or more detachments, 33% of which occurred during exercise.
  • C. ANDRODERM®.
  • ANDRODERM® (Watson Laboratories, Inc., Corona, Calif.) is a testosterone-containing patch applied to non-scrotal skin. The circular patch has a total surface area of 37 cm.2 The patch consists of a liquid reservoir containing 12.2 mg of testosterone and a permeation-enhanced vehicle containing ethanol, water, monoglycerides, fatty acid esters, and gelling agents. The suggested dose of two patches, applied each night in a rotating manner on the back, abdomen, upper arm, or thigh, delivers 4.1 to 6.8 mg of testosterone.
  • The steady state pharmacokinetic profile of a clinical study involving ANDRODERM® is shown in FIG. 4. In general, upon repeated application of the ANDRODERM® patch, serum testosterone levels increase gradually for eight hours after each application and then remain at this plateau level for about another eight hours before declining.
  • In clinical trials, ANDRODERM® is associated with skin irritation in about a third of the patients, and 10% to 15% of subjects have been reported to discontinue the treatment because of chronic skin irritation. Preapplication of corticosteroid cream at the site of application of ANDRODERM® has been reported to decrease the incidence and severity of the skin irritation. A recent study, however, found that the incidence of skin reactions sufficiently noxious enough to interrupt therapy was as high as 52%. See Parker et al., Experience with Transdermal Testosterone Replacement in Hypogonadal Men, 50 Clinical Endocrinology (Oxf) 57-62 (1999). The study reported: Two-thirds of respondents found the Andropatch unsatisfactory. Patches were variously described as noisy, visually indiscrete, embarrassing, unpleasant to apply and remove, and generally to be socially unacceptable. They fell off in swimming pools and showers, attracted ribald comments from sporting partners, and left bald red marks over trunk and limbs. Dogs, wives, and children were distracted by noise of the patches with body movements. Those with poor mobility or manual dexterity (and several were over 70 years of age) found it difficult to remove packaging an apply patches dorsally.
  • d. Transdermal Patch Summary.
  • In sum, the transdermal patch generally offers an improved pharmacokinetic profile compared to other currently used testosterone delivery mechanisms. However, as discussed above, the clinical and survey data shows that all of these patches suffer from significant drawbacks, such as buritus, burn-like blisters, and erythema. Moreover, one recent study has concluded that the adverse effects associated with transdermal patch systems are “substantially higher” than reported in clinical trials. See Parker, supra. Thus, the transdermal patch still remains an inadequate testosterone replacement therapy alternative for most men.
  • 5. DHT Gels.
  • Researchers have recently begun investigating the application of DHT to the skin in a transdermal gel. However, the pharmacokinetics of a DHT-gel is markedly different from that of a testosterone gel. Application of DHT-gel results in decreased serum testosterone, E2, LH, and FSH levels. Thus, DHT gels are not effective at increasing testosterone levels in hypogonadal men. Accordingly, there is a definite need for a testosterone formulation that safely and effectively provides an optimal and predictable pharmacokinetic profile.
  • SUMMARY OF INVENTION
  • The foregoing problems are solved and a technical advance is achieved with the present invention. The present invention generally comprises a testosterone gel. Daily transdermal application of the gel in hypogonadal men results in a unique pharmacokinetic steady-state profile for testosterone. Long-term treatment further results in, for example, increased bone mineral density, enhanced libido, enhanced erectile frequency and satisfaction, increased positive mood, increased muscle strength, and improved body composition without significant skin irritation. The present invention is also directed to a unique method of administering the testosterone gel employing a packet having a polyethylene liner compatible with the components of the gel.
  • BRIEF DESCRIPTION OF DRAWINGS
  • FIG. 1 is a graph of testosterone concentrations, DHT concentrations, and the DHT/T ratio for patients receiving a subdermal testosterone pellet implant over a period of 300 days after implantation.
  • FIG. 2 shows a typical pharmacokinetic testosterone profile for both the 40 cm2 and 60 cm2 patch. Studies have also shown that after two to four weeks of continuous daily use, the average plasma concentration of DHT and DHT/T increased four to five times above normal. The high serum DHT levels are presumably caused by the increased metabolism of 5α-reductase in the scrotal skin.
  • FIG. 3 is a 24-hour testosterone pharmacokinetic profile for patients receiving the TESTODERM® TTS patch.
  • FIG. 4 is a 24-hour testosterone pharmacokinetic profile for patients receiving the ANDRODERM® patch.
  • FIG. 5(a) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men prior to receiving 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 5(b) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men on the first day of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 5(c) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men on day 30 of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel, or the testosterone patch (by initial treatment group).
  • FIG. 5(d) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men on day 90 of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 5(e) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men on day 180 of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by final treatment group).
  • FIG. 5(f) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men on day 0, 1, 30, 90, and 180 of treatment with 5.0 g/day of AndroGel®.
  • FIG. 5(g) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men on day 0, 1, 30, 90, and 180 of treatment with 10.0 g/day of AndroGel®.
  • FIG. 5(h) is a graph showing the 24-hour testosterone pharmacokinetic profile for hypogonadal men on day 0, 1, 30, 90, and 180 of treatment with the testosterone patch.
  • FIG. 6(a) is a graph showing the 24-hour free testosterone pharmacokinetic profile for hypogonadal men on day 1 of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 6(b) is a graph showing the 24-hour free testosterone pharmacokinetic profile for hypogonadal men on day 30 of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 6(c) is a graph showing the 24-hour free testosterone pharmacokinetic profile for hypogonadal men on day 90 of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 6(d) is a graph showing the 24-hour free testosterone pharmacokinetic profile for hypogonadal men on day 180 of treatment with either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by final treatment group).
  • FIG. 6(e) is a graph showing the 24-hour free testosterone pharmacokinetic profile for hypogonadal men on day 0, 1, 30, 90, and 180 of treatment with 5.0 g/day of AndroGel®.
  • FIG. 6(f) is a graph showing the 24-hour free testosterone pharmacokinetic profile for hypogonadal men on day 0, 1, 30, 90, and 180 of treatment with 10.0 g/day of AndroGel®.
  • FIG. 6(g) is a graph showing the 24-hour free testosterone pharmacokinetic profile for hypogonadal men on day 0, 1, 30, 90, and 180 of treatment with the testosterone patch.
  • FIG. 7 is a graph showing the DHT concentrations on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 8 is a graph showing the DHT/T ratio on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 9 is a graph showing the total androgen concentrations (DHT+T) on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 10 is a graph showing the E2 concentrations on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 11 is a graph showing the SHBG concentrations on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 12(a) is a graph showing the FSH concentrations on days 0 through 180 for men having primary hypogonadism and receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 12(b) is a graph showing the FSH concentrations on days 0 through 180 for men having secondary hypogonadism and receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 12(c) is a graph showing the FSH concentrations on days 0 through 180 for men having age-associated hypogonadism and receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 12(d) is a graph showing the FSH concentrations on days 0 through 180 for men having hypogonadism of an unknown origin and receiving either 5.0 g/day of AndroGel®, 10.0of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 13(a) is a graph showing the LH concentrations on days 0 through 180 for men having primary hypogonadism and receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 13(b) is a graph showing the LH concentrations on days 0 through 180 for men having secondary hypogonadism and receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 13(c) is a graph showing the LH concentrations on days 0 through 180 for men having age-associated hypogonadism and receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 13(d) is a graph showing the LH concentrations on days 0 through 180 for men having hypogonadism of an unknown origin and receiving either 5.0 g/day of AndroGel®, 10.0 of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 14(a) is a bar graph showing the change in hip BMD for hypogonadal men after 180 days of treatment with 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 14(b) is a bar graph showing the change in spine BMD for hypogonadal men after 180 days of treatment with 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 15 is a graph showing PTH concentrations on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 16 is a graph showing SALP concentrations on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 17 is a graph showing the osteocalcin concentrations on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 18 is a graph showing the type I procollagen concentrations on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 19 is a graph showing the N-telopeptide/Cr ratio on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 20 is a graph showing the Ca/Cr ratio on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch (by initial treatment group).
  • FIG. 21(a) is a graph showing sexual motivation scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 21(b) is a graph showing overall sexual desire scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 21(c) is a graph showing sexual enjoyment (with a partner) scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 22(a) is a graph showing sexual performance scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 22(b) is a graph showing erection satisfaction performance scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 22(c) is a graph showing percent erection scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 23(a) is a graph showing positive mood scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 23(b) is a graph showing negative mood scores on days 0 through 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 24(a) is a bar graph showing the change in leg strength on days 90 and 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 24(b) is a bar graph showing the change in arm strength on days 90 and 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 25(a) is a bar graph showing the change in total body mass on days 90 and 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 25(b) is a bar graph showing the change in lean body mass on days 90 and 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 25(c) is a bar graph showing the change in fat mass on days 90 and 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • FIG. 25(d) is a bar graph showing the change in percent body fat on days 90 and 180 for hypogonadal men receiving either 5.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, 10.0 g/day of AndroGel®, or the testosterone patch.
  • DETAILED DESCRIPTION
  • While the present invention may be embodied in many different forms, several specific embodiments are discussed herein with the understanding that the present disclosure is to be considered only as an exemplification of the principles of the invention, and it is not intended to limit the invention to the embodiments illustrated.
  • The present invention is directed to a pharmaceutical composition for percutaneous administration comprising at least one active pharmaceutical ingredient (e.g., testosterone) in a hydroalcoholic gel. In a broad aspect of the invention, the active ingredients employed in the composition may include anabolic steroids such as androisoxazole, bolasterone, clostebol, ethylestrenol, formyldienolone, 4-hydroxy-19-nortestosterone, methenolone, methyltrienolone, nandrolone, oxymesterone, quinbolone, stenbolone, trenbolone; androgenic steroids such as boldenone, fluoxymesterone, mestanolone, mesterolone, methandrostenolone, 17-methyltestosterone, 17 Alpha-methyl-testosterone 3-cyclopentyl enol ether, norethandrolone, normethandrone, oxandrolone, oxymetholone, prasterone, stanlolone, stanozolol, dihydrotestosterone, testosterone; and progestogens such as anagestone, chlormadinone acetate, delmadinone acetate, demegestone, dimethisterone, dihydrogesterone, ethinylestrenol, ethisterone, ethynodiol, ethynodiol diacetate, flurogestone acetate, gestodene, gestonorone caproate, haloprogesterone, 17-hydroxy-16-methylene-progesterone, 17 Beta-hydroxyprogesterone, 17 Alpha-hydroxyprogesterone caproate, medrogestone, medroxyprogesterone, megestrol acetate, melengestrol, norethindrone, norethindrone acetate, norethynodrel, norgesterone, norgestimate, norgestrel, norgestrienone, 19-norprogesterone, norvinisterone, pentagestrone, progesterone, promegestone, quingestrone, and trengestone; and all enantiomers, isomers and derivatives of these compounds. (Based upon the list provided in The Merck Index, Merck & Co. Rahway, N.J. (1998)).
  • In addition to the active ingredient, the gel comprises one or more lower alcohols, such as ethanol or isopropanol; a penetration enhancing agent; a thickener; and water. Additionally, the present invention may optionally include salts, emollients, stabilizers, antimicrobials, fragrances, and propellants.
  • A “penetration enhancer” is an agent known to accelerate the delivery of the drug through the skin. These agents also have been referred to as accelerants, adjuvants, and absorption promoters, and are collectively referred to herein as “enhancers.” This class of agents includes those with diverse mechanisms of action including those which have the function of improving the solubility and diffusibility of the drug, and those which improve percutaneous absorption by changing the ability of the stratum corneum to retain moisture, softening the skin, improving the skin's permeability, acting as penetration assistants or hair-follicle openers or changing the state of the skin such as the boundary layer.
  • The penetration enhancer of the present invention is a functional derivative of a fatty acid, which includes isosteric modifications of fatty acids or non-acidic derivatives of the carboxylic functional group of a fatty acid or isosteric modifications thereof. In one embodiment, the functional derivative of a fatty acid is an unsaturated alkanoic acid in which the COOH group is substituted with a functional derivative thereof, such as alcohols, polyols, amides and substituted derivatives thereof. The term “fatty acid” means a fatty acid that has four (4) to twenty-four (24) carbon atoms. Non-limiting examples of penetration enhancers include C8-C22 fatty acids such as isostearic acid, octanoic acid, and oleic acid; C8-C22 fatty alcohols such as oleyl alcohol and lauryl alcohol; lower alkyl esters of C8-C22 fatty acids such as ethyl oleate, isopropyl myristate, butyl stearate, and methyl laurate; di(lower)alkyl esters of C6-C8 diacids such as diisopropyl adipate; monoglycerides of C8-C22 fatty acids such as glyceryl monolaurate; tetrahydrofurfiuryl alcohol polyethylene glycol ether; polyethylene glycol, propylene glycol; 2-(2-ethoxyethoxy) ethanol; diethylene glycol monomethyl ether; alkylaryl ethers of polyethylene oxide; polyethylene oxide monomethyl ethers; polyethylene oxide dimethyl ethers; dimethyl sulfoxide; glycerol; ethyl acetate; acetoacetic ester; N-alkylpyrrolidone; and terpenes.
  • The thickeners used herein may include anionic polymers such as polyacrylic acid (CARBOPOL® by B.F. Goodrich Specialty Polymers and Chemicals Division of Cleveland, Ohio), carboxymethylcellulose and the like. Additional thickeners, enhancers and adjuvants may generally be found in United States Pharmacopeia/National Formulary (2000); Remington's The Science and Practice of Pharmacy, Meade Publishing Co.
  • The amount of drug to be incorporated in the composition varies depending on the particular drug, the desired therapeutic effect, and the time span for which the gel is to provide a therapeutic effect. The composition is used in a “pharmacologically effective amount.” This means that the concentration of the drug is such that in the composition it results in a therapeutic level of drug delivered over the term that the gel is to be used. Such delivery is dependent on a number of variables including the drug, the form of drug, the time period for which the individual dosage unit is to be used, the flux rate of the drug from the gel, surface area of application site, etc. The amount of drug necessary can be experimentally determined based on the flux rate of the drug through the gel, and through the skin when used with and without enhancers.
  • One such testosterone gel has only recently been made available in the United States under the trademark AndroGel® by Unimed Pharmaceuticals, Inc., Deerfield, Ill., one of the assignees of this application. In one embodiment, the gel is comprised of the following substances in approximate amounts:
    TABLE 5
    Composition of AndroGel ®
    AMOUNT (w/w)
    PER 100 g OF
    SUBSTANCE GEL
    Testosterone 1.0 g
    Carbopol 980 0.90 g
    Isopropyl myristate 0.50 g
    0.1 N NaOH 4.72 g
    Ethanol (95% w/w) 72.5 g*
    Purified water (qsf) 100 g

    *Corresponding to 67 g of ethanol.
  • One skilled in the art will appreciate that the constituents of this formulation may be varied in amounts yet continue to be within the spirit and scope of the present invention. For example, the composition may contain about 0.1 to about 10.0 g of testosterone, about 0.1 to about 5.0 g Carbopol, about 0.1 to about 5.0 g isopropyl myristate, and about 30.0 to about 98.0 g ethanol; or about 0.1% to about 10.0% of testosterone, about 0.1% to about 5.0% Carbopol, about 0.1% to about 5.0% isopropyl myristate, and about 30.0% to about 98.0% ethanol, on a weight to weight basis of the composition.
  • A therapeutically effective amount of the gel is rubbed onto a given area of skin by the user. The combination of the lipophilic testosterone with the hydroalcoholic gel helps drive the testosterone in to the outer layers of the skin where it is absorbed and then slowly released into the blood stream. As demonstrated by the data presented herein, the administration of the gel of the present invention has a sustained effect.
  • Toxicity and therapeutic efficacy of the active ingredients can be determined by standard pharmaceutical procedures, e.g., for determining LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds which exhibit large therapeutic indices are preferred. While compounds that exhibit toxic side effects may be used, care should be taken to design a delivery system that targets such compounds to the site of affected tissue in order to minimize potential damage to uninfected cells and, thereby, reduce side effects.
  • The term “treatment” as used herein refers to any treatment of a human condition or disease and includes: (1) preventing the disease or condition from occurring in a subject which may be predisposed to the disease but has not yet been diagnosed as having it, (2) inhibiting the disease or condition, i.e., arresting its development, (3) relieving the disease or condition, i.e., causing regression of the condition, or (4) relieving the conditions caused by the disease, i.e., stopping the symptoms of the disease.
  • Although the examples of the present invention involve the treatment of disorders associated with hypogonadal men, the composition and method of the present invention may be used to treat these disorders in humans and animals of any kind, such as dogs, pigs, sheep, horses, cows, cats, zoo animals, and other commercially bred farm animals.
  • The present invention is further illustrated by the following examples, which should not be construed as limiting in any way. The contents of all cited references throughout this application are hereby expressly incorporated by reference. The practice of the present invention will employ, unless otherwise indicated, conventional techniques of pharmacology and pharmaceutics, which are within the skill of the art.
  • EXAMPLE 1 Treatment of Hypogonadism in Male Subjects
  • One embodiment of the present invention involves the transdermal application of AndroGel® as a method of treating male hypogonadism. As demonstrated below, application of the gel results in a unique pharmacokinetic profile for testosterone, as well as concomitant modulation of several other sex hormones. Application of the testosterone gel to hypogonadal male subjects also results in (1) increased bone mineral density, (2) enhanced libido, (3) enhanced erectile capability and satisfaction, (4) increased positive mood, (5) increased muscle strength, and (6) better body composition, such increased total body lean mass and decreased total body fat mass. Moreover, the gel is not generally associated with significant skin irritation.
  • Methods. In this example, hypogonadal men were recruited and studied in 16 centers in the United States. The patients were between 19 and 68 years and had single morning serum testosterone levels at screening of less than or equal to 300 ng/dL (10.4 nmol/L). A total of 227 patients were enrolled: 73, 78, and 76 were randomized to receive 5.0 g/day of AndroGel® (delivering 50 mg/day of testosterone to the skin of which about 10% or 5 mg is absorbed), 10.0 g/day of AndroGel® (delivering 100 mg/day of testosterone to the skin of which about 10% or 10 mg is absorbed), or the ANDRODERM® testosterone patch (“T patch”) (delivering 50 mg/day of testosterone), respectively.
  • As shown in the following table, there were no significant group-associated differences of the patients' characteristics at baseline.
    TABLE 6
    Baseline Characteristics of the Hypogonadal Men
    AndroGel ® AndroGel ®
    Treatment Group T patch (5.0 g/day) (10.0 g/day)
    No of subjects 76 73 78
    enrolled
    Age (years) 51.1 51.3 51.0
    Range (years) 28-67 23-67 19-68
    Height (cm) 179.3 ± 0.9  175.8 ± 0.8  178.6 ± 0.8 
    Weight (kg) 92.7 ± 1.6  90.5 ± 1.8  91.6 ± 1.5 
    Serum testosterone 6.40 ± 0.41 6.44 ± 0.39 6.49 ± 0.37
    (nmol/L)
    Causes of
    hypogonadism
    Primary 34 26 34
    hypogonadism
    Klinefelter's 9 5 8
    Syndrome
    Post Orchidectomy/ 2 1 3
    Anorchia
    Primary Testicular 23 20 23
    Failure
    Secondary 15 17 12
    hypogonadism
    Kallman's Syndrome 2 2 0
    Hypothalimic 6 6 3
    Pituitary Disorder
    Pituitary Tumor 7 9 9
    Aging 6 13 6
    Not classified 21 17 26
    Years diagnosed 5.8 ± 1.1 4.4 ± 0.9  5.7 ± 1.24
    Number previously 50 (65.8%) 38 (52.1%) 46 (59.0%)
    treated with
    testosterone
    Type of Previous
    Hormonal Treatment
    Intramuscular
    26 20 28
    injections
    Transdermal patch 12 7 8
    All others 12 11 10
    Duration of treat- 5.8 ± 1.0 5.4 ± 0.8  4.6 ± 80.7
    ment (years)
  • Forty-one percent (93/227) of the subjects had not received prior testosterone replacement therapy. Previously treated hypogonadal men were withdrawn from testosterone ester injection for at least six weeks and oral or transdermal androgens for four weeks before the screening visit. Aside from the hypogonadism, the subjects were in good health as evidenced by medical history, physical examination, complete blood count, urinalysis, and serum biochemistry. If the subjects were on lipid-lowering agents or tranquilizers, the doses were stabilized for at least three months prior to enrollment. Less than 5% of the subjects were taking supplemental calcium or vitamin D during the study. The subjects had no history of chronic medical illness, alcohol or drug abuse. They had a normal rectal examination, a PSA level of less than 4 ng/mL, and a urine flow rate of 12 mL/s or greater. Patients were excluded if they had a generalized skin disease that might affect the testosterone absorption or prior history of skin irritability with ANDRODERM® patch. Subjects weighing less than 80% or over 140% of their ideal body weight were also excluded.
  • The randomized, multi-center, parallel study compared two doses of AndroGel® with the ANDRODERM® testosterone patch. The study was double-blind with respect to the AndroGel® dose and open-labeled for the testosterone patch group. For the first three months of the study (days 1 to 90), the subjects were randomized to receive 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, or two non-scrotal patches. In the following three months (days 91 to 180), the subjects were administered one of the following treatments: 5.0 g/day of AndroGel®, 10.0 g/day of AndroGel®, 7.5 g/day of AndroGel®, or two non-scrotal patches. Patients who were applying AndroGel® had a single, pre-application serum testosterone measured on day 60 and, if the levels were within the normal range of 300 to 1,000 ng/dL (10.4 to 34.7 nmol/L), then they remained on their original dose. Patients with testosterone levels less than 300 ng/dL and who were originally assigned to apply 5.0 g/day of AndroGel® and those with testosterone levels more than 1,000 ng/dL who had received 10.0 g/day of AndroGel® were then reassigned to administer 7.5 of AndroGel® for days 91 to 180.
  • Accordingly, at 90 days, dose adjustments were made in the AndroGel® groups based on the pre-application serum testosterone levels on day 60. Twenty subjects in the 5.0 g/day AndroGel® group had the dose increased to 7.5 g/day. Twenty patients in the 10.0 g/day AndroGel® group had the AndroGel® dose reduced to 7.5 g/day. There were three patients in the testosterone patch group who were switched to 5.0 g/day AndroGel® because of patch intolerance. One 10.0 g/day AndroGel® subject was adjusted to receive 5.0 g/day and one 5.0 AndroGel® subject had the dose adjusted to 2.5 g/day. The number of subjects enrolled into day 91 to 180 of the study thus consisted of 51 receiving 5.0 g/day of AndroGel®, 40 receiving 7.5 g/day of AndroGel®, 52 receiving 10.0 g/day of AndroGel®, and 52 continuing on the ANDRODERM® patch. The treatment groups in this example may thus be characterized in two ways, either by “initial” or by the “final” treatment group.
  • Subjects returned to the study center on days 0, 30, 60, 90, 120, 150, and 180 for a clinical examination, skin irritation and adverse event assessments. Fasting blood samples for calcium, inorganic phosphorus, parathyroid hormone (“PTH”), osteocalcin, type I procollagen, and skeletal specific alkaline phosphatase (“SALP”) were collected on days 0, 30, 90, 120, and 180. In addition, a fasting two-hour timed urine collection for urine creatinine, calcium, and typecollagen cross-linked N-telopeptides (“N-telopeptide”) were collected on days 0, 30, 90, 120, and 180. Other tests performed were as follows: (1) Hematology: hemoglobin, hematocrit, red blood cell count, platelets, white blood cell counts with differential analysis (neutrophils, lymphocytes, monocytes, eosinophils, and basophils); (2) Chemistry: alkaline phosphatase, alanine aminotransferase, serum glutamic pyruvic transaminase (“ALT/SGPT”), asparate aminotransferase/serum glutamin axaloacetic transaminase (“AST/SGOT”), total bilirubin, creatinine, glucose, and elecrolytes (sodium, potassium, choride, bicarbonate, calcium, and inorganic phosphorus); (3) Lipids: total cholesterol, high-density lipoprotein (“HDL”), low-density lipoprotein (“LDL”), and triglycerides; (4) Urinalysis: color, appearance, specific gravity, pH, protein, glucose, ketones, blood, bilirubin, and nitrites; and (5) Other: PSA (screening days 90-180), prolactin (screening), and testosterone (screening) including electrolytes, glucose, renal, and liver function tests and lipid profile, were performed at all clinic visits. Bone mineral density (“BMD”) was analyzed at day 0 and day 180.
  • A. AndroGel® and ANDRODERM® patch.
  • Approximately 250 g of AndroGel® was packaged in multidose glass bottles that delivered 2.25 g of the gel for each actuation of the pump. Patients assigned to apply 5.0 g/day of AndroGel® testosterone were given one bottle of AndroGel® and one bottle of placebo gel (containing vehicle but no testosterone), while those assigned to receive 10.0 g/day of AndroGel® were dispensed two bottles of the active AndroGel®. The patients were then instructed to apply the bottle contents to the right and left upper arms/shoulders and to the right and left sides of the abdomen on an alternate basis. For example, on the first day of the study, patients applied two actuations from one bottle, one each to the left and right upper arm/shoulder, and two actuations from the second bottle, one each to the left and right abdomen. On the following day of treatment, the applications were reversed. Alternate application sites continued throughout the study. After application of the gel to the skin, the gel dried within a few minutes. Patients washed their hands thoroughly with soap and water immediately after gel application.
  • The 7.5 g/day AndroGel® group received their dose in an open-label fashion. After 90 days, for the subjects titrated to the AndroGel® 7.5 g/day dose, the patients were supplied with three bottles, one containing placebo and the other two AndroGel®. The subjects were instructed to apply one actuation from the placebo bottle and three actuations from a AndroGel® bottle to four different sites of the body as above. The sites were rotated each day taking the same sequence as described above.
  • ANDRODERM® testosterone patches each delivering 2.5 mg/day of testosterone were provided to about one-third of the patients in the study. These patients were instructed to apply two testosterone patches to a clean, dry area of skin on the back, abdomen, upper arms, or thighs once per day. Application sites were rotated with approximately seven days interval between applications to the same site.
  • On study days when the patients were evaluated, the gel/patches were applied following pre-dose evaluations. On the remaining days, the testosterone gel or patches were applied at approximately 8:00 a.m. for 180 days.
  • B. Study Method and Results:
  • 1. Hormone Pharmacokinetics.
  • On days 0, 1, 30, 90, and 180, the patients had multiple blood samples for testosterone and free testosterone measurements at 30, 15 and 0 minutes before and 2, 4, 8, 12, 16, and 24 hours after AndroGel® or patch application. In addition, subjects returned on days 60, 120, and 150 for a single blood sampling prior to application of the gel or patch. Serum DHT, E2, FSH, LH and SHBG were measured on samples collected before gel application on days 0, 30, 60, 90, 120, 150, and 180. Sera for all hormones were stored frozen at 20° C. until assay. All samples for a patient for each hormone were measured in the same assay whenever possible. The hormone assays were then measured at the Endocrine Research Laboratory of the UCLA-Harbor Medical Center.
  • The following table summarizes the pharmacokinetic parameters were measured for each patient:
    TABLE 7
    Pharmacokinetic Parameters
    AUC0-24 area under the curve from 0 to 24 hours, determined
    using the linear trapezoidal rule.
    Cbase or Co Baseline concentration
    Cavg time-averaged concentration over the 24-hour dosing
    interval determined by AUC0-24/24
    Cmax maximum concentration during the 24-hour
    dosing interval
    Cmin minimum concentration during the 24-hour
    dosing interval
    Tmax time at which Cmax occurred
    Tmin time at which Cmin occurred
    Fluctuation extent of variation in the serum concentration over the
    Index course of a single day, calculated as (Cmax − Cmin)/Cavg
    Accumulation increase in the daily drug exposure with continued
    ratio dosing, calculated as the ratio of the AUC at
    steady on a particular day over the AUC on
    day 1 (eg., AUCday 30/AUCday 1)
    Net AUC0-24 AUC0-24 on days 30, 90, 180 - AUC0-24 on day 0
  • a. Testosterone Pharmacokinetics:
  • (1) Methods.
  • Serum testosterone levels were measured after extraction with ethylacetate and hexane by a specific radioimmunoassay (“RIA”) using reagents from ICN (Costa Mesa, Calif.). The cross reactivities of the antiserum used in the testosterone RIA were 2.0% for DHT, 2.3% for androstenedione, 0.8% for 3-Beta-androstanediol, 0.6% for etiocholanolone and less than 0.01% for all other steroids tested. The lower limit of quantitation (“LLQ”) for serum testosterone measured by this assay was 25 ng/dL (0.87 nmol/L). The mean accuracy of the testosterone assay, determined by spiking steroid free serum with varying amounts of testosterone (0.9to 52 nmol/L), was 104% and ranged from 92% to 117%. The intra-assay and inter-assay coefficients of the testosterone assay were 7.3 and 11.1%, respectively, at the normal adult male range. In normal adult men, testosterone concentrations range from 298 to 1,043 ng/dL (10.33 to 36.17 nmol/L) as determined at the UCLA-Harbor Medical Center.
  • (2) Baseline Concentration.
  • As shown in Table 8 and FIG. 5(a), at baseline, the average serum testosterone concentrations over 24 hours (Cavg) were similar in the groups and below the adult normal range. Moreover the variations of the serum concentration (based on maximum and minimum concentrations during the 24-hour period, Cmax and Cmin, respectively) during the day were also similar in the three groups. FIG. 5(a) shows that the mean testosterone levels had a maximum level between 8 to 10 a.m. (i.e., at 0 to 2 hours) and the minimum 8 to 12 hours later, demonstrating a mild diurnal variation of serum testosterone. About one-third of the patients in each group had Cavg within the lower normal adult male range on day 0 (24/73 for the 5.0 g/day AndroGel® group, 26/78 for the 10.0 g/day AndroGel® group, and 25/76 for testosterone patch group). All except three of the subjects met the enrollment criterion of serum testosterone less than 300 ng/dL (10.4 nmol/L) on admission.
    TABLE 8(a)
    Baseline Phamacokinetic Parameters by Initial
    Treatment Group (Mean ± SD)
    5.0 g/day T-Gel 10.0 g/day T-gel T-patch
    N 73 78 76
    Cavg (ng/dL) 237 ± 130 248 ± 140 237 ± 139
    Cmax (ng/dL) 328 ± 178 333 ± 194 314 ± 179
    Tmax*(hr)  4.0 (0.0-24.5) 7.9 (0.0-24.7) 4.0 (0.0-24.3)
    Cmin (ng/dL) 175 ± 104 188 ± 112 181 ± 112
    Tmin* (hr) 8.01 (0.0-24.1) 8.0 (0.0-24.0) 8.0 (0.0-23.9)
    Fluc Index (ratio) 0.627 ± 0.479 0.556 ± 0.384 0.576 ± 0.341

    *Median (Range*)
  • TABLE 8(b)
    Baseline Testosterone Pharmacokinetic Parameters by Final Treatment Group (Mean ± SD)
    Doses Received During Initial => Extended Treatment Phases
    5.0 g/day 5.0 => 7.5 g/day 10.0 => 7.5 g/day 10.0 g/day
    T-gel T-gel T-gel T-gel T-patch
    N 53 20 20 58 76
    Cavg (ng/dL) 247 ± 137 212 ± 109 282 ± 157 236 ± 133 237 ± 140
    Cmax (ng/dL) 333 ± 180 313 ± 174 408 ± 241 307 ± 170 314 ± 179
    Tmax* (hr) 4.0 (0.0-24.5) 4.0 (0.0-24.0) 19.7 (0.0-24.3)  4.0 (0.0-24.7) 4.0 (0.0-24.3)
    Cmin (ng/dL) 185 ± 111 150 ± 80  206 ± 130 182 ± 106 181 ± 112
    Tmin* (hr) 8.0 (0.0-24.1) 11.9 (0.0-24.0)  8.0 (0.0-23.3) 8.0 (0.0-24.0) 8.0 (0.0-23.9)
    Fluc Index (ratio) 0.600 ± 0.471 0.699 ± 0.503 0.678 ± 0.580 0.514 ± 0.284 0.576 ± 0.341

    *Median (range)
  • (3) Day 1.
  • FIG. 5(b) and Tables 8(c)-(d) show the pharmacokinetic profile for all three initial treatment groups after the first application of transdermal testosterone. In general, treatment with AndroGel® and the testosterone patch produced increases in testosterone concentrations sufficiently large to bring the patients into the normal range in just a few hours. However, even on day 1, the pharmacokinetic profiles were markedly different in the AndroGel® and patch groups. Serum testosterone rose most rapidly in the testosterone patch group reaching a maximum concentration (Cmax) at about 12 hours (Tmax). In contrast, serum testosterone rose steadily to the normal range after AndroGel® application with Cmax levels achieved by 22 and 16 hours in the 5.0 g/day AndroGel® group and the 10.0 g/day AndroGel® group, respectively.
    TABLE 8(c)
    Testosterone Pharmacokinetic Parameters on Day
    1 by Initial Treatment Group (Mean ± SD)
    5.0 g/day T-Gel 10.0 g/day T-gel T-patch
    N 73 76 74
    Cavg (ng/dL) 398 ± 156 514 + 227 482 ± 204
    Cmax (ng/dL) 560 ± 269 748 ± 349 645 ± 280
    Tmax*(hr) 22.1 (0.0-25.3) 16.0 (0.0-24.3) 11.8 (1.8-24.0)
    Cmin (ng/dL) 228 ± 122 250 ± 143 232 ± 132
    Tmin* (hr)  1.9 (0.0-24.0)  0.0 (0.0-24.2)  1.5 (0.0-24.0)

    *Median (Range)
  • TABLE 8(d)
    Testosterone Phamacokinetic Parameters on Day 1 by Final Treatment Group (Mean ± SD)
    Doses Received During Initial => Extended Treatment Phases
    5.0 g/day 5.0 => 7.5 g/day 10.0 => 7.5 g/day 10.0 g/day
    T-gel T-gel T-gel T-gel T-patch
    N 53 20 19 57 74
    Cavg (ng/dL) 411 ± 160 363 ± 143 554 ± 243 500 ± 223 482 ± 204
    Cmax (ng/dL) 573 ± 285 525 ± 223 819 ± 359 724 ± 346 645 ± 280
    Tmax* (hr) 22.1 (0.0-25.3)  19.5 (1.8-24.3)  15.7 (3.9-24.0)  23.0 (0.0-24.3)  11.8 (1.8-24.0) 
    Cmin (ng/dL) 237 ± 125 204 ± 112 265 ± 154 245 ± 140 232 ± 132
    Tmin* (hr) 1.8 (0.0-24.0) 3.5 (0.0-24.0) 1.9 (0.0-24.2) 0.0 (0.0-23.8) 1.5 (0.0-24.0)
    Fluc Index (ratio) 0.600 ± 0.471 0.699 ± 0.503 0.678 ± 0.580 0.514 ± 0.284 0.576 ± 0.341

    *Median (range)
  • (4) Days 30, 90, and 180.
  • FIGS. 5(c) and 5(d) show the unique 24-hour pharmacokinetic profile of AndroGel®-treated patients on days 30 and 90. In the AndroGel® groups, serum testosterone levels showed small and variable increases shortly after dosing. The levels then returned to a relatively constant level. In contrast, in the testosterone patch group, patients exhibited a rise over the first 8 to 12 hours, a plateau for another 8 hours, and then a decline to the baseline of the prior day. Further, after gel application on both days 30 and 90, the Cavg in the 10.0 g/day AndroGel® group was 1.4 fold higher than in the 5.0 g/day AndroGel® group and 1.9 fold higher than the testosterone patch group. The testosterone patch group also had a Cmin substantially below the lower limit of the normal range. On day 30, the accumulation ratio was 0.94 for testosterone patch group, showing no accumulation. The accumulation ratios at 1.54 and 1.9 were significantly higher in the 5.0 g/day AndroGel® group and 10.0 g/day AndroGel® group, respectively. The differences in accumulation ratio among the groups persisted on day 90. This data indicates that the AndroGel® preparations had a longer effective half-life than testosterone patch.
  • FIG. 5(e) shows the 24-hour pharmacokinetic profile for the treatment groups on day 180. In general, as Table 8(e) shows, the serum testosterone concentrations achieved and the pharmacokinetic parameters were similar to those on days 30 and 90 in those patients who continued on their initial randomized treatment groups. Table 8(f) shows that the patients titrated to the 7.5 g/day AndroGel® group were not homogeneous. The patients that were previously in the 10.0 g/day group tended to have higher serum testosterone levels than those previously receiving 5.0 g/day. On day 180, the Cavg in the patients in the 10.0 g/day group who converted to 7.5 g/day on day 90 was 744 ng/dL, which was 1.7 fold higher than the Cavg of 450in the patients titrated to 7.5 g/day from 5.0 g/day. Despite adjusting the dose up by 2.5 g/day in the 5.0 to 7.5 g/day group, the Cavg remained lower than those remaining in the 5.0 group. In the 10.0 to 7.5 g/day group, the Cavg became similar to those achieved by patients remaining in the 10.0 g/day group without dose titration. These results suggest that many of the under-responders may actually be poorly compliant patients. For example, if a patient does not apply AndroGel® properly (e.g., preferentially from the placebo container or shortly before bathing), then increasing the dose will not provide any added benefit.
  • FIGS. 5(f)-(h) compare the pharmacokinetic profiles for the 5.0 g/day AndroGel® group, the 10.0 g/day AndroGel® g/day group, and the testosterone patch group at days 0, 1, 30, 90, and 180, respectively. In general, the mean serum testosterone levels in the testosterone patch group remained at the lower limit of the normal range throughout the treatment period. In contrast, the mean serum testosterone levels remained at about 490-570 ng/dL for the 5.0 g/day AndroGel® group and about 630-860 ng/dL AndroGel® for the 10.0 g/day group.
    TABLE 8(e)
    Testosterone Phamacokinetic Parameters on Day
    1 by Initial Treatment Group (Mean ± SD)
    5.0 g/ 10.0 g/
    day T-Gel day T-gel T-patch
    Day 30 N = 66 N = 74 N = 70
    Cavg (ng/dL) 566 ± 262 792 ± 294 419 ± 163
    Cmax (ng/dL) 876 ± 466 1200 ± 482 576 ± 223
    Tmax*(hr) 7.9 (0.0-24.0) 7.8 (0.0-24.3) 11.3 (0.0-24.0) 
    Cmin (ng/dL) 361 ± 149 505 ± 233 235 ± 122
    Tmin* (hr) 8.0 (0.0-24.1) 8.0 (0.0-25.8) 2.0 (0.0-24.2)
    Fluc Index 0.857 ± 0.331 0.895 ± 0.434 0.823 ± 0.289
    (ratio)
    Accum Ratio 1.529 ± 0.726 1.911 ± 1.588 0.937 ± 0.354
    (ratio)
    Day 90 N = 65 N = 73 N = 64
    Cavg (ng/dL) 553 ± 247 792 ± 276 417 ± 157
    Cmax (ng/dL) 846 ± 444 1204 ± 570 597 ± 242
    Tmax*(hr) 4.0 (0.0-24.1) 7.9 (0.0-25.2) 8.1 (0.0-25.0)
    Cmin (ng/dL) 354 ± 147 501 ± 193 213 ± 105
    Tmin* (hr) 4.0 (0.0-25.3) 8.0 (0.0-24.8) 2.0 (0.0-24.0)
    Fluc Index 0.851 ± 0.402 0.859 ± 0.399 0.937 ± 0.442
    (ratio)
    Accum Ratio 1.615 ± 0.859 1.927 ± 1.310 0.971 ± 0.453
    (ratio)
    Day 180 N = 63 N = 68 N = 45
    Cavg (ng/dL) 520 ± 227 722 ± 242 403 ± 163
    Cmax (ng/dL) 779 ± 359 1091 ± 437 580 ± 240
    Tmax*(hr) 4.0 (0.0-24.0) 7.9 (0.0-24.0) 10.0 (0.0-24.0) 
    Cmin (ng/dL) 348 ± 164 485 ± 184 223 ± 114
    Tmin* (hr) 11.9 (0.0-24.0)  11.8 (0.0-27.4)  2.0 (0.0-25.7)
    Fluc Index 0.845 ± 0.379 0.829 ± 0.392 0.891 ± 0.319
    (ratio)
    Accum Ratio 1.523 ± 1.024 1.897 ± 2.123 0.954 ± 0.4105
    (ratio)

    *Median (Range)
  • TABLE 8(f)
    Testosterone Phamacokinetic Parameters on Days 30, 90, 180 by Final Treatment Group (Mean ± SD)
    Doses Received During Initial => Extended Treatment Phases
    5.0 g/day 5.0 => 7.5 g/day 10.0 => 7.5 g/day 10.0 g/day
    T-gel T-gel T-gel T-gel T-patch
    Day 30 N = 47 N = 19 N = 19 N = 55 N = 70
    Cavg (ng/dL) 604 ± 288 472 ± 148 946 ± 399 739 ± 230 419 ± 163
    Cmax (ng/dL) 941 ± 509 716 ± 294 1409 ± 556  1128 ± 436  576 ± 223
    Tmax* (hr) 7.9 (0.0-24.0) 8.0 (0.0-24.0) 8.0 (0.0-24.3) 7.8 (0.0-24.3) 11.3 (0.0-24.0) 
    Cmin (ng/dL) 387 ± 159 296 ± 97  600 ± 339 471 ± 175 235 ± 122
    Tmin* (hr) 8.1 (0.0-24.1) 1.7 (0.0-24.1) 11.4 (0.0-24.1)  8.0 (0.0-25.8) 2.0 (0.0-24.2)
    Fluc Index (ratio) 0.861 ± 0.341 0.846 ± 0.315 0.927 ± 0.409 0.884 ± 0.445 0.823 ± 0.289
    Accum Ratio (ratio) 1.543 ± 0.747 1.494 ± 0.691 2.053 ± 1.393 1.864 ± 1.657 0.937 ± 0.354
    Day 90 N = 45 N = 20 N = 18 N = 55 N = 64
    Cavg (ng/dL) 596 ± 266 455 ± 164 859 ± 298 771 ± 268 417 ± 157
    Cmax (ng/dL) 931 ± 455 654 ± 359 1398 ± 733  1141 ± 498  597 ± 242
    Tmax* (hr) 3.8 (0.0-24.1) 7.7 (0.0-24.0) 7.9 (0.0-24.0) 7.9 (0.0-25.2) 8.1 (0.0-25.0)
    Cmin (ng/dL) 384 ± 147 286 ± 125 532 ± 181 492 ± 197 213 ± 105
    Tmin* (hr) 7.9 (0.0-25.3) 0.0 (0.0-24.0) 12.0 (0.0-24.1)  4.0 (0.0-24.8) 2.0 (0.0-24.0)
    Fluc Index (ratio) 0.886 ± 0.391 0.771 ± 0.425 0.959 ± 0.490 0.826 ± 0.363 0.937 ± 0.442
    Accum Ratio (ratio) 1.593 ± 0.813 1.737 ± 1.145 1.752 ± 0.700 1.952 ± 1.380 0.971 ± 0.453
    Day 180 N = 44 N = 18 N = 19 N = 48 N = 41
    Cavg (ng/dL) 555 ± 225 450 ± 219 744 ± 320 713 ± 209 408 ± 165
    Cmax (ng/dL) 803 ± 347 680 ± 369 1110 ± 468  1083 ± 434  578 ± 245
    Tmax* (hr) 5.8 (0.0-24.0) 2.0 (0.0-24.0) 7.8 (0.0-24.0) 7.7 (0.0-24.0) 10.6 (0.0-24.0) 
    Cmin (ng/dL) 371 ± 165 302 ± 150 505 ± 233 485 ± 156 222 ± 116
    Tmin* (hr) 11.9 (0.0-24.0)  9.9 (0.0-24.0) 12.0 (0.0-24.0)  8.0 (0.0-27.4) 2.0 (0.0-25.7)
    Fluc Index (ratio) 0.853 ± 0.402 0.833 ± 0.335 0.824 ± 0.298 0.818 ± 0.421 0.866 ± 0.311
    Accum Ratio (ratio) 1.541 ± 0.917 NA NA 2.061 ± 2.445 0.969 ± 0.415

    *Median (range)
  • (5) Dose Proportionality for AndroGel®.
  • Table 8(g) shows the increase in AUC0-24 on days 30, 90, and 180 from the pretreatment baseline (net AUC0-24). In order to assess dose-proportionality, the bioequivalence assessment was performed on the log-transformed AUCs using “treatment” as the only factor. The AUCs were compared after subtracting away the AUC contribution from the endogenous secretion of testosterone (the AUC on day 0) and adjusting for the two-fold difference in applied doses. The AUC ratio on day 30 was 0.95 (90% C.I.: 0.75-1.19) and on day 90 was 0.92 (90% C.I.: 0.73-1.17). When the day 30 and day 90 data was combined, the AUC ratio was 0.93 (90% C.I.: 0.79-1.10).
  • The data shows dose proportionality for AndroGel® treatment. The geometric mean for the increase in AUC0-24 from day 0 to day 30 or day 90 was twice as great for the 10.0 g/day group as for the 5.0 g/day group. A 125 ng/dL mean increase in serum testosterone Cavg level was produced by each 2.5 g/day of AndroGel®. In other words, the data shows that 0.1 g/day of AndroGel® produced, on the average, a 5 ng/dL increase in serum testosterone concentration. This dose proportionality aids dosing adjustment by the physician. Because AndroGel® is provided in 2.5 g packets (containing 25 mg of testosterone), each 2.5 g packet will produce, on average, a 125 ng/dL increase in the Cavg for serum total testosterone.
    TABLE 8(g)
    Net AUC0-24 (nmol*h/L) on Days 30, 90, and
    180 after Transdermal Testosterone Application
    T Patch T gel 5.0 g/day T gel 10.0 g/day
    Day
    30 154 ± 18 268 ± 28 446 ± 30
    Day 90 157 ± 20 263 ± 29 461 ± 28
    Day 180 160 ± 25 250 ± 32 401 ± 27
  • The increase in AUC0-24 from pretreatment baseline achieved by the 10.0 g/day and the 5.0 g/day groups were approximately 2.7 and 1.7 fold higher than that resulting from application of the testosterone patch.
  • b. Pharmacokinetics of Serum Free Testosterone Concentration:
  • (1) Methods.
  • Serum free testosterone was measured by RIA of the dialysate, after an overnight equilibrium dialysis, using the same RIA reagents as the testosterone assay. The LLQ of serum free testosterone, using the equilibrium dialysis method, was estimated to be 22 pmol/L. When steroid free serum was spiked with increasing doses of testosterone in the adult male range, increasing amounts of free testosterone were recovered with a coefficient of variation that ranged from 11.0-18.5%. The intra- and interassay coefficients of free testosterone were 15% and 16.8% for adult normal male values, respectively. As estimated by the UCLA-Harbor Medical Center, free testosterone concentrations range from 3.48-17.9 ng/dL (121-620 pmol/L) in normal adult men.
  • (2) Pharmacokinetic Results.
  • In general, as shown in Table 9, the pharmacokinetic parameters of serum free testosterone mirrored that of serum total testosterone as described above. At baseline (day 0), the mean serum free testosterone concentrations (Cavg) were similar in all three groups which were at the lower limit of the adult male range. The maximum serum free testosterone concentration occurred between 8 and 10 a.m., and the minimum about 8 to 16 hours later. This data is consistent with the mild diurnal variation of serum testosterone.
  • FIG. 6(a) shows the 24-hour pharmacokinetic profiles for the three treatment groups on day 1. After application of the testosterone patch, the serum free testosterone levels peaked at 12 about 4 hours earlier than those achieved by the AndroGel® groups The serum free testosterone levels then declined in the testosterone patch group whereas in the AndroGel® groups, the serum free testosterone levels continued to rise.
  • FIGS. 6(b) and 6(c) show the pharmacokinetic profiles of free testosterone in the AndroGel®-treated groups resembled the unique testosterone profiles on days 30 and 90. After AndroGel® application, the mean serum free testosterone levels in the three groups were within normal range. Similar to the total testosterone results, the free testosterone Cavg achieved by the 10.0 g/day group was 1.4 fold higher than the 5.0 g/day group and 1.7 fold higher than the testosterone patch group. Moreover, the accumulation ratio for the testosterone patch was significantly less than that of the 5.0 g/day AndroGel® group and the 10.0 g/day AndroGel® group.
  • FIG. 6(d) shows the free testosterone concentrations by final treatment groups on day 180. In general, the free testosterone concentrations exhibited a similar pattern as serum testosterone. The 24-hour pharmacokinetic parameters were similar to those on days 30 and 90 in those subjects who remained in the three original randomized groups. Again, in the subjects titrated to receive 7.5 g/day of AndroGel®, the group was not homogenous. The free testosterone Cavg in the patients with doses adjusted upwards from 5.0 to 7.5 g/day remained 29% lower than those of subjects remaining in the 5.0 g/day group. The free testosterone Cavg in the patients whose doses were decreased from 10.0 to 7.5 g/day was 11% higher than those in remaining in the 10.0 g/day group.
  • FIGS. 6(e)-(g) show the free testosterone concentrations in the three groups of subjects throughout the 180-day treatment period. Again, the free testosterone levels followed that of testosterone. The mean free testosterone levels in all three groups were within the normal range with the 10.0 g/day group maintaining higher free testosterone levels than both the 5.0 g/day and the testosterone patch groups.
    TABLE 9
    Free Testosterone Pharmacokinetic Parameters by Final Treatment (Mean ± SD)
    Doses Received During Initial =>Extended Treatment Phases
    5.0 g/day 5.0 => 7.5 g/day 10.0 => 7.5 g/day 10/0 g/day
    T-gel T-gel T-gel Tgel T-patch
    Day 0 N = 53 N = 20 N = 20 N = 58 N = 76
    Cavg (ng/dL) 4.52 ± 3.35 4.27 ± 3.45 4.64 ± 3.10 4.20 ± 3.33 4.82 ± 3.64
    Cmax (ng/dL) 5.98 ± 4.25 6.06 ± 5.05 6.91 ± 4.66 5.84 ± 4.36 6.57 ± 4.90
    Tmax* (hr) 4.0 (0.0-24.5) 2.0 (0.0-24.0) 13.5 (0.0-24.2)  2.1 (0.0-24.1) 3.8 (0.0-24.0)
    Cmin (ng/dL) 3.23 ± 2.74 3.10 ± 2.62 3.14 ± 2.14 3.12 ± 2.68 3.56 ± 2.88
    Tmin* (hr) 8.0 (0.0-24.2) 9.9 (0.0-16.0) 4.0 (0.0-23.3) 8.0 (0.0-24.0) 7.9 (0.0-24.0)
    Fluc Index (ratio) 0.604 ± 0.342 0.674 ± 0.512 0.756 ± 0.597 0.634 ± 0.420 0.614 ± 0.362
    Day 1 N = 53 N = 20 N = 19 N = 57 N = 74
    Cavg (ng/dL) 7.50 ± 4.83 6.80 ± 4.82 9.94 ± 5.04 8.93 ± 6.09 9.04 ± 4.81
    Cmax (ng/dL) 10.86 ± 7.45  10.10 ± 7.79  15.36 ± 7.31  13.20 ± 8.61  12.02 ± 6.14 
    Tmax* (hr) 16.0 (0.0-25.3)  13.9 (0.0-24.3)  15.7 (2.0-24.0)  23.5 (1.8-24.3)  12.0 (1.8-24.0) 
    Cmin (ng/dL) 4.30 ± 3.33 3.69 ± 3.24 3.88 ± 2.73 4.40 ± 3.94 4.67 ± 3.52
    Tmin* (hr) 0.0 (0.0-24.1) 1.8 (0.0-24.0) 0.0 (0.0-24.2) 0.0 (0.0-23.9) 0.0 (0.0-24.0)
    Day 30 N = 47 N = 19 N = 19 N = 55 N = 70
    Cavg (ng/dL) 11.12 ± 6.22  7.81 ± 3.94 16.18 ± 8.18  13.37 ± 7.13  8.12 ± 4.15
    Cmax (ng/dL) 16.93 ± 10.47 11.62 ± 6.34  25.14 ± 10.80 19.36 ± 9.75  11.48 ± 5.78 
    Tmax* (hr) 8.0 (0.0-27.8) 8.0 (0.0-26.3) 8.0 (0.0-24.3) 8.0 (0.0-24.3) 8.0 (0.0-24.0)
    Cmin (ng/dL) 6.99 ± 3.82 4.78 ± 3.10 9.99 ± 7.19 8.25 ± 5.22 4.31 ± 3.20
    Tmin* (hr) 4.0 (0.0-24.1) 3.5 (0.0-24.1) 11.4 (0.0-24.1)  7.8 (0.0-25.8) 2.0 (0.0-24.8)
    Fluc Index (ratio) 0.853 ± 0.331 0.872 ± 0.510 1.051 ± 0.449 0.861 ± 0.412 0.929 ± 0.311
    Accum Ratio (ratio) 1.635 ± 0.820 1.479 ± 0.925 2.065 ± 1.523 1.953 ± 1.626 0.980 ± 0.387
    Day 90 N = 45 N = 20 N = 18 N = 55 N = 64
    Cavg (ng/dL) 12.12 ± 7.78  8.06 ± 3.78 17.65 ± 8.62  13.11 ± 5.97  8.50 ± 5.04
    Cmax (ng/dL) 18.75 ± 12.90 10.76 ± 4.48  25.29 ± 12.42 18.61 ± 8.20  12.04 ± 6.81 
    Tmax* (hr) 4.0 (0.0-24.0) 9.7 (0.0-24.0) 8.0 (0.0-24.0) 8.0 (0.0-25.2) 11.6 (0.0-25.0) 
    Cmin (ng/dL) 7.65 ± 4.74 4.75 ± 2.86 10.56 ± 6.07  8.40 ± 4.57 4.38 ± 3.70
    Tmin* (hr) 8.0 (0.0-24.0) 1.9 (0.0-24.0) 5.9 (0.0-24.1) 4.0 (0.0-24.8) 2.0 (0.0-24.1)
    Fluc Index (ratio) 0.913 ± 0.492 0.815 ± 0.292 0.870 ± 0.401 0.812 ± 0.335 0.968 ± 0.402
    Accum Ratio (ratio) 1.755 ± 0.983 1.916 ± 1.816 1.843 ± 0.742 2.075 ± 1.866 1.054 ± 0.498
    Day 180 N = 44 N = 18 N = 19 N = 48 N = 41
    Cavg (ng/dL) 11.01 ± 5.24  7.80 ± 4.63 14.14 ± 7.73  12.77 ± 5.70  7.25 ± 4.90
    Cmax (ng/dL) 16.21 ± 7.32  11.36 ± 6.36  22.56 ± 12.62 18.58 ± 9.31  10.17 ± 5.90 
    Tmax* (hr) 7.9 (0.0-24.0) 2.0 (0.0-23.9) 7.8 (0.0-24.0) 8.0 (0.0-24.0) 11.1 (0.0-24.0) 
    Cmin (ng/dL) 7.18 ± 3.96 5.32 ± 4.06 9.54 ± 6.45 8.23 ± 4.01 3.90 ± 4.20
    Tmin* (hr) 9.9 (0.0-24.2) 7.9 (0.0-24.0) 8.0 (0.0-23.2) 11.8 (0.0-27.4)  2.5 (0.0-25.7)
    Fluc Index (ratio) 0.897 ± 0.502 0.838 ± 0.378 0.950 ± 0.501 0.815 ± 0.397 0.967 ± 0.370
    Accum Ratio (ratio) 1.712 ± 1.071 NA NA 2.134 ± 1.989 1.001 ± 0.580

    *Median (Range)
  • C. Serum DHT Concentrations.
  • Serum DHT was measured by RIA after potassium permanganate treatment of the sample followed by extraction. The methods and reagents of the DHT assay were provided by DSL (Webster, Tex.). The cross reactivities of the antiserum used in the RIA for DHT were 6.5% for 3-β-androstanediol, 1.2% for 3-α-androstanediol, 0.4% for 3-α-androstanediol glucuronide, and 0.4% for testosterone (after potassium permanganate treatment and extraction), and less than 0.01% for other steroids tested. This low cross-reactivity against testosterone was further confirmed by spiking steroid free serum with 35 nmol/L (1,000 pg/dL) of testosterone and taking the samples through the DHT assay. The results even on spiking with over 35 nmol/L of testosterone was measured as less than 0.1 nmol/L of DHT. The LLQ of serum DHT in the assay was 0.43 nmol/L. The mean accuracy (recovery) of the DHT assay determined by spiking steroid free serum with varying amounts of DHT from 0.43 nmol/L to 9 nmol/L was 101% and ranged from 83 to 114%. The intra-assay and inter-assay coefficients of variation for the DHT assay were 7.8 and 16.6%, respectively, for the normal adult male range. The normal adult male range of DHT was 30.7-193.2 ng/dL (1.06 to 6.66 nmol/L) as determined by the UCLA-Harbor Medical Center.
  • As shown in Table 10, the pretreatment mean serum DHT concentrations were between 36 and 42 ng/dL, which were near the lower limit of the normal range in all three initial treatment groups. None of the patients had DHT concentrations above the upper limit of the normal range on the pretreatment day, although almost half (103 patients) had concentrations less than the lower limit.
  • FIG. 7 shows that after treatment, the differences between the mean DHT concentrations associated with the different treatment groups were statistically significant, with patients receiving AndroGel® having a higher mean DHT concentration than the patients using the patch and showing dose-dependence in the mean serum DHT concentrations. Specifically, after testosterone patch application mean serum DHT levels rose to about 1.3 fold above the baseline. In contrast, serum DHT increased to 3.6 and 4.8 fold above baseline after application of 5.0 and 10.0 g/day of AndroGel®, respectively.
    TABLE 10
    DHT Concentrations (ng/dL) on Each of the Observation Days By Initial Treatment (Mean ± SD)
    Day 0 Day 30 Day 60 Day 90 Day 120 Day 150 Day 180
    5.0 g/day N = 73 N = 69 N = 70 N = 67 N = 65 N = 63 N = 65
    T-gel 36.0 ± 19.9 117.6 ± 74.9  122.4 ± 99.4  130.1 ± 99.2  121.8 ± 89.2 144.7 ± 110.5 143.7 ± 105.9
    10.0 g/day N = 78 N = 78 N = 74 N = 75 N = 68 N = 67 N = 71
    T-gel 42.0 ± 29.4 200.4 ± 127.8 222.0 ± 126.6 207.7 ± 111.0 187.3 ± 97.3 189.1 ± 102.4 206.1 ± 105.9
    T-Patch N = 76 N = 73 N = 68 N = 66 N = 49 N = 46 N = 49
    37.4 ± 21.4 50.8 ± 34.6 49.3 ± 27.2 43.6 ± 26.9  53.0 ± 52.8 54.0 ± 42.5 52.1 ± 34.3
    Across RX 0.6041 0.0001 0.0001 0.0001 0.0001 0.0001 0.0001
  • The increase in DHT concentrations are likely attributed to the concentration and location of 5α-reductase in the skin. For example, the large amounts of 5α-reductase in the scrotal skin presumably causes an increase in DHT concentrations in the TESTODERM® patch. In contrast, the ANDRODERM® and TESTODERM TTS® patches create little change in DTH levels because the surface area of the patch is small and little 5α-reductase is located in nonscrotal skin. AndroGel® presumably causes an increase in DHT levels because the gel is applied to a relatively large skin area and thus exposes testosterone to greater amounts of the enzyme.
  • To date, elevated DHT levels have not been reported to have any adverse clinical effects. Moreover, there is some evidence to suggest that increased DHT levels may inhibit prostate cancer.
  • d. DHT/T Ratio.
  • The UCLA-Harbor Medical Center reports a DHT/T ratio of 0.052-0.328 for normal adult men. In this example, the mean ratios for all three treatments were within the normal range on day 0. As shown in FIG. 8 and Table 11, there were treatment and concentration-dependent increases observed over the 180-day period. Specifically, the AndroGel® treatment groups showed the largest increase in DHT/T ratio. However, the mean ratios for all of the treatment groups remained within the normal range on all observation days.
    TABLE 11
    DHT/T Ratio on Each of the Observation Days By Initial Treatment (Mean ± SD)
    Day 0 Day 30 Day 60 Day 90 Day 120 Day 150 Day 180
    5.0 g/day N = 73 N = 68 N = 70 N = 67 N = 65 N = 62 N = 64
    T-gel 0.198 ± 0.137 0.230 ± 0.104 0.256 ± 0.132 0.248 ± 0.121 0.266 ± 0.119 0.290 ± 0.145 0.273 ± 0.160
    10.0 g/day N = 78 N = 77 N = 74 N = 74 N = 68 N = 67 N = 71
    T-gel 0.206 ± 0.163 0.266 ± 0.124 0.313 ± 0.160 0.300 ± 0.131 0.308 ± 0.145 0.325 ± 0.142 0.291 ± 0.124
    T-Patch N = 76 N = 73 N = 68 N = 65 N = 49 N = 46 N = 46
    0.204 ± 0.135 0.192 ± 0.182 0.175 ± 0.102 0.175 ± 0.092 0.186 ± 0.134 0.223 ± 0.147 0.212 ± 0.160
    Across RX 0.7922 0.0001 0.0001 0.0001 0.0001 0.0001 0.0002
  • e. Total Androgen (DHT+T).
  • The UCLA-Harbor Medical Center has determined that the normal total androgen concentration is 372 to 1,350 ng/dL. As shown in FIG. 9 and Table 12, the mean pre-dose total androgen concentrations for all three treatments were below the lower limit of the normal range on pretreatment day 0. The total androgen concentrations for both AndroGel® groups were within the normal range on all treatment observation days. In contrast, the mean concentrations for patients receiving the testosterone patch was barely within the normal range on day 60 and 120, but were below the lower normal limit on days 30, 90, 150, and 180.
    TABLE 12
    Total Androgen (DHT + T) (ng/dL) on Each of the Observation
    Days By Initial Treatment (Mean ± SD)
    Day 0 Day 30 Day 60 Day 90 Day 120 Day 150 Day 180
    5.0 g/day N = 73 N = 68 N = 70 N = 67 N = 65 N = 62 N = 64
    T-gel 281 ± 150 659 ± 398 617 ± 429 690 ± 431 574 ± 331 631 ± 384 694 ± 412
    10.0 g/day N = 78 N = 77 N = 74 N = 74 N = 68 N = 67 N = 71
    T-gel 307 ± 180 974 ± 532 1052 ± 806  921 ± 420 827 ± 361 805 ± 383 944 ± 432
    T-Patch N = 76 N = 73 N = 68 N = 65 N = 49 N = 46 N = 46
    282 ± 159 369 ± 206 392 ± 229 330 ± 173 378 ± 250 364 ± 220 355 ± 202
    Across RX 0.7395 0.0001 0.0001 0.0001 0.0001 0.0001 0.0001
  • f. E2 Concentrations.
  • Serum E2 levels were measured by a direct assay without extraction with reagents from ICN (Costa Mesa, Calif.). The intra-assay and inter-assay coefficients of variation of E2 were 6.5 and 7.1% respectively. The UCLA-Harbor Medical Center reported an average E2 concentration ranging from 7.1 to 46.1 pg/mL (63 to 169 pmol/L) for normal adult male range. The LLQ of the E2 was 18 pmol/L. The cross reactivities of the E2 antibody were 6.9% for estrone, 0.4% for equilenin, and less than 0.01% for all other steroids tested. The accuracy of the E2 assay was assessed by spiking steroid free serum with increasing amount of E2 (18 to 275 pmol/L). The mean recovery of E2 compared to the amount added was 99.1 % and ranged from 95 to 101 %.
  • FIG. 10 depicts the E2 concentrations throughout the 180-day study. The pretreatment mean E2 concentrations for all three treatment groups were 23-24 pg/mL. During the study, the E2 levels increased by an average 9.2% in the testosterone patch during the treatment period, 30.9% in the 5.0 g/day AndroGel® group, and 45.5% in the 10.0 g/day AndroGel® group. All of the mean concentrations fell within the normal range.
    TABLE 13
    Estradiol Concentration (pg/mL) on Each of the Observation Days By Initial Treatment (Mean ± SD)
    Day 0 Day 30 Day 60 Day 90 Day 120 Day 150 Day 180
    5.0 g/day N = 73 N = 69 N = 68 N = 67 N = 64 N = 65 N = 65
    T-gel 23.0 ± 9.2 29.2 ± 11.0 28.1 ± 10.0 31.4 ± 11.9 28.8 ± 9.9 30.8 ± 12.5 32.3 ± 13.8
    10.0 g/day N = 78 N = 78 N = 74 N = 75 N = 71 N = 66 N = 71
    T-gel 24.5 ± 9.5 33.7 ± 11.5 36.5 ± 13.5 37.8 ± 13.3  34.6 ± 10.4 35.0 ± 11.1 36.3 ± 13.9
    T-Patch N = 76 N = 72 N = 68 N = 66 N = 50 N = 49 N = 49
    23.8 ± 8.2 25.8 ± 9.8  24.8 ± 8.0  25.7 ± 9.8  25.7 ± 9.4 27.0 ± 9.2  26.9 ± 9.5 
    Across RX 0.6259 0.0001 0.0001 0.0001 0.0001 0.0009 0.0006
  • E2 is believed to be important for the maintenance of normal bone. In addition, E2 has a positive effect on serum lipid profiles.
  • g. Serum SHBG Concentrations.
  • Serum SHBG levels were measured with a fluoroimmunometric assay (“FIA”) obtained from Delfia (Wallac, Gaithersberg, Md.). The intra- and interassay coefficients were 5% and 12% respectively. The LLQ was 0.5 nmol/L. The UCLA-Harbor Medical Center determined that the adult normal male range for the SHBG assay is 0.8 to 46.6 nmol/L.
  • As shown in FIG. 11 and Table 11, the serum SHBG levels were similar and within the normal adult male range in the three treatment groups at baseline. None of the treatment groups showed major changes from the baseline on any of the treatment visit days. After testosterone replacement, serum SHBG levels showed a small decrease in all three groups. The most marked change occurred in the 10.0 g/day AndroGel® group.
    TABLE 14
    SHBG Concentration (nmol/L) on Each of the Observation Days By Initial Treatment (Mean ± SD)
    Day 0 Day 30 Day 60 Day 90 Day 120 Day 150 Day 180
    5.0 g/day N = 73 N = 69 N = 69 N = 67 N = 66 N = 65 N = 65
    T-gel 26.2 ± 14.9 24.9 ± 14.0 25.9 ± 14.4 25.5 ± 14.7 25.2 ± 14.1 24.9 ± 12.9 24.2 ± 13.6
    10.0 g/day N = 78 N = 78 N = 75 N = 75 N = 72 N = 68 N = 71
    T-gel 26.6 ± 17.8 24.8 ± 14.5 25.2 ± 15.5 23.6 ± 14.7 25.5 ± 16.5 23.8 ± 12.5 24.0 ± 14.5
    T-Patch N = 76 N = 72 N = 68 N = 66 N = 50 N = 49 N = 49
    30.2 ± 22.6 28.4 ± 21.3 28.2 ± 23.8 28.0 ± 23.6 26.7 ± 16.0 26.7 ± 16.4 25.8 ± 15.1
    Across RX 0.3565 0.3434 0.5933 0.3459 0.8578 0.5280 0.7668
  • h. Gonadotropins.
  • Serum FSH and LH were measured by highly sensitive and specific solid-phase FIA assays with reagents provided by Delfia (Wallac, Gaithersburg, Md.). The intra-assay coefficient of variations for LH and FSH fluroimmunometric assays were 4.3 and 5.2%, respectively; and the interassay variations for LH and FSH were 11.0% and 12.0%, respectively. For both LH and FSH assays, the LLQ was determined to be 0.2 IU/L. All samples obtained from the same subject were measured in the same assay. The UCLA-Harbor Medical Center reports that the adult normal male range for LH is 1.0-8.1 U/L and for FSH is 1.0-6.9U/L.
  • (1) FSH.
  • Table 15(a)-(d) shows the concentrations of FSH throughout the 180-day treatment depending on the cause of hypogonadism: (1) primary, (2) secondary, (3) age-associated, or (4) unknown.
  • As discussed above, patients with primary hypogonadism have an intact feedback inhibition pathway, but the testes do not secrete testosterone. As a result, increasing serum testosterone levels should lead to a decrease in the serum FSH concentrations. In this example, a total of 94 patients were identified as having primary hypogonadism. For these patients, the mean FSH concentrations in the three treatment groups on day 0 were 21-26 mlU/mL, above the upper limit of the normal range. As shown in FIG. 12(a) and Table 15(a), the mean FSH concentrations decreased during treatment in all three treatment regimens. However, only the 10.0 g/day AndroGel® group reduced the mean concentrations to within the normal range during the first 90 days of treatment. Treatment with the 10.0 g/day AndroGel® group required approximately 120 days to reach steady state. The mean FSH concentration in patients applying 5.0 g/day of AndroGel® showed an initial decline that was completed by day 30 and another declining phase at day 120 and continuing until the end of treatment. Mean FSH concentrations in the patients receiving the testosterone patch appeared to reach steady state after 30 days but were significantly higher than the normal range.
    TABLE 15(a)
    FSH Concentrations (mlU/mL) on Each of the Observation
    Days by Initial Treatment Group for Patients Having
    Primary Hypogonadism (Mean ± SD)
    N 5 g/day N 10 g/day N T-patch
    Day
    0 26 21.6 ± 21.0 33 20.9 ± 15.9  34 25.5 ± 25.5
    Day 30 23 10.6 ± 15.0 34 10.6 ± 14.1  31 21.4 ± 24.6
    Day 60 24 10.8 ± 16.9 32 7.2 ± 12.6 31 21.7 ± 23.4
    Day 90 24 10.4 ± 19.7 31 5.7 ± 10.1 30 19.5 ± 20.0
    Day 120 24  8.1 ± 15.2 28 4.6 ± 10.2 21 25.3 ± 28.4
    Day 150 22  6.7 ± 15.0 29 5.3 ± 11.0 21 18.6 ± 24.0
    Day 180 24  6.2 ± 11.3 28 5.3 ± 11.2 22 24.5 ± 27.4
  • Patients with secondary hypogonadism have a deficient testosterone negative feedback system. As shown in FIG. 12(b), of 44 patients identified as having secondary hypogonadism, the mean FSH concentrations decreased during treatment, although the decrease over time was not statistically significant for the testosterone patch. The patients in the 5.0 g/day AndroGel® group showed a decrease in the mean FSH concentration by about 35% by day 30, with no further decrease evident by day 60. Beyond day 90, the mean FSH concentration in the patients appeared to slowly return toward the pretreatment value. By day 30, all of the 10.0 g/day AndroGel® group had FSH concentrations less than the lower limit.
    TABLE 15(b)
    FSH Concentrations (mlU/mL) on Each of the Observation
    Days by Initial Treatment Group for Patients Having
    Secondary Hypogonadism (Mean ± SD)
    N 5 g/day N 10 g/day N T-patch
    Day
    0 17 4.2 ± 6.6 12 2.1 ± 1.9 15 5.1 ± 9.0
    Day 30 16 2.8 ± 5.9 12 0.2 ± 0.1 14 4.2 ± 8.0
    Day 60 17 2.8 ± 6.1 12 0.2 ± 0.1 13 4.2 ± 7.4
    Day 90 15 2.9 ± 5.6 12 0.2 ± 0.1 14 4.9 ± 9.0
    Day 120 14 3.0 ± 6.1 12 0.1 ± 0.1 12  6.1 ± 10.7
    Day 150 14 3.5 ± 7.5 12 0.2 ± 0.2 11 4.6 ± 6.5
    Day 180 14 3.7 ± 8.6 12 0.1 ± 0.1 12 4.9 ± 7.4
  • Twenty-five patients were diagnosed with age-associated hypogonadism. As shown in FIG. 12(c), the 5.0 g/day AndroGel® group had a mean pretreatment FSH concentration above the normal range. The mean concentration for this group was within the normal range by day 30 and had decreased more than 50% on days 90 and 180. The decrease in FSH mean concentration in the 10.0 g/day AndroGel® group showed a more rapid response. The concentrations in all six patients decreased to below the lower normal limit by day 30 and remained there for the duration of the study. The six patients who received the testosterone patch exhibited no consistent pattern in the mean FSH level; however, there was an overall trend towards lower FHS levels with continued treatment.
    TABLE 15(c)
    FSH Concentrations (mlU/mL) on Each of the Observation
    Days by Initial Treatment Group for Patients Having
    Age-Related Hypogonadism (Mean ± SD)
    N 5 g/day N 10 g/day N T-patch
    Day
    0 13 8.0 ± 9.1 6 5.2 ± 1.9 6 4.7 ± 1.7
    Day 30 12 4.6 ± 7.4 6 0.4 ± 0.3 6 3.7 ± 2.0
    Day 60 12 3.9 ± 6.6 6 0.3 ± 0.3 4 4.3 ± 3.3
    Day 90 11 3.8 ± 7.0 6 0.4 ± 0.7 4 3.5 ± 1.9
    Day 120 11 4.2 ± 8.3 6 0.4 ± 0.7 4 4.2 ± 3.3
    Day 150 11 4.3 ± 8.1 5 0.2 ± 0.2 4 3.4 ± 2.7
    Day 180 11 4.0 ± 7.2 6 0.2 ± 0.2 4 2.7 ± 2.1
  • Sixty-four patients in the study suffered from unclassified hypogonadism. As shown in FIG. 12(d), the patients showed a marked and comparatively rapid FSH concentration decrease in all three groups, with the greatest decrease being in the 10.0 g/day AndroGel® group. The 10.0 AndroGel® group produced nearly a 90% decrease in the mean FSH concentration by day 30 and maintained the effect to day 180. The 5.0 g/day AndroGel® group produced about a 75% drop in mean FSH concentration by day 30 and stayed at that level for the remainder of treatment. The 21 patients receiving the testosterone patch had a 50% decrease in the mean FSH concentration by day 30, a trend that continued to day 90 when the concentration was about one-third of its pretreatment value.
    TABLE 15(d)
    Concentrations (mlU/mL) for FSH on Each of the Observation
    Days by Initial Treatment Group for Patients Having
    Unknown-Related Hypogonadism (Mean ± SD)
    N 5 g/day N 10 g/day N T-patch
    Day
    0 17 4.0 ± 1.8 26 4.1 ± 1.6 21 3.7 ± 1.4
    Day 30 17 1.1 ± 1.0 26 0.5 ± 0.5 21 1.8 ± 0.8
    Day 60 16 1.1 ± 1.1 26 0.3 ± 0.3 18 1.6 ± 1.0
    Day 90 17 1.1 ± 1.1 25 0.4 ± 0.7 18 1.2 ± 0.9
    Day 120 16 1.2 ± 1.4 26 0.4 ± 0.6 12 1.4 ± 1.0
    Day 150 17 1.4 ± 1.4 23 0.3 ± 0.5 13 1.4 ± 1.2
    Day 180 16 1.0 ± 0.9 24 0.4 ± 0.4 11 1.3 ± 0.9
  • This data shows that feedback inhibition of FSH secretion functioned to some extent in all four subpopulations. The primary hypogonadal population showed a dose-dependency in both the extent and rate of the decline in FSH levels. The sensitivity of the feedback process appeared to be reduced in the secondary and age-associated groups in that only the highest testosterone doses had a significant and prolonged impact on FSH secretion. In contrast, the feedback inhibition pathway in the patients in the unclassified group was quite responsive at even the lowest dose of exogenous testosterone.
  • (2) LH.
  • The response of LH to testosterone was also examined separately for the same four subpopulations. Tables 16(a)-(d) shows the LH concentrations throughout the treatment period.
  • As shown in FIG. 13(a) and Table 16(a), the LH concentrations prior to treatment were about 175% of the upper limit of the normal range in primary hypogonadal patients. The mean LH concentrations decreased during treatment in all groups. However, only the AndroGel® groups decreased the mean LH concentrations enough to fall within the normal range. As with FSH, the primary hypogonadal men receiving AndroGel® showed dose-dependence in both the rate and extent of the LH response.
    TABLE 16(a)
    Concentrations for LH (mlU/mL) on Each of the
    Observation Days for Patients Having Primary
    Hypogonadism (Summary of Mean ± SD)
    N 5 g/day N 10 g/day N T-patch
    Day
    0 26 12.2 ± 12.1 33 13.9 ± 14.9  33 13.3 ± 14.3
    Day 30 23 5.6 ± 7.6 34 5.9 ± 8.1  31 10.9 ± 12.9
    Day 60 24 6.8 ± 9.0 32 4.8 ± 10.0 31 10.8 ± 11.8
    Day 90 24 5.9 ± 9.5 31 4.2 ± 11.0 30 10.0 ± 11.7
    Day 120 24  6.4 ± 11.9 28 3.8 ± 10.4 21 11.5 ± 11.5
    Day 150 22 4.4 ± 8.5 29 4.0 ± 11.3 21 7.4 ± 6.0
    Day 180 24 4.8 ± 6.8 28 4.0 ± 11.9 22 11.2 ± 10.5
  • The secondary hypogonadal men were less sensitive to exogenous testosterone. For the 44 patients identified as having secondary hypogonadism, the pretreatment mean concentrations were all within the lower limit normal range. The mean LH concentrations decreased during treatment with all three regimens as shown in FIG. 13(b) and Table 16(b).
    TABLE 16(b)
    Concentrations for LH (mlU/mL) on Each of the
    Observation Days for Patients Having Secondary
    Hypogonadism (Summary of Mean ± SD)
    N 5 g/day N 10 g/day N T-patch
    Day
    0 17 1.8 ± 2.6 12 1.4 ± 1.8 15 1.6 ± 3.1
    Day 30 16 1.1 ± 2.2 12 0.2 ± 0.2 14 0.4 ± 0.4
    Day 60 17 1.4 ± 3.8 12 0.2 ± 0.2 13 0.6 ± 0.5
    Day 90 15 1.2 ± 2.4 12 0.2 ± 0.2 14 0.7 ± 1.0
    Day 120 14 1.6 ± 4.0 12 0.2 ± 0.2 12 0.8 ± 0.8
    Day 150 14 1.6 ± 3.5 12 0.2 ± 0.2 11 1.2 ± 2.0
    Day 180 14 1.5 ± 3.7 12 0.2 ± 0.2 12 1.4 ± 2.1
  • None of the 25 patients suffering from age-associated hypogonadism had pretreatment LH concentrations outside of the normal range as shown in FIG. 13(c) and Table 16(c). The overall time and treatment effects were significant for the AndroGel® patients but not those patients using the testosterone patch.
    TABLE 16(c)
    Concentrations for LH (mlU/mL) on Each of the
    Observation Days for Patients Having Age-Related
    Hypogonadism (Summary of Mean ± SD)
    N 5 g/day N 10 g/day N T-patch
    Day
    0 13 3.2 ± 1.1 6 2.4 ± 1.8 6 2.9 ± 0.6
    Day 30 12 1.1 ± 1.0 6 0.1 ± 0.0 6 1.8 ± 1.1
    Day 60 12 0.8 ± 0.7 6 0.2 ± 0.3 5 3.4 ± 2.8
    Day 90 11 0.9 ± 1.2 6 0.1 ± 0.0 4 2.3 ± 1.4
    Day 120 11 1.0 ± 1.4 6 0.1 ± 0.0 4 2.2 ± 1.4
    Day 150 11 1.3 ± 1.5 5 0.1 ± 0.0 4 1.9 ± 1.2
    Day 180 11 1.8 ± 2.1 6 0.1 ± 0.0 4 1.4 ± 1.0
  • Of the 64 patients suffering from an unclassified hypogonadism, none of the patients had a pretreatment LH concentration above the upper limit. Fifteen percent, however, had pretreatment concentrations below the normal limit. The unclassified patients showed comparatively rapid LH concentration decreases in all treatment groups as shown in FIG. 13(d) and Table 16(d).
    TABLE 16(d)
    Concentrations for LH (mlU/mL) on Each of the Observation
    Days for Patients Having Unknown-Related Hypogonadism
    (Summary of Mean ± SD)
    N 5 g/day N 10 g/day N T-patch
    Day
    0 17 1.8 ± 1.2 26 2.5 ± 1.5 21 2.5 ± 1.5
    Day 30 17 0.3 ± 0.3 26 0.3 ± 0.3 21 1.3 ± 1.3
    Day 60 17 0.4 ± 0.5 26 0.3 ± 0.3 18 1.2 ± 1.4
    Day 90 17 0.5 ± 0.5 26 0.3 ± 0.4 18 1.0 ± 1.4
    Day 120 17 0.4 ± 0.4 26 0.4 ± 0.5 12 1.2 ± 1.1
    Day 150 17 0.8 ± 1.1 23 0.3 ± 0.4 13 1.1 ± 1.1
    Day 180 15 0.3 ± 0.4 25 0.4 ± 0.4 11 1.5 ± 1.3
  • (3) Summary: LH and FSH.
  • Patients receiving AndroGel® or the testosterone patch achieve “hormonal steady state” only after long-term treatment. Specifically, data involving FSH and LH show that these hormones do not achieve steady-state until many weeks after treatment. Because testosterone concentrations are negatively inhibited by FSH and LH, testosterone levels do not achieve true steady state until these other hormones also achieve steady state. However, because these hormones regulate only endogenous testosterone (which is small to begin with in hypogonadal men) in an intact feedback mechanism (which may not be present depending on the cause of hypogonadism), the level of FSH and/or LH may have little effect on the actual testosterone levels achieved. The net result is that the patients do not achieve a “hormonal steady state” for testosterone even though the Cavg, Cmin, and Cmax for testosterone remains relatively constant after a few days of treatment.
  • 2. Bone Mineral Density (“BMD”) and Similar Markers:
  • a. BMD.
  • BMD was assessed by dual energy X-ray absorptiometry (“DEXA”) using Hologic QDR 2000 or 4500 A (Hologic, Waltham, Mass.) on days 0 and 180 in the lumbar spine and left hip regions. BMD of spine was calculated as the average of BMD at L1 to L4. BMD of the left hip, which included Ward's triangle, was calculated by the average of BMD from neck, trochanter, and intertrochanter regions. The scans were centrally analyzed and processed at Hologic. BMD assessments were performed at 13 out of the 16 centers (206 out of 227 subjects) because of the lack of the specific DEXA equipment at certain sites.
  • Table 17 and FIGS. 14(a)-14(b) show that before treatment, the BMD of the hip or the spine was not different among the three treatment groups. Significant increases in BMD occurred only in subjects in the AndroGel® 10.0 g/day group and those who switched from AndroGel® 10.0 to 7.5 g/day groups. The increases in BMD were about 1% in the hip and 2% in the spine during the six-month period. Average increases in BMD of 0.6% and 1% in the hip and spine were seen in those receiving 5.0 g/day of AndroGel® but no increase was observed in the testosterone patch group.
    TABLE 17
    BMD Concentrations on Day 0 and Day 180 by
    Final Treatment Group Mean (± SD)
    Final % Change from
    Treatment Group N Day 0 N Day 180 N Day 0 to Day 180
    Hip
    5.0 g/day T-gel 50 1.026 ± 0.145 41 1.022 ± 0.145 41 0.7 ± 2.1
    5.0 to 7.5 g/day T-gel 16 1.007 ± 0.233 15 1.011 ± 0.226 15 1.0 ± 4.9
    10.0 to 7.5 g/day T-gel 20 1.002 ± 0.135 19 1.026 ± 0.131 19 1.3 ± 2.4
    10.0 g/day T-gel 53 0.991 ± 0.115 44 0.995 ± 0.130 44 1.1 ± 1.9
    T-Patch 67 0.982 ± 0.166 37 0.992 ± 0.149 37 −0.2 ± 2.9  
    Spine
    5.0 g/day T-gel 50 1.066 ± 0.203 41 1.072 ± 0.212 41 1.0 ± 2.9
    5.0 to 7.5 g/day T-gel 16 1.060 ± 0.229 15 1.077 ± 0.217 15 0.4 ± 5.5
    10.0 to 7.5 g/day T-gel 19 1.049 ± 0.175 19 1.067 ± 0.175 18 1.4 ± 3.2
    10.0 g/day T-gel 53 1.037 ± 0.126 44 1.044 ± 0.124 44 2.2 ± 3.1
    T-Patch 67 1.058 ± 0.199 36 1.064 ± 0.205 36 −0.2 ± 3.4  

    Note:

    Day 0 and Day 180 are arithmetic means, while percent change is a geometric mean.
  • The baseline hip and spine BMD and the change in BMD on day 180 were not significantly correlated with the average serum testosterone concentration on day 0. The changes in BMD in the hip or spine after testosterone replacement were not significantly different in subjects with hypogonadism due to primary, secondary, aging, or unclassified causes; nor were they different between naive and previously testosterone replaced subjects. The change in BMD in the spine was negatively correlated with baseline BMD values, indicating that the greatest increase in BMD occurred in subjects with the lowest initial BMD. The increase in BMD in the hip (but not in the spine) after testosterone treatment was correlated with the change in serum testosterone levels.
  • b. Bone Osteoblastic Activity Markers.
  • The results described above are supported by measurements of a number of serum and urine markers of bone formation. Specifically, the mean concentrations of the serum markers (PTH, SALP, osteocalcin, type I procollagen) generally increase during treatment in all treatment groups. In addition, the ratios of two urine markers of bone formation (N-telopeptide/creatinine ratio and calcium/creatinine ratio) suggests a decrease in bone resorption.
  • (1) PTH (Parathyroid or Calciotropic Hormone).
  • Serum intact PTH was measured by two site immunoradiometric assay (“IRMA”) kits from Nichol's Institute (San Juan Capistrano, Calif.). The LLC for the PTH assay was 12.5 ng/L. The intra- and inter-assay coefficients of variation were 6.9 and 9.6%, respectively. The UCLA-Harbor Medical Center has reported previously that the normal male adult range of PTH is 6.8 to 66.4 ng/L.
  • Table 18 provides the PTH concentrations over the 180-day study. FIG. 15 shows that the mean serum PTH levels were within the normal male range in all treatment groups at baseline. Statistically significant increases in serum PTH were observed in all subjects as a group at day 90 without inter-group differences. These increases in serum PTH were maintained at day 180 in all three groups.
    TABLE 18
    PTH Concentrations on Each of the Observation Days by Final Treatment Group (Mean ± SD)
    5 g/day 5 => 7.5 g/day 10 => 7.5 g/day 10 g/day
    N T-gel N T-gel N T-gel N T-gel N T-Patch
    Day
    0 53 16.31 ± 8.81  20 17.70 ± 9.66  20 18.02 ± 8.18 58 14.99 ± 6.11 75 15.60 ± 6.57 
    Day 30 49 17.91 ± 10.36 20 18.33 ± 8.02  20 17.45 ± 5.67 58 18.04 ± 8.95 72 18.33 ± 10.92
    Day 90 47 21.32 ± 11.47 20 21.25 ± 10.96 19 17.10 ± 6.04 54 20.01 ± 9.77 66 21.45 ± 13.71
    Day 120 46 21.19 ± 11.42 19 21.42 ± 13.20 20  19.62 ± 9..96 50  22.93 ± 12.57 46 21.07 ± 11.44
    Day 180 46 22.85 ± 12.89 19 21.34 ± 11.08 19  21.02 ± 10.66 51  25.57 ± 15.59 46 25.45 ± 16.54
  • (2) SALP.
  • SALP was quantitated by IRMA using reagents supplied by Hybritech (San Diego, Calif.). The LLQ for the SALP assay was 3.8 μg/L.; and the intra- and inter-assay precision coefficients were 2.9 and 6.5%, respectively. The UCLA-Harbor Medical Center reported that the adult normal male concentration of SALP ranges from 2.4 to 16.6 μg/L.
  • The pretreatment SALP concentrations were within the normal range. FIG. 16 and Table 19 show that SALP levels increased with testosterone treatment in the first 90 days and reached statistical difference in the testosterone patch group. Thereafter serum SALP plateaued in all treatment groups.
    TABLE 19
    SALP Concentrations on Each of the Observation Days by Final Treatment Group (Mean ± SD)
    5 g/day 5 => 7.5 g/day 10 => 7.5 10 g/day
    N T-gel N T-gel N g/day T-gel N T-gel N T-Patch
    Day
    0 53  9.96 ± 5.61 20 12.36 ± 4.62 20 10.48 ± 3.68 58 9.80 ± 3.57 76 10.44 ± 3.77
    Day 30 49 10.20 ± 6.77 20 11.38 ± 4.09 20 11.83 ± 4.32 58 9.93 ± 3.88 71 10.86 ± 3.75
    Day 90 47 11.64 ± 7.98 20 11.97 ± 5.03 20 10.97 ± 3.18 55 9.56 ± 3.12 65 11.99 ± 9.36
    Day 120 46 11.71 ± 7.85 19 12.12 ± 5.25 20 11.61 ± 2.58 48 9.63 ± 3.58 45 11.63 ± 4.72
    Day 180 45 11.12 ± 7.58 19 11.67 ± 5.35 19 11.22 ± 3.44 51 9.19 ± 2.42 46 11.47 ± 3.77
  • (3) Osteocalcin.
  • Serum osteocalcin was measured by an IRMA from Immutopics (San Clemente, Calif.). The LLQ was 0.45 μg/L. The intra- and inter-assay coefficients were 5.6 and 4.4%, respectively. The UCLA-Harbor Medical Center reports that the normal male adult range for the osteocalcin assay ranges from 2.9 to 12.7 μg/L.
  • As shown in FIG. 17 and Table 20, the baseline mean serum osteocalcin levels were within the normal range in all treatment groups. During the first 90-day treatment, mean serum osteocalcin increased with testosterone replacement in all subjects as a group without significant differences between the groups. With continued treatment serum osteocalcin either plateaued or showed a decrease by day 180.
    TABLE 20
    Osteocalcin Concentrations on Each of the Observation Days by Final Treatment Group (Mean ± SD)
    5 g/day 5 => 7.5 g/day 10 => 7.5 g/day 10 g/day
    N T-gel N T-gel N T-gel N T-gel N T-Patch
    Day
    0 53 4.62 ± 1.55 20 5.01 ± 2.03 20 4.30 ± 1.28 58 4.58 ± 1.92 76 4.53 ± 1.54
    Day 30 49 4.63 ± 1.65 20 5.35 ± 2.06 20 4.48 ± 1.72 58 4.91 ± 2.08 72 5.17 ± 1.61
    Day 90 47 4.91 ± 2.15 20 5.29 ± 1.87 19 4.76 ± 1.50 55 4.83 ± 2.13 66 5.18 ± 1.53
    Day 120 46 4.95 ± 1.97 18 4.97 ± 1.60 20 4.71 ± 1.39 49 4.61 ± 2.01 47 4.98 ± 1.87
    Day 180 45 4.79 ± 1.82 19 4.89 ± 1.54 19 4.47 ± 1.49 51 3.76 ± 1.60 46 5.15 ± 2.18
  • (4) Type I Procollagen.
  • Serum type I procollagen was measured using a RIA kit from Incstar Corp (Stillwater, Minn.). The LLQ of the procollagen assay was 5 μg/L, and the intra- and inter-assay precisions were 6.6 and 3.6%, respectively. The UCLA-Harbor Medical Center reports that the normal adult male concentration of type I procollagen ranges from 56 to 310 μg/L.
  • FIG. 18 and Table 21 show that serum procollagen generally followed the same pattern as serum osteocalcin. At baseline the mean levels were similar and within the normal range in all treatment groups. With transdermal treatment, serum procollagen increased significantly in all subjects as a group without treatment group differences. The increase in procollagen was highest on day 30 and then plateaued until day 120. By day 180, the serum procollagen levels returned to baseline levels.
    TABLE 21
    Procollagen Concentrations on Each of the Observation Days by Final Treatment Group (Mean ± SD)
    5 g/day 5 => 7.5 g/day 10 => 75 g/day 10 g/day
    N T-gel N T-gel N T-gel N T-gel N T-Patch
    Day
    0 53 115.94 ± 43.68 20 109.27 ± 32.70 20 120.93 ± 28.16 58 125.33 ± 57.57 76 122.08 ± 51.74
    Day 30 49 141.09 ± 64.02 20 141.41 ± 77.35 20 147.25 ± 49.85 58 149.37 ± 60.61 71 139.26 ± 59.12
    Day 90 47 137.68 ± 68.51 20 129.02 ± 60.20 29 144.60 ± 58.20 55 135.59 ± 51.54 66 130.87 ± 49.91
    Day 120 46 140.07 ± 81.48 19 133.61 ± 54.09 20 139.00 ± 64.96 50 128.48 ± 45.56 46 130.39 ± 42.22
    Day 180 45 119.78 ± 49.02 19 108.78 ± 35.29 19 123.51 ± 39.30 51 108.52 ± 38.98 45 120.74 ± 56.10
  • c. Urine Bone Turnover Markers: N-telopeptide/Cr and Ca/Cr Ratios.
  • Urine calcium and creatinine were estimated using standard clinical chemistry procedures by an autoanalyzer operated by the UCLA-Harbor Pathology Laboratory. The procedures were performed using the COBAS MIRA automated chemistry analyzer system manufactured by Roche Diagnostics Systems. The sensitivity of the assay for creatinine was 8.9 mg/dL and the LLQ was 8.9 mg/dL. According to the UCLA-Harbor Medical Center, creatinine levels in normal adult men range from 2.1 mM to 45.1 mM. The sensitivity of the assay for calcium was 0.7 mg/dL and the LLQ was 0.7 mg/dL. The normal range for urine calcium is 0.21 mM to 7.91N-telopeptides were measured by an enzyme-linked immunosorbant assay (“ELISA”) from Ostex (Seattle, Wash.). The LLQ for the N-telopeptide assay was 5 nM bone collagen equivalent (“BCE”). The intra- and inter-assay had a precision of 4.6 and 8.9%, respectively. The normal range for the N-telopeptide assay was 48-2529 nM BCE. Samples containing low or high serum/urine bone marker levels were reassayed after adjusting sample volume or dilution to ensure all samples would be assayed within acceptable precision and accuracy.
  • The normal adult male range for the N-telopeptide/Cr ratio is 13 to 119 nM BCE/nM Cr. As shown in FIG. 19 and Table 22, urinary N-telopeptide/Cr ratios were similar in all three treatment groups at baseline but decreased significantly in the AndroGel® 10.0 g/day group but not in the AndroGel® 5.0 g/day or testosterone patch group during the first 90 days of treatment. The decrease was maintained such that urinary N-telopeptide/Cr ratio remained lower than baseline in AndroGel® 10.0 g/day and in those subjects adjusted to 7.5 g/day from 10.0 g/day group at day 180. This ratio also decreased in the testosterone patch treatment group by day 180.
    TABLE 22
    N-Telopeptide/Cr Ratio on Each of the Observation
    Days by Initial Treatment Group (Mean ± SD)
    Initial Treatment 5.0 g. day 10.0 g/day Across-group
    Group N T-gel N T-gel N T-Patch p-value
    Day
    0 71  90.3 ± 170.3 75  98.0 ± 128.2 75 78.5 ± 82.5  0.6986
    Day 30 65 74.6 ± 79.3 73 58.4 ± 66.4 66 91.6 ± 183.6 0.3273
    Day 90 62 70.4 ± 92.6 73 55.2 ± 49.1 63 75.0 ± 113.5 0.5348
    Day 120 35 78.8 ± 88.2 36 46.6 ± 36.4 21 71.2 ± 108.8 0.2866
    Day 180 64 68.2 ± 81.1 70 46.9 ± 43.1 47 49.4 ± 40.8  0.2285
  • The normal range for Ca/Cr ratio is 0.022 to 0.745 mM/mM. FIG. 20 shows no significant difference in baseline urinary Ca/Cr ratios in the three groups. With transdermal testosterone replacement therapy, urinary Ca/Cr ratios did not show a significant decrease in any treatment group at day 90. With continued testosterone replacement to day 180, urinary Ca/Cr showed marked variation without significant changes in any treatment groups.
    TABLE 23
    Ca/Cr Ratio on Each of the Observation Days by Initial Treatment Group (Mean ± SD)
    Initial Treatment 5.0 g. day 10.0 g/day Across-group
    Group N T-gel N T-gel N T-Patch p-value
    Day
    0 71 0.150 ± 0.113 75 0.174 ± 0.222 75 0.158 ± 0.137 0.6925
    Day 30 65 0.153 ± 0.182 73 0.128 ± 0.104 66 0.152 ± 0.098 0.3384
    Day 90 63 0.136 ± 0.122 73 0.113 ± 0.075 63 0.146 ± 0.099 0.2531
    Day 120 36 0.108 ± 0.073 36 0.117 ± 0.090 21 0.220 ± 0.194 0.0518
    Day 180 64 0.114 ± 0.088 70 0.144 ± 0.113 47 0.173 ± 0.108 0.0398
  • Interestingly, the change in Ca/Cr ratio from baseline at day 90 was inversely related to the baseline Ca/Cr ratios. Similarly, the change in urine N-telopeptide/Cr ratio was also inversely proportional to the baseline N-telopeptide/Cr ratio (r=−0.80, p=0.0001). Thus subjects with the highest bone resorption markers at baseline showed the largest decreases of these markers during transdermal testosterone replacement. The decreases in urinary bone resorption markers were most prominent in subjects who had highest baseline values, suggesting that hypogonadal subjects with the most severe metabolic bone disease responded most to testosterone replacement therapy.
  • d. Serum Calcium.
  • Serum calcium showed no significant inter-group differences at baseline, nor significant changes after testosterone replacement. Serum calcium levels showed insignificant changes during testosterone replacement.
  • 3. Libido, Sexual Performance, and Mood.
  • Sexual function and mood were assessed by questionnaires the patients answered daily for seven consecutive days before clinic visits on day 0 and on days 30, 60, 90, 120, 150, and 180 days during gel and patch application. The subjects recorded whether they had sexual day dreams, anticipation of sex, flirting, sexual interaction (e.g., sexual motivation parameters) and orgasm, erection, masturbation, ejaculation, intercourse (e.g., sexual performance parameters) on each of the seven days. The value was recorded as 0 (none) or 1 (any) for analyses and the number of days the subjects noted a parameter was summed for the seven-day period. The average of the four sexual motivation parameters was taken as the sexual motivation score and that of the five sexual motivation parameters as the sexual motivation mean score (0 to 7). The subjects also assessed their level of sexual desire, sexual enjoyment, and satisfaction of erection using a seven-point Likert-type scale (0 to 7) and the percent of full erection from 0 to 100%. The subjects rated their mood using a 0 to 7 score. The parameters assessed included positive mood responses: alert, friendly, full of energy, well/good feelings and negative mood responses: angry, irritable, sad, tired, nervous. Weekly average scores were calculated. The details of this questionnaire had been described previously and are fully incorporated by reference. See Wang et al., “Testosterone Replacement Therapy Improves Mood in Hypogonadal Men A Clinical Research Center Study,” 81 J. Clinical Endocrinology & Metabolism 3578-3583 (1996).
  • a. Libido.
  • As shown in FIG. 21(a), at baseline, sexual motivation was the same in all treatment groups. After transdermal testosterone treatment, overall sexual motivation showed significant improvement. The change in the summary score from baseline, however, was not different among the three treatment groups.
  • Libido was assessed from responses on a linear scale of: (1) overall sexual desire, (2) of sexual activity without a partner, and (3) enjoyment of sexual activity with a partner. As shown in FIG. 21(b) and Table 24, as a group, overall sexual desire increased after transdermal testosterone treatment without inter-group difference. Sexual enjoyment with and without a partner (FIG. 21(c) and Tables 25 and 26) also increased as a group.
  • Similarly the sexual performance score improved significantly in all subjects as a group. The improvement in sexual performance from baseline values was not different between transdermal preparations.
    TABLE 24
    Overall Sexual Desire Changes From Day 0 to Day
    180 by Initial Treatment Group (Mean ± SD)
    Initial Treatment Change From Within-Group
    Group N Day 0 N Day 180 N Day 0 to Day 180 p-value
    5.0 g/day T-gel 69 2.1 ± 1.6 63 3.5 ± 1.6 60 1.4 ± 1.9 0.0001
    10.0 g/day T-gel 77 2.0 ± 1.4 68 3.6 ± 1.6 67 1.5 ± 1.9 0.0001
    T-Patch 72 2.0 ± 1.6 47 3.1 ± 1.9 45 1.6 ± 2.1 0.0001
    Across-Groups 0.8955 0.2247 0.8579
    p-value
  • TABLE 25
    Level of Sexual Enjoyment Without a Partner Changes From Day
    0 to Day 180 by Initial Treatment Group (Mean ± SD)
    Initial Treatment Change From Within-Group
    Group N Day 0 N Day 180 N Day 0 to Day 180 p-value
    5.0 g/day T-gel 60 1.5 ± 1.9 51 1.9 ± 1.9 44 0.8 ± 1.4 0.0051
    10.0 g/day T-gel 63 1.2 ± 1.4 53 2.2 ± 1.9 48 1.1 ± 1.6 0.0001
    T-Patch 66 1.4 ± 1.8 44 2.2 ± 2.3 40 1.0 ± 1.9 0.0026
    Across-Groups 0.6506 0.7461 0.6126
    p-value
  • TABLE 26
    Level of Sexual Enjoyment With a Partner Change from Day
    0 to Day 180 by Initial Treatment Group (Mean ± SD)
    Initial Treatment Change From Within-Group
    Group N Day 0 N Day 180 N Day 0 to Day 180 p-value
    5.0 g/day T-gel 64 2.1 ± 2.1 55 2.6 ± 2.2 48 0.4 ± 2.2 0.0148
    10.0 g/day T-gel 66 1.8 ± 1.7 58 3.0 ± 2.2 52 1.0 ± 2.3 0.0053
    T-Patch 61 1.5 ± 1.7 40 2.2 ± 2.4 35 0.7 ± 2.3 0.1170
    Across-Groups 0.2914 0.1738 0.3911
    p-value
  • b. Sexual Performance.
  • FIG. 22(a) shows that while all treatment groups had the same baseline sexual performance rating, the rating improved with transdermal testosterone treatment in all groups. In addition, as a group, the subjects' self-assessment of satisfaction of erection (FIG. 22(b) and Table 27) and percent full erection (FIG. 22(c) and Table 28) were also increased with testosterone replacement without significant differences between groups.
  • The improvement in sexual function was not related to the dose or the delivery method of testosterone. Nor was the improvement related to the serum testosterone levels achieved by the various testosterone preparations. The data suggest that once a threshold (serum testosterone level probably at the low normal range) is achieved, normalization of sexual function occurs. Increasing serum testosterone levels higher to the upper normal range does not further improve sexual motivation or performance.
    TABLE 27
    Satisfaction with Duration of Erection Change from Day 0
    to Day 180 by Initial Treatment Group (Mean ± SD)
    Initial Treatment Change From Within-Group
    Group N Day 0 N Day 180 N Day 0 to Day 180 p-value
    5.0 g/day T-gel 55 2.5 ± 2.1 57 4.3 ± 1.8 44 1.9 ± 2.0 0.0001
    10/0 g/day T-gel 64 2.9 ± 1.9 58 4.5 ± 1.7 53 1.5 ± 2.0 0.0001
    T-Patch 45 3.4 ± 2.1 34 4.5 ± 2.0 20 1.3 ± 2.1 0.0524
    Across-Groups 0.1117 0.7093 0.5090
    p-value
  • TABLE 28
    Percentage of Full Erection Change from Day 0 to Day
    180 by Initial Treatment Group (Mean ± SD)
    Initial Treatment Change From Within-Group
    Group N Day 0 N Day 180 N Day 0 to Day 180 p-value
    5.0 g/day T-gel 53 53.1 ± 24.1 57 67.4 ± 22.5 43 18.7 ± 22.1 0.0001
    10.0 g/day T-gel 62 59.6 ± 22.1 59 72.0 ± 20.2 52 10.4 ± 23.4 0.0001
    T-Patch 47 56.5 ± 24.7 33 66.7 ± 26.7 19 12.7 ± 20.3 0.0064
    Across-Groups 0.3360 0.4360 0.1947
    p-value
  • c. Mood.
  • The positive and negative mood summary responses to testosterone replacement therapy are shown in FIGS. 23(a) and 23(b). All three treatment groups had similar scores at baseline and all subjects as a group showed improvement in positive mood. Similarly, the negative mood summary scores were similar in the three groups at baseline and as a group the responses to transdermal testosterone applications showed significant decreases without showing between group differences. Specifically, positive mood parameters, such as sense of well being and energy level, improved and negative mood parameters, such as sadness and irritability, decreased. The improvement in mood was observed at day 30 and was maintained with continued treatment. The improvement in mood parameters was not dependent on the magnitude of increase in the serum testosterone levels. Once the serum testosterone increased into the low normal range, maximal improvement in mood parameters occurred. Thus, the responsiveness in sexual function and mood in hypogonadal men in response to testosterone therapy appeared to be dependent on reaching a threshold of serum testosterone at the low normal range.
  • 4. Muscle Strength.
  • Muscle strength was assessed on days 0, 90, and 180. The one-repetitive maximum (“1-RM”) technique was used to measure muscle mass in bench press and seated leg press exercises. The muscle groups tested included those in the hips, legs, shoulders, arms, and chest. The 1-RM technique assesses the maximal force generating capacity of the muscles used to perform the test. After a 5-10 minute walking and stretching period, the test began with a weight believed likely to represent the patient's maximum strength. The test was repeated using increments of about 2-10 pounds until the patient was unable to lift additional weight with acceptable form. Muscle strength was assessed in 167 out of the 227 patients. Four out of 16 centers did not participate in the muscle strength testing because of lack of the required equipment.
  • The responses of muscle strength testing by the arm/chest and leg press tests are shown in FIG. 24(a) and 24(b) and Table 29. There were no statistical significant differences in arm/chest or leg muscle strength among the three groups at baseline. In general, muscle strength improved in both the arms and legs in all three treatment groups without inter-group differences at both day 90 and 180. The results showed an improvement in muscle strength at 90 and 180 days, more in the legs than the arms, which was not different across treatment groups nor on the different days of assessment. Adjustment of the dose at day 90 did not significantly affect the muscle strength responses to transdermal testosterone preparations.
    TABLE 29
    Muscle Strength - Days 0, 90, and 180 Levels and Change (lbs.) from
    Day 0 to Day 90 and from Day 0 to Day 180 by Final Treatment Group
    Arm/Chest (Bench
    Final Treatment Seated Leg Press Press)
    Group Study Day N Mean ± SD (lbs.) N Mean ± SD (lbs.)
    5.0 g/day T-gel 0 37 356.8 ± 170.0 37 100.5 ± 37.4
    90 30 396.4 ± 194.3 31 101.2 ± 30.7
    Δ 0-90  30 25.8 ± 49.2 31  4.0 ± 10.0
    180 31 393.4 ± 196.6 31  99.7 ± 31.4
    Δ 0-180 31 19.9 ± 62.4 31  1.3 ± 13.0
    7.5 g/day T-gel 0 16 302.8 ± 206.5 16 102.8 ± 48.9
    (from 5.0 g/day) 90 15 299.8 ± 193.9 15 109.5 ± 47.6
    Δ 0-90  15 17.0 ± 88.4 15  5.0 ± 21.3
    180 14 300.6 ± 203.0 14 108.5 ± 49.3
    Δ 0-180 14 −0.1 ± 110.2 14  5.6 ± 30.4
    7.5 g/day T-gel 0 14 363.4 ± 173.8 14 123.3 ± 54.7
    (From 10.0 g/day) 90 14 401.6 ± 176.6 14 134.6 ± 57.5
    Δ 0-90  14 38.2 ± 42.9 14  11.3 ± 10.5
    180 12 409.9 ± 180.2 14 132.3 ± 61.5
    Δ 0-180 12 33.9 ± 67.3 14  9.0 ± 18.7
    10.0 g/day T-gel 0 45 345.9 ± 186.9 43 114.7 ± 55.1
    90 43 373.5 ± 194.8 41 119.8 ± 54.2
    Δ 0-90  43 27.6 ± 45.1 41  4.6 ± 12.8
    180 36 364.4 ± 189.1 34 112.0 ± 45.5
    Δ 0-180 36 32.2 ± 72.3 34  1.9 ± 14.8
    T-Patch 0 55 310.4 ± 169.7 54  99.2 ± 43.1
    90 46 344.9 ± 183.9 46 106.2 ± 44.0
    Δ 0-90  46 25.4 ± 37.0 46  3.2 ± 12.0
    180 36 324.8 ± 199.0 35 104.8 ± 44.8
    Δ 0-180 36 15.2 ± 54.7 35  2.3 ± 15.7
  • 5. Body Composition.
  • Body composition was measured by DEXA with Hologic 2000 or 4500A series on days 0, 90, and 180. These assessments were done in 168 out of 227 subjects because the Hologic DEXA equipment was not available at 3 out of 16 study centers. All body composition measurements were centrally analyzed and processed by Hologic (Waltham, Mass.).
  • At baseline, there were no significant differences in total body mass (“TBM”), total body lean mass (“TLN”), percent fat (“PFT”), and total body fat mass (“TFT”) in the three treatment groups. As shown in FIGS. 25(a) and Table 30, all treatment groups incurred an overall increase in TBM. The increase in TBM was mainly due to the increases in TLN. FIG. 25(b) and Table 30 show that after 90 days of testosterone replacement the increase in TLN was significantly higher in the 10.0 g/day AndroGel® group than in the other two groups. At day 180, the increases in TLN were further enhanced or maintained in all AndroGel® treated groups, as well as in the testosterone patch group.
  • FIGS. 25(c) and (d) show that the TFT and the PFT decreased in all transdermal AndroGel® treatment groups. At 90 days of treatment, TFT was significantly reduced in the 5.0 g/day and 10.0 g/day AndroGel® groups, but was not changed in the testosterone patch group. This decrease was maintained at day 180. Correspondingly, at day 90 and 180, the decrease in PFT remained significantly lower in all AndroGel® treated groups but not significantly reduced in the testosterone patch group.
  • The increase in TLN and the decrease in TFT associated with testosterone replacement therapy showed significant correlations with the serum testosterone level attained by the testosterone patch and the different doses of AndroGel®. Testosterone gel administered at 10.0 increased lean mass more than the testosterone patch and the 5.0 g/day AndroGel® groups. The changes were apparent on day 90 after treatment and were maintained or enhanced at day 180. Such changes in body composition was significant even though the subjects were withdrawn from prior testosterone therapy for six weeks. The decrease in TFT and PFT was also related to the serum testosterone achieved and were different across the treatment groups. The testosterone patch group did not show a decrease in PFT or TFT after 180 days of treatment. Treatment with AndroGel® (5.0 to 10.0 g/day) for 90 days reduced PFT and TFT. This decrease was maintained in the 5.0 and 7.5 g/day groups at 180 days but were further lowered with continued treatment with the higher dose of the AndroGel®.
    TABLE 30
    Mean Change in Body Composition Parameters
    (DEXA) From Baseline to Day 90 and Baseline
    to Day 180 By Final Treatment Groups
    Final
    Treatment Mean Change from Day 0-Day 90
    Group N TFT (g) TLN (g) TBM (g) PFT
    5.0 g/day 43 −782 ± 2105 1218 ± 2114  447 ± 1971 −1.0 ± 2.2
    T-gel
    7.5 g/day 12 −1342 ± 3212  1562 ± 3321  241 ± 3545 −1.0 ± 3.1
    (from 5.0
    g/day)
    7.5 g/day 16 −1183 ± 1323  3359 ± 2425 2176 ± 2213 −2.0 ± 1.5
    (from 10.0
    g/day)
    10.0 g/day 45 −999 ± 1849 2517 ± 2042 1519 ± 2320 −1.7 ± 1.8
    T-gel
    T-Patch 52   11 ± 1769  1205 ± 1913 1222 ± 2290 −0.4 ± 1.6
    Final
    Treatment Mean Change from Day 0-Day 180
    Group N TFT (g) TLN (g) TBM (g) PFT
    5.0 g/day 38  −972 ± 3191 1670 ± 2469  725 ± 2357 −1.3 ± 3.1
    T-gel
    7.5 g/day 13 −1467 ± 3851 2761 ± 3513 1303 ± 3202 −1.5 ± 3.9
    (from 5.0
    g/day)
    7.5 g/day 16 −1333 ± 1954 3503 ± 1726 2167 ± 1997 −2.2 ± 1.7
    (from 10.0
    g/day)
    10.0 g/day 42 −2293 ± 2509 3048 ± 2284  771 ± 3141 −2.9 ± 2.1
    T-gel
    T-Patch 34   293 ± 2695   997 ± 2224 1294 ± 2764 −0.3 ± 2.2
  • 6. Lipid profile and blood chemistry.
  • The serum total, HDL, and LDL cholesterol levels at baseline were not significantly different in all treatment groups. With transdermal testosterone replacement, there were no overall treatment effects nor inter-group differences in serum concentrations of total, HDL- and LDL-cholesterol (FIG. 5(d)) and triglycerides (data not shown). There was a significant change of serum total cholesterol concentrations as a group with time (p=0.0001), the concentrations on day 30, 90, and 180 were significantly lower than day 0.
  • Approximately 70 to 95% of the subjects had no significant change in their serum lipid profile during testosterone replacement therapy. Total cholesterol levels which were initially high were lowered into the normal range (of each center's laboratory) at day 180 in 17.2, 20.4, and 12.2% of subjects on testosterone patch, AndroGel® 5.0 g/day and AndroGel® 10.0 g/day, respectively. Serum HDL-cholesterol levels (initially normal) were reduced to below the normal range (of each center's laboratory) in 9.8, 4.0, 9.1, and 12.5% of subjects at day 180 in the testosterone patch, AndroGel® 5.0, 7.5, and 10.0 g/day groups, respectively. There was no clinically significant changes in renal or liver function tests in any treatment group.
  • 7. Skin Irritations.
  • Skin irritation assessments were performed at every clinic visit using the following scale: 0=no erythema; 1=minimal erythema; 2=moderate erythema with sharply defined borders; 3=intense erythema with edema; and 4=intense erythema with edema and blistering/erosion.
  • Tolerability of the daily application of AndroGel® at the tested dosages was much better than with the permeation-enhanced testosterone patch. Minimal skin irritation (erythema) at the application site was noted in three patients in the AndroGel® 5.0 g/day group (5.7%) and another three in the AndroGele 10.0 g/day group (5.3%). Skin irritation varying in intensity from minimal to severe (mild erythema to intense edema with blisters) occurred in 65.8% of patients in the patch group. Because of the skin irritation with the testosterone patch, 16 subjects discontinued the study; 14 of these had moderate to severe skin reactions at the medication sites. No patients who received AndroGel® discontinued the study because of adverse skin reactions. The open system and the lower concentration of alcohol in the AndroGel® formulation markedly reduced skin irritation resulting in better tolerability and continuation rate on testosterone replacement therapy.
  • Moreover, based on the difference in the weight of the dispensed and returned AndroGel® bottles, the mean compliance was 93.1% and 96.0% for the 5.0 g/day and 10.0 g/day AndroGel® groups during days 1-90, respectively. Compliance remained at over 93% for the three AndroGel® groups from days 91-180. In contrast, based on counting the patches returned by the subjects, the testosterone patch compliance was 65% during days 1-90 and 74% during days 91-180. The lower compliance in the testosterone patch group was mostly due to skin reactions from the subjects' records.
    TABLE 31
    Incidence of Skin-Associated Adverse Events: Day 1 to
    Day 180 in Patients Who Remained on Initial Treatment
    5.0 g/day T-gel 10.0 g/day T-gel T-Patch
    N = 53 N = 57 N = 73
    Total 16 (30.2%) 18 (31.6%) 50 (68.5%)
    Application 3 (5.7%) 3 (5.3%) 48 (65.8%)
    Site Reaction
    Acne 1 (1.9%) 7 (12.3%) 3 (4.1%)
    Rash 4 (7.5%) 4 (7.0%) 2 (2.7%)
    Skin Disorder 2 (3.8%) 1 (1.8%) 1 (1.4%)
    Skin Dry 2 (3.8) 0 (0.0%) 1 (1.4%)
    Sweat 0 (0.0%) 2 (3.5%) 0 (0.0%)
    Reaction 2 (3.6%) 1 (1.7%) 0 (0.0%)
    Unevaluable
    Cyst 0 (0.0%) 0 (0.0%) 2 (2.7%)
  • EXAMPLE 2 Gel Delivery Dosage Forms and Devices
  • The present invention is also directed to a method for dispensing and packaging the gel. In one embodiment, the invention comprises a hand-held pump capable of delivering about 2.5 g of testosterone gel with each actuation. In another embodiment, the gel is packaged in foil packets comprising a polyethylene liner. Each packet holds about 2.5 g of testosterone gel. The patient simply tears the packet along a perforated edge to remove the gel. However, because isopropyl myristate binds to the polyethylene liner, additional isopropyl myristate is added to the gel in order to obtain a pharmaceutically effective gel when using this delivery embodiment. Specifically, when dispensing the gel via the foil packet, about 41% more isopropyl myristate is used in the gel composition (i.e., about 0.705 g instead of about 0.5 g in Table 5), to compensate for this phenomenon.
  • The composition can also be dispensed from a rigid multi-dose container (e.g., with a hand pump) having a larger foil packet of the composition inside the container. Such larger packets also comprise a polyethylene liner as above.
  • Both embodiments permit a patient to deliver accurate but incremental amounts of gel (e.g., either 2.5 g, 5.0 g, 7.5 g, etc.) to the body. These delivery mechanisms thus permit the gel to be administered in unit dose form depending on the particular needs and characteristics of the patient.
  • Although the invention has been described with respect to specific embodiments and examples, it should be appreciated that other embodiments utilizing the concept of the present invention are possible without departing from the scope of the invention. The present invention is defined by the claimed elements, and any and all modifications, variations, or equivalents that fall within the true spirit and scope of the underlying principles.

Claims (73)

1. A hydroalcoholic gel pharmaceutical composition, comprising:
(a) about 0.1% to about 10% testosterone weight to weight of the composition;
(b) an alcohol;
(c) a penetration enhancer; and
(d) a gelling agent;
wherein upon administration of the pharmaceutical composition to skin of a male subject, the testosterone is absorbed into bloodstream of the subject at a rate and duration that maintains a circulating serum concentration of the testosterone greater than about 400 ng testosterone per dl serum during a time period beginning about 2 hours after administration and ending about 24 hours after administration.
2. The pharmaceutical composition of claim 1, wherein the serum testosterone concentration is maintained between about 400 ng testosterone per dl serum to about 105 ng testosterone per dl serum.
3. The pharmaceutical composition of claim 1, wherein for each about 0.1 gram per day administration of the composition to the skin, an increase of at least about 5 ng/dl in serum testosterone concentration results in the subject.
4. The pharmaceutical composition of claim 1, wherein the composition is a dosage form of approximately 0.1 g, 2.5 g, 5 g, 7.5 g, or 10 g.
5. The pharmaceutical composition of claim 4, wherein the dose is approximately a 5 g dose delivering about 50 mg to about 100 mg of testosterone to the skin.
6. The pharmaceutical composition of claim 4, wherein the dose is approximately 7.5 g dose delivering about 50 mg to about 100 mg of testosterone to the skin.
7. The pharmaceutical composition of claim 4, wherein the dose is approximately a 10 g dose delivering 50 mg to about 100 mg of testosterone to the skin.
8. The pharmaceutical composition of claim 1, wherein maximum serum testosterone concentration in the subject is reached about 16 hours after administration of the composition on day one of administration.
9. The pharmaceutical composition of claim 1, wherein the subject is a male human.
10. The pharmaceutical composition of claim 9, wherein the serum testosterone concentration in the subject is at least 490 ng/dl to about 860 ng/dl after at least about 30 days of daily administration.
11. The pharmaceutical composition of claim 9, wherein upon administration of the composition to the skin of the subject after at least about 30 days of daily administration, the testosterone absorbs into the bloodstream of the subject at a rate and duration that maintains a serum dihydrotestosterone concentration in the subject greater than about 54 ng/dl.
12. The pharmaceutical composition of claim 11, wherein a ratio of serum dihydrotestosterone concentration to serum testosterone concentration of greater than about 0.23 is achieved in the subject after at least about 30 days of daily administration.
13. The pharmaceutical composition of claim 9, wherein upon administration of the pharmaceutical composition to the skin of the subject after at least about 30 days of daily administration, the testosterone absorbs into the bloodstream of the subject at a rate and duration that maintains a total serum androgen concentration in the subject greater than about 372 ng/dl.
14. The pharmaceutical composition of claim 9, wherein upon administration of the composition to the skin of the subject after at least about 30 days of daily administration, the testosterone absorbs into the bloodstream of the subject at a rate and duration that maintains a serum estradiol concentration in the subject greater than about 28 pg/ml.
15. The pharmaceutical composition of claim 9, wherein upon administration of the composition to the skin of the subject after at least about 30 days of daily administration, the testosterone absorbs into the bloodstream of the subject at a rate and duration that maintains a serum follicle stimulating hormone concentration in the subject less than about 11 mIU/ml.
16. The pharmaceutical composition of claim 15, wherein the serum follicle stimulating hormone concentration in the subject is less than about 3.7 mIU/ml after at least about 30 days of daily administration.
17. The pharmaceutical composition of claim 15, wherein the serum follicle stimulating hormone concentration is within or below the normal range after at least about 30 days of daily administration.
18. The pharmaceutical composition of claim 9, wherein upon administration of the composition to the skin of the subject after at least about 30 days of daily administration, the testosterone absorbs into the bloodstream of the subject at a rate and duration that maintains a serum luteinizing hormone concentration within normal range of an adult male.
19. The pharmaceutical composition of claim 18, wherein the normal range is less than about 6.8 mIU/ml serum luteinizing hormone after at least about 30 days of daily administration.
20. The pharmaceutical composition of claim 9, wherein upon administration of the pharmaceutical composition to the skin of the subject after at least about 30 days of daily administration, the testosterone has an accumulation ratio in the subject greater than about 1.5.
21. The pharmaceutical composition of claim 9, wherein upon administration of the pharmaceutical composition to the skin of the subject after at least about 30 days of daily administration the testosterone has a net AUC0-24 in the subject greater than 220 nmol*h/l.
22. The pharmaceutical composition of claim 1, wherein the testosterone comprises an enantiomer, a racemic mixture, a derivative, a base, or a salt thereof.
23. The pharmaceutical composition of claim 1, wherein the composition is a dosage form containing about 1.0 gram to about 10 grams of the composition.
24. The pharmaceutical composition of claim 1, wherein the composition is a dosage form containing about 2.5 grams to about 7.5 grams of the composition.
25. The pharmaceutical composition of claim 1, wherein the composition is a dosage form containing about 2.5 grams to about 5.0 grams of the composition.
26. The pharmaceutical composition of claim 1, wherein the composition comprises about 0.5% to about 5% (w/w) testosterone.
27. The pharmaceutical composition of claim 1, wherein the composition comprises about 1% (w/w) testosterone.
28. The pharmaceutical composition of claim 1, wherein the permeation enhancer comprises isopropyl myristate in an amount of about 0.25% to about 2.5% (w/w) of the composition.
29. The pharmaceutical composition of claim 1, wherein the composition comprises isopropyl myristate in an amount of about 0.5% (w/w) of the composition.
30. The pharmaceutical composition of claim 1, wherein the gelling agent is polyacrylic acid.
31. The pharmaceutical composition of claim 30, wherein the composition comprises about 0.9% (w/w) polyacrylic acid.
32. The pharmaceutical composition of claim 1, wherein the composition comprises about 40% to about 90% (w/w) alcohol.
33. A method of treating hypogonadism in a male subject in need thereof, comprising administering a pharmaceutical composition to skin of the subject in a pharmacologically effective amount to treat the hypogonadism, wherein the composition comprises:
(a) about 0.1% to about 10% testosterone weight to weight of the composition;
(b) an alcohol;
(c) a penetration enhancer; and
(d) a gelling agent;
wherein the testosterone is absorbed into bloodstream of the subject at a rate and duration that maintains a circulating serum concentration of the testosterone greater than about 400 ng testosterone per dl serum during a time period beginning about 2 hours after administration and ending about 24 hours after administration.
34. The method of claim 33, wherein the composition is administered daily for at least about 7 days.
35. The method of claim 33, wherein the composition is administered daily for at least 30 days.
36. The method of claim 33, wherein the composition is administered daily for at least about 180 days.
37. The method of claim 33, wherein the administration of the composition exhibits dose proportionality.
38. The method of claim 33, wherein the testosterone comprises an enantiomer, racemic mixture, a derivative, a base, or a salt thereof.
39. The method of claim 33, wherein the subject is a male human.
40. The method of claim 39, wherein the serum testosterone concentration is maintained between about 400 ng testosterone per dl serum to about 1050 ng testosterone per dl serum.
41. The method of claim 39, wherein after at least about 30 days of daily administration serum testosterone concentration in the subject is at least about 490 ng/dl to about 860 ng/dl.
42. The method of claim 39, wherein after at least about 30 days of daily administration serum dihydrotestosterone concentration in the subject is greater than about 54 ng/dl.
43. The method of claim 39, wherein after at least about 30 days of daily administration a ratio of serum dihydrotestosterone concentration to serum testosterone concentration of greater than about 0.23 is achieved in the subject.
44. The method of claim 39, wherein after at least about 30 days of daily administration total serum androgen concentration in the subject is greater than about 372 ng/dl.
45. The method of claim 39, wherein after at least about 30 days of daily administration serum estradiol concentration in the subject is greater than about 28 pg/ml.
46. The method of claim 39, wherein the subject has primary hypogonadism prior to administration.
47. The method of claim 46, wherein after at least about 30 days of daily administration serum follicle stimulating hormone concentration in the subject is less than about 11 mIU/ml.
48. The method of claim 39, wherein the subject has secondary hypogonadism prior to administration.
49. The method of claim 48, wherein after at least about 30 days of daily administration serum follicle stimulating hormone concentration in the subject is less than about 3.7 mIU/ml.
50. The method of claim 39, wherein the subject has a pretreatment serum follicle stimulating hormone concentration greater than a normal range of a normal subject.
51. The method of claim 50, wherein after at least about 30 days of daily administration the serum follicle stimulating hormone concentration is within or below the normal range.
52. The method of claim 39, wherein the subject has a pretreatment serum luteinizing hormone concentration greater than a normal range of a subject having primary hypogonadism.
53. The method of claim 52, wherein after at least about 30 days of daily administration serum luteinizing hormone concentration is within the normal range.
54. The method of claim 53, wherein after at least about 30 days of daily administration the serum luteinizing hormone concentration is less than about 6.8 mIU/ml.
55. The method of claim 39, wherein after at least about 30 days of daily administration the testosterone has an accumulation ratio in the subject greater than about 1.5.
56. The method of claim 39, wherein after at least about 30 days of daily administration the testosterone has a net ACU0-24 in the subject greater than 220 nmol*h/l.
57. The method of claim 33, wherein for each about 0.1 gram per day administration of the composition to the skin, an increase of at least about 5 ng/dl in serum testosterone concentration results in the subject.
58. The method of claim 33, wherein the composition is provided to the subject for daily administration in a dose of approximately 0.1 g, 2.5 g, 5 g, 7.5 g, or 10 g.
59. The method of claim 58, wherein the dose is approximately a 5 g dose delivering about 50 mg to about 100 mg of testosterone to the skin.
60. The method of claim 58, wherein the dose is approximately a 7.5 mg dose delivering about 50 mg to about 100 mg of testosterone to the skin.
61. The method of claim 58, wherein the dose is approximately a 10 g dose delivering 50 mg to about 100 mg of testosterone to the skin.
62. The method of claim 58, wherein the composition is provided to the subject in one or more packets.
63. The method of claim 33, wherein maximum serum testosterone concentration in the subject is reached about 16 hours after administration of the composition on day one of administration.
64. The method of claim 33, wherein the composition is a dosage form containing about 1.0 gram to about 10 grams of the composition.
65. The method of claim 33, wherein the composition is a dosage form containing about 2.5 grams to about 7.5 grams of the composition.
66. The method of claim 33, wherein the composition is a dosage form containing about 2.5 grams to about 5.0 grams of the composition.
67. The method of claim 33, wherein the composition comprises about 0.5% to about 5% (w/w) testosterone.
68. The method of claim 33, wherein the composition comprises about 1% (w/w) testosterone.
69. The method of claim 33, wherein the penetration enhancer comprises isopropyl myristate in an amount of about 0.25% to about 2.5% (w/w) of the composition.
70. The method of claim 69, wherein the composition comprises about 0.5 (w/w) isopropyl myristate.
71. The method of claim 33, wherein the gelling agent is polyacrylic acid.
72. The method of claim 71, wherein the composition comprises about 0.9% (w/w) polyacrylic acid.
73. The method of claim 33, wherein the composition comprises about 40% to about 90% alcohol.
US10/867,445 2000-08-30 2004-06-14 Pharmaceutical composition and method for treating hypogonadism Abandoned US20050142173A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US10/867,445 US20050142173A1 (en) 2000-08-30 2004-06-14 Pharmaceutical composition and method for treating hypogonadism
US13/071,264 US20110201586A1 (en) 2000-08-30 2011-03-24 Pharmaceutical composition and method for treating hypogonadism
US13/071,276 US20110172196A1 (en) 2000-08-30 2011-03-24 Pharmaceutical composition and method for treating hypogonadism
US13/902,035 US20130261097A1 (en) 2000-08-30 2013-05-24 Pharmaceutical composition and method for treating hypogonadism
US13/942,245 US9132089B2 (en) 2000-08-30 2013-07-15 Pharmaceutical composition and method for treating hypogonadism
US13/965,499 US9125816B2 (en) 2000-08-30 2013-08-13 Pharmaceutical composition and method for treating hypogonadism
US14/800,949 US20150313914A1 (en) 2000-08-30 2015-07-16 Pharmaceutical composition and method for treating hypogonadism

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09/651,777 US6503894B1 (en) 2000-08-30 2000-08-30 Pharmaceutical composition and method for treating hypogonadism
US10/248,267 US20030232072A1 (en) 2000-08-30 2003-01-03 Pharmaceutical composition and method for treating hypogonadism
US10/867,445 US20050142173A1 (en) 2000-08-30 2004-06-14 Pharmaceutical composition and method for treating hypogonadism

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/248,267 Continuation US20030232072A1 (en) 2000-08-30 2003-01-03 Pharmaceutical composition and method for treating hypogonadism

Related Child Applications (5)

Application Number Title Priority Date Filing Date
US13/071,276 Continuation US20110172196A1 (en) 2000-08-30 2011-03-24 Pharmaceutical composition and method for treating hypogonadism
US13/071,264 Continuation US20110201586A1 (en) 2000-08-30 2011-03-24 Pharmaceutical composition and method for treating hypogonadism
US13/902,035 Continuation US20130261097A1 (en) 2000-08-30 2013-05-24 Pharmaceutical composition and method for treating hypogonadism
US13/942,245 Continuation US9132089B2 (en) 2000-08-30 2013-07-15 Pharmaceutical composition and method for treating hypogonadism
US13/965,499 Continuation US9125816B2 (en) 2000-08-30 2013-08-13 Pharmaceutical composition and method for treating hypogonadism

Publications (1)

Publication Number Publication Date
US20050142173A1 true US20050142173A1 (en) 2005-06-30

Family

ID=24614189

Family Applications (13)

Application Number Title Priority Date Filing Date
US09/651,777 Expired - Lifetime US6503894B1 (en) 2000-08-30 2000-08-30 Pharmaceutical composition and method for treating hypogonadism
US10/046,454 Abandoned US20020183296A1 (en) 2000-08-30 2001-10-19 Pharmaceutical composition and method for treating hypogonadism
US10/033,101 Abandoned US20030050292A1 (en) 2000-08-30 2001-10-19 Pharmaceutical composition and method for treating hypogonadism
US10/248,267 Abandoned US20030232072A1 (en) 2000-08-30 2003-01-03 Pharmaceutical composition and method for treating hypogonadism
US10/825,540 Abandoned US20050113353A1 (en) 2000-08-30 2004-04-15 Pharmaceutical composition and method for treating hypogonadism
US10/828,678 Abandoned US20050112181A1 (en) 2000-08-30 2004-04-20 Pharmaceutical composition and method for treating hypogonadism
US10/867,445 Abandoned US20050142173A1 (en) 2000-08-30 2004-06-14 Pharmaceutical composition and method for treating hypogonadism
US13/071,264 Abandoned US20110201586A1 (en) 2000-08-30 2011-03-24 Pharmaceutical composition and method for treating hypogonadism
US13/071,276 Abandoned US20110172196A1 (en) 2000-08-30 2011-03-24 Pharmaceutical composition and method for treating hypogonadism
US13/902,035 Abandoned US20130261097A1 (en) 2000-08-30 2013-05-24 Pharmaceutical composition and method for treating hypogonadism
US13/942,245 Expired - Fee Related US9132089B2 (en) 2000-08-30 2013-07-15 Pharmaceutical composition and method for treating hypogonadism
US13/965,499 Expired - Fee Related US9125816B2 (en) 2000-08-30 2013-08-13 Pharmaceutical composition and method for treating hypogonadism
US14/800,949 Abandoned US20150313914A1 (en) 2000-08-30 2015-07-16 Pharmaceutical composition and method for treating hypogonadism

Family Applications Before (6)

Application Number Title Priority Date Filing Date
US09/651,777 Expired - Lifetime US6503894B1 (en) 2000-08-30 2000-08-30 Pharmaceutical composition and method for treating hypogonadism
US10/046,454 Abandoned US20020183296A1 (en) 2000-08-30 2001-10-19 Pharmaceutical composition and method for treating hypogonadism
US10/033,101 Abandoned US20030050292A1 (en) 2000-08-30 2001-10-19 Pharmaceutical composition and method for treating hypogonadism
US10/248,267 Abandoned US20030232072A1 (en) 2000-08-30 2003-01-03 Pharmaceutical composition and method for treating hypogonadism
US10/825,540 Abandoned US20050113353A1 (en) 2000-08-30 2004-04-15 Pharmaceutical composition and method for treating hypogonadism
US10/828,678 Abandoned US20050112181A1 (en) 2000-08-30 2004-04-20 Pharmaceutical composition and method for treating hypogonadism

Family Applications After (6)

Application Number Title Priority Date Filing Date
US13/071,264 Abandoned US20110201586A1 (en) 2000-08-30 2011-03-24 Pharmaceutical composition and method for treating hypogonadism
US13/071,276 Abandoned US20110172196A1 (en) 2000-08-30 2011-03-24 Pharmaceutical composition and method for treating hypogonadism
US13/902,035 Abandoned US20130261097A1 (en) 2000-08-30 2013-05-24 Pharmaceutical composition and method for treating hypogonadism
US13/942,245 Expired - Fee Related US9132089B2 (en) 2000-08-30 2013-07-15 Pharmaceutical composition and method for treating hypogonadism
US13/965,499 Expired - Fee Related US9125816B2 (en) 2000-08-30 2013-08-13 Pharmaceutical composition and method for treating hypogonadism
US14/800,949 Abandoned US20150313914A1 (en) 2000-08-30 2015-07-16 Pharmaceutical composition and method for treating hypogonadism

Country Status (24)

Country Link
US (13) US6503894B1 (en)
EP (3) EP2070517A3 (en)
KR (1) KR100866715B1 (en)
CN (3) CN101077350B (en)
AT (1) ATE429920T1 (en)
AU (2) AU9059801A (en)
BR (1) BRPI0113670B8 (en)
CY (1) CY1110907T1 (en)
DE (1) DE60138553D1 (en)
DK (1) DK1313482T3 (en)
EA (1) EA009815B1 (en)
ES (1) ES2326621T3 (en)
HU (1) HU230401B1 (en)
IL (2) IL154668A0 (en)
MA (1) MA27127A1 (en)
NO (1) NO335506B1 (en)
NZ (1) NZ524473A (en)
OA (1) OA12386A (en)
PL (1) PL205875B1 (en)
PT (1) PT1313482E (en)
SI (1) SI1313482T1 (en)
UA (1) UA76958C2 (en)
WO (1) WO2002017926A1 (en)
ZA (3) ZA200301705B (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060100186A1 (en) * 2003-06-18 2006-05-11 White Hillary D Trandsdermal compositions and methods for treatment of fibromyalgia and chronic fatigue syndrome
US20070237822A1 (en) * 2005-10-12 2007-10-11 Ramana Malladi Testosterone gel and method of use
US20110118227A1 (en) * 2003-06-18 2011-05-19 White Mountain Pharma, Inc. Methods for the Treatment of Fibromyalgia and Chronic Fatigue Syndrome
US20110172196A1 (en) * 2000-08-30 2011-07-14 Dudley Robert E Pharmaceutical composition and method for treating hypogonadism
US9642863B2 (en) 2010-11-18 2017-05-09 White Mountain Pharma, Inc. Methods for treating chronic or unresolvable pain and/or increasing the pain threshold in a subject and pharmaceutical compositions for use therein
US9757388B2 (en) 2011-05-13 2017-09-12 Acerus Pharmaceuticals Srl Intranasal methods of treating women for anorgasmia with 0.6% and 0.72% testosterone gels
US10111888B2 (en) 2011-05-13 2018-10-30 Acerus Biopharma Inc. Intranasal 0.15% and 0.24% testosterone gel formulations and use thereof for treating anorgasmia or hypoactive sexual desire disorder
US10668084B2 (en) 2011-05-13 2020-06-02 Acerus Biopharma Inc. Intranasal lower dosage strength testosterone gel formulations and use thereof for treating anorgasmia or hypoactive sexual desire disorder
US11090312B2 (en) 2013-03-15 2021-08-17 Acerus Biopharma Inc. Methods of treating hypogonadism with transnasal testerosterone bio-adhesive gel formulations in male with allergic rhinitis, and methods for preventing an allergic rhinitis event

Families Citing this family (94)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AUPN814496A0 (en) * 1996-02-19 1996-03-14 Monash University Dermal penetration enhancer
US5968547A (en) 1997-02-24 1999-10-19 Euro-Celtique, S.A. Method of providing sustained analgesia with buprenorphine
CN1879630A (en) * 1998-06-11 2006-12-20 内部研究股份有限公司 Pharmaceutical compositions and uses for androst-5-ene-3 beta,17 beta-diol
US6248363B1 (en) * 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US20030236236A1 (en) * 1999-06-30 2003-12-25 Feng-Jing Chen Pharmaceutical compositions and dosage forms for administration of hydrophobic drugs
US20040198706A1 (en) * 2003-03-11 2004-10-07 Carrara Dario Norberto R. Methods and formulations for transdermal or transmucosal application of active agents
US8980290B2 (en) 2000-08-03 2015-03-17 Antares Pharma Ipl Ag Transdermal compositions for anticholinergic agents
US7198801B2 (en) * 2000-08-03 2007-04-03 Antares Pharma Ipl Ag Formulations for transdermal or transmucosal application
ATE485837T1 (en) * 2000-08-03 2010-11-15 Antares Pharma Ipl Ag COMPOSITION FOR TRANSDERMAL AND/OR TRANSMUCOSAL ADMINISTRATION OF ACTIVE INGREDIENTS WHICH GUARANTEES ADEQUATE THERAPEUTIC LEVELS
US20070225379A1 (en) * 2001-08-03 2007-09-27 Carrara Dario Norberto R Transdermal delivery of systemically active central nervous system drugs
US20030139384A1 (en) * 2000-08-30 2003-07-24 Dudley Robert E. Method for treating erectile dysfunction and increasing libido in men
US20040092494A9 (en) * 2000-08-30 2004-05-13 Dudley Robert E. Method of increasing testosterone and related steroid concentrations in women
US20040002482A1 (en) * 2000-08-30 2004-01-01 Dudley Robert E. Androgen pharmaceutical composition and method for treating depression
WO2002051421A2 (en) * 2000-12-22 2002-07-04 Dr. August Wolff Gmbh & Co. Gel composition and trans-scrotal application of a composition for the treatment of hypogonadism
NZ551013A (en) 2001-07-09 2008-04-30 Repros Therapeutics Inc Composition comprising 0-29% cis-clomiphene and 79-100% trans-clomiphene testosterone deficiency in men
US7737185B2 (en) * 2001-07-09 2010-06-15 Repros Therapeutics Inc. Methods and compositions with trans-clomiphene
MXPA04006017A (en) 2001-12-20 2005-06-08 Femmepharma Inc Vaginal delivery of drugs.
TWI339122B (en) * 2002-03-15 2011-03-21 Lab Besins Iscovesco Androgen pharmaceutical composition,kit containing the same and method for using the same
EP1511494B1 (en) * 2002-03-15 2020-08-19 Unimed Pharmaceuticals, LLC Androgen pharmaceutical composition and method for treating depression
MY139721A (en) * 2002-04-19 2009-10-30 Cpex Pharmaceuticals Inc Pharmaceutical composition
NZ537360A (en) 2002-06-25 2006-09-29 Acrux Dds Pty Ltd Transdermal delivery rate control using amorphous pharmaceutical compositions
US20040115287A1 (en) * 2002-12-17 2004-06-17 Lipocine, Inc. Hydrophobic active agent compositions and methods
US7485623B2 (en) * 2002-12-18 2009-02-03 Laboratoires Besins International Sa Reduction of breast density with 4-hydroxy tamoxifen
US7786172B2 (en) * 2002-12-18 2010-08-31 Laboratories Besins International Treatment of mastalgia with 4-hydroxy tamoxifen
US9173836B2 (en) 2003-01-02 2015-11-03 FemmeParma Holding Company, Inc. Pharmaceutical preparations for treatments of diseases and disorders of the breast
EP1578421A4 (en) 2003-01-02 2009-04-22 Femmepharma Holding Co Inc Pharmaceutical preparations for treatments of diseases and disorders of the breast
JP5490346B2 (en) * 2003-04-01 2014-05-14 ブザン ヘルスケア ルクセンブルグ エスアーエールエル Prevention and treatment of breast cancer with 4-hydroxy tamoxifen
US7879357B2 (en) * 2003-04-28 2011-02-01 Bayer Schering Pharma Ag Pharmaceutical composition in the form of a hydrogel for transdermal administration of active ingredients
AU2004246812B8 (en) * 2003-06-09 2010-01-14 Besins Healthcare Luxembourg Sarl Treatment and prevention of excessive scarring with 4-hydroxy tamoxifen
US20050042182A1 (en) * 2003-08-13 2005-02-24 Moshe Arkin Topical compositions of urea
US20050037040A1 (en) * 2003-08-13 2005-02-17 Moshe Arkin Topical compositions of urea and ammonium lactate
US20050036953A1 (en) * 2003-08-13 2005-02-17 Moshe Arkin Topical compositions of ammonium lactate
US20050025833A1 (en) * 2003-07-16 2005-02-03 Chaim Aschkenasy Pharmaceutical composition and method for transdermal drug delivery
US20050020552A1 (en) * 2003-07-16 2005-01-27 Chaim Aschkenasy Pharmaceutical composition and method for transdermal drug delivery
US20050042268A1 (en) * 2003-07-16 2005-02-24 Chaim Aschkenasy Pharmaceutical composition and method for transdermal drug delivery
MXPA06003316A (en) * 2003-10-10 2006-06-08 Antares Pharma Ipl Ag Transdermal pharmaceutical formulation for minimizing skin residues.
US20060003002A1 (en) * 2003-11-03 2006-01-05 Lipocine, Inc. Pharmaceutical compositions with synchronized solubilizer release
EP1550440A1 (en) * 2003-12-15 2005-07-06 Laboratoires Besins International Use of 4-hydroxytamoxifen for the preparation of a medicament for the treatment of gynecomastia
US7968532B2 (en) * 2003-12-15 2011-06-28 Besins Healthcare Luxembourg Treatment of gynecomastia with 4-hydroxy tamoxifen
DK1694319T3 (en) * 2003-12-15 2008-11-17 Besins Int Lab Uses of 4-hydroxytamoxifene in the manufacture of a drug for the treatment of gynecomastia
US20050143362A1 (en) * 2003-12-31 2005-06-30 Mclane Michael W. Danazol for treatment of hypogonadism in the adult male
US20050271597A1 (en) * 2004-02-13 2005-12-08 Keith Alec D Prostate hypertrophy treatment composition and method
US7507769B2 (en) * 2004-03-22 2009-03-24 Laboratoires Besins International Treatment and prevention of benign breast disease with 4-hydroxy tamoxifen
EP1579857A1 (en) * 2004-03-22 2005-09-28 Laboratoires Besins International Chemically stable compositions of 4-hydroxy tamoxifen
US20050244502A1 (en) * 2004-04-28 2005-11-03 Mathias Neil R Composition for enhancing absorption of a drug and method
US7425340B2 (en) * 2004-05-07 2008-09-16 Antares Pharma Ipl Ag Permeation enhancing compositions for anticholinergic agents
MX2007000050A (en) * 2004-07-14 2007-07-10 Repros Therapeutics Inc Trans-clomiphene for the treatment of benign prostate hypertrophy, prostate cancer, hypogonadism, elevated triglycerides and high cholesterol.
EP1634583A1 (en) * 2004-09-09 2006-03-15 Laboratoires Besins International Testosterone gels comprising propylene glycol as penetration enhancer
MX2007009077A (en) * 2005-02-04 2007-09-13 Repros Therapeutics Inc Methods and materials with trans-clomiphene for the treatment of male infertility.
JP5785680B2 (en) * 2005-03-22 2015-09-30 リプロス セラピューティクス インコーポレイテッド Dosing regimen for trans-clomiphene
US20070088012A1 (en) * 2005-04-08 2007-04-19 Woun Seo Method of treating or preventing type-2 diabetes
CN101217874B (en) * 2005-04-12 2012-06-06 优尼麦德药物股份有限公司 Method of treating or preventing bone deterioration or osteoporosis
JP2008536851A (en) * 2005-04-13 2008-09-11 ユニメド ファーマスーティカルズ インク How to increase testosterone and related steroid levels in women
WO2007124250A2 (en) 2006-04-21 2007-11-01 Antares Pharma Ipl Ag Methods of treating hot flashes with formulations for transdermal or transmucosal application
US8067399B2 (en) 2005-05-27 2011-11-29 Antares Pharma Ipl Ag Method and apparatus for transdermal or transmucosal application of testosterone
ES2607454T3 (en) * 2005-06-03 2017-03-31 Acrux Dds Pty Ltd Method and composition for transdermal testosterone delivery
US20070065494A1 (en) * 2005-08-03 2007-03-22 Watson Laboratories, Inc. Formulations and Methods for Enhancing the Transdermal Penetration of a Drug
US7641675B2 (en) * 2006-03-08 2010-01-05 Warsaw Orthopedic, Inc. Flexible bone plates and methods for dynamic spinal stabilization
TW200815045A (en) * 2006-06-29 2008-04-01 Jazz Pharmaceuticals Inc Pharmaceutical compositions of ropinirole and methods of use thereof
EP1891945A1 (en) * 2006-07-31 2008-02-27 Laboratoires Besins International Treatment and prevention of excessive scarring
EP2046310B1 (en) * 2006-07-31 2011-11-02 Besins Healthcare Luxembourg SARL Treatment and prevention of excessive scarring
US8206390B2 (en) * 2006-11-02 2012-06-26 Warsaw Orthopedic, Inc. Uni-directional ratcheting bone plate assembly
WO2008067991A2 (en) * 2006-12-08 2008-06-12 Antares Pharma Ipl Ag Skin-friendly drug complexes for transdermal administration
CA2674078C (en) * 2006-12-26 2012-03-20 Femmepharma Holding Company, Inc. Topical administration of danazol
AU2007343579B2 (en) 2007-01-11 2011-11-17 Acrux Dds Pty Ltd Spreading implement
KR100871531B1 (en) * 2007-01-16 2008-12-05 익수제약 주식회사 Topical preparation of testosterone
US20080261937A1 (en) * 2007-03-23 2008-10-23 Dudley Robert E Pharmaceutical compositions and method for treating pediatric hypogonadism
EA018775B1 (en) * 2007-03-23 2013-10-30 Юнимед Фармасьютикалз Ллк Use of testosterone as hydroalcoholic gel formulation for treating pediatric hypogonadism
CA2866905C (en) 2007-10-16 2016-04-19 Repros Therapeutics, Inc. Selected estrogen receptor modulator (serm) for type 2 diabetes
EP2257220B1 (en) * 2008-03-13 2017-06-14 Liebel-Flarsheim Company LLC Multi-function, foot-activated controller for imaging system
EP2340043A4 (en) * 2008-10-31 2012-09-19 Moberg Derma Ab Topical composition comprising a combination of at least two penetration enhancing agents
US11304960B2 (en) * 2009-01-08 2022-04-19 Chandrashekar Giliyar Steroidal compositions
US10080760B2 (en) 2009-10-27 2018-09-25 Besins Healthcare Luxembourg Sarl Transdermal pharmaceutical compositions comprising active agents
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
US20180153904A1 (en) 2010-11-30 2018-06-07 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
KR20230041081A (en) * 2011-05-15 2023-03-23 에이세러스 바이오파마 인크. Intranasal testosterone bio-adhesive gel formulations and use thereof for treating male hypogonadism
UA113291C2 (en) 2011-08-04 2017-01-10 TRANSCLOMYPHENE METABOLITES AND THEIR APPLICATIONS
WO2014070523A1 (en) 2012-11-02 2014-05-08 Repros Therapeutics Inc. Trans-clomiphene for use in cancer therapy
US10201549B2 (en) 2013-06-14 2019-02-12 Professional Compounding Centers Of America (Pcca) Testosterone combined with anastrozole injection solutions
USD749225S1 (en) 2013-11-26 2016-02-09 Acrux Dds Pty Ltd Topical spreading applicator
USD750788S1 (en) 2013-11-26 2016-03-01 Acrux Dds Pty Ltd Topical spreading applicator
US8785426B1 (en) 2013-12-13 2014-07-22 Upsher-Smith Laboratories, Inc. Testosterone gel compositions and related methods
US11439668B2 (en) 2014-05-23 2022-09-13 JangoBio, LLC Methods to differentiate stem cells into hormone-producing cells
US11253549B2 (en) * 2014-05-23 2022-02-22 JangoBio, LLC Methods to rebalance the hypothalamic-pituitary-gonadal axis
US20160051498A1 (en) * 2014-08-20 2016-02-25 Professional Compounding Centers Of America (Pcca) Transdermal Pharmaceutical Compositions Including Testosterone and a C-SERM
US9498485B2 (en) 2014-08-28 2016-11-22 Lipocine Inc. Bioavailable solid state (17-β)-hydroxy-4-androsten-3-one esters
US20170246187A1 (en) 2014-08-28 2017-08-31 Lipocine Inc. (17-ß)-3-OXOANDROST-4-EN-17-YL TRIDECANOATE COMPOSITIONS AND METHODS OF THEIR PREPARATION AND USE
US20170014417A1 (en) 2015-07-14 2017-01-19 Lipp Life Sciences Llc Pharmaceutical administration system for the transdermal application of vardenafil
US9824976B1 (en) * 2016-08-16 2017-11-21 Infineon Technologies Americas Corp. Single-sided power device package
EP3544614A4 (en) 2016-11-28 2020-08-05 Lipocine Inc. Oral testosterone undecanoate therapy
WO2019140087A1 (en) 2018-01-10 2019-07-18 Celista Pharmaceuticals Llc Testosterone transdermal spray with film

Citations (40)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US568112A (en) * 1896-09-22 Vehicle-wheel
US4078060A (en) * 1976-05-10 1978-03-07 Richardson-Merrell Inc. Method of inducing an estrogenic response
US4861764A (en) * 1986-11-17 1989-08-29 Macro Chem. Corp. Percutaneous absorption enhancers, compositions containing same and method of use
US5152997A (en) * 1990-12-11 1992-10-06 Theratech, Inc. Method and device for transdermally administering testosterone across nonscrotal skin at therapeutically effective levels
US5200190A (en) * 1988-10-11 1993-04-06 Sekisui Kagaku Kogyo Kabushiki Kaisha Percutaneous pharmaceutical preparation
US5223261A (en) * 1988-02-26 1993-06-29 Riker Laboratories, Inc. Transdermal estradiol delivery system
US5231087A (en) * 1988-03-16 1993-07-27 Cellegy Pharmaceuticals, Inc. Treatment of skin diseases and tumors with esters and amides of monocarboxylic acids
US5232703A (en) * 1989-07-21 1993-08-03 Izhak Blank Estradiol compositions and methods for topical application
US5238944A (en) * 1988-12-15 1993-08-24 Riker Laboratories, Inc. Topical formulations and transdermal delivery systems containing 1-isobutyl-1H-imidazo[4,5-c]quinolin-4-amine
US5550107A (en) * 1989-03-10 1996-08-27 Endorecherche Inc. Combination therapy for the treatment of estrogen-sensitive disease
US5643899A (en) * 1992-06-19 1997-07-01 Cellegy Pharmaceuticals, Inc. Lipids for epidermal moisturization and repair of barrier function
US5723114A (en) * 1993-03-19 1998-03-03 Cellegy Pharmaceuticals Inc. Penetration enhancing compositions and methods of their use
US5730987A (en) * 1996-06-10 1998-03-24 Omar; Lotfy Ismail Medication for impotence containing lyophilized roe and a powdered extract of Ginkgo biloba
US5760096A (en) * 1996-10-18 1998-06-02 Thornfeldt; Carl R. Potent penetration enhancers
US5788984A (en) * 1988-10-27 1998-08-04 Schering Aktiengesellschaft Gestodene-containing agent for transdermal administration
US5788983A (en) * 1989-04-03 1998-08-04 Rutgers, The State University Of New Jersey Transdermal controlled delivery of pharmaceuticals at variable dosage rates and processes
US5942545A (en) * 1998-06-15 1999-08-24 Macrochem Corporation Composition and method for treating penile erectile dysfunction
US5952000A (en) * 1996-10-30 1999-09-14 Theratech, Inc. Fatty acid esters of lactic acid salts as permeation enhancers
US5955455A (en) * 1993-01-19 1999-09-21 Endorecherche, Inc. Therapeutic methods and delivery systems utilizing sex steroid precursors
US5968919A (en) * 1997-10-16 1999-10-19 Macrochem Corporation Hormone replacement therapy drug formulations for topical application to the skin
US6007837A (en) * 1996-07-03 1999-12-28 Alza Corporation Drug delivery devices and process of manufacture
US6019997A (en) * 1997-01-09 2000-02-01 Minnesota Mining And Manufacturing Hydroalcoholic compositions for transdermal penetration of pharmaceutical agents
US6200591B1 (en) * 1998-06-25 2001-03-13 Anwar A. Hussain Method of administration of sildenafil to produce instantaneous response for the treatment of erectile dysfunction
US6319913B1 (en) * 1997-11-10 2001-11-20 Cellegy Pharmaceuticals, Inc. Penetration enhancing and irritation reducing systems
US20020128176A1 (en) * 1999-01-27 2002-09-12 Wolf-Georg Forssmann Treatment of erectile dysfunctions with C-Type natriuretic polypeptide (CNP) as a monotherapy or in combination with phosphodiesterase inhibitors
US6503894B1 (en) * 2000-08-30 2003-01-07 Unimed Pharmaceuticals, Inc. Pharmaceutical composition and method for treating hypogonadism
US20030022877A1 (en) * 2000-08-30 2003-01-30 Dudley Robert E. Method of increasing testosterone and related steroid concentrations in women
US20030027804A1 (en) * 2001-06-27 2003-02-06 Van Der Hoop Roland Gerritsen Therapeutic combinations for the treatment of hormone deficiencies
US6562370B2 (en) * 1999-12-16 2003-05-13 Dermatrends, Inc. Transdermal administration of steroid drugs using hydroxide-releasing agents as permeation enhancers
US6562369B2 (en) * 1999-12-16 2003-05-13 Dermatrends, Inc. Transdermal administration of androgenic drugs hydroxide-releasing agents as permeation enhancers
US6586000B2 (en) * 1999-12-16 2003-07-01 Dermatrends, Inc. Hydroxide-releasing agents as skin permeation enhancers
US20040072810A1 (en) * 2001-11-07 2004-04-15 Besins International Belgique Pharmaceutical composition in the form of a gel or a solution based on dihydrotestosterone, process for preparing it and uses thereof
US20040110732A1 (en) * 2002-12-10 2004-06-10 Besins International Belgique Pharmaceutical composition for transdermal or transmucosal administration comprising at least one progestin and/or at least one oestrogen, process for preparing it and uses thereof
US6818226B2 (en) * 1996-02-19 2004-11-16 Acrux Dds Pty. Ltd. Dermal penetration enhancers and drug delivery systems involving same
US20050020552A1 (en) * 2003-07-16 2005-01-27 Chaim Aschkenasy Pharmaceutical composition and method for transdermal drug delivery
US7097853B1 (en) * 1994-09-14 2006-08-29 3M Innovative Properties Company Matrix for transdermal drug delivery
US7320968B2 (en) * 2002-04-19 2008-01-22 Bentley Pharmaceuticals, Inc. Pharmaceutical composition
US20110269729A1 (en) * 2005-10-12 2011-11-03 Ramana Malladi Testosterone Gel And Method Of Use
US20110306582A1 (en) * 2002-03-15 2011-12-15 Dudley Robert E Androgen pharmaceutical composition and method for treating depression
US20120058981A1 (en) * 2000-08-30 2012-03-08 Dudley Robert E Androgen pharmaceutical composition and method for treating depression

Family Cites Families (329)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2155658A (en) 1936-01-08 1939-04-25 Chemische Forschungs Gmbh Surgical and medical preparations
NL109113C (en) 1957-03-27
GB916778A (en) 1959-10-15 1963-01-30 Vismara Francesco Spa Oral compositions containing 3-substituted 17-ª--methyl testosterones
US3121042A (en) * 1959-05-04 1964-02-11 Ercoli Alberto Oral compositions containing 3-enolethers of methyltestosterone
GB941634A (en) 1960-11-16 1963-11-13 Upjohn Co Improvements in or relating to steroids and the manufacture thereof
US3218283A (en) 1962-07-26 1965-11-16 Monsanto Co Novel solutions of poly-(acrylic anhydride) and poly-(methacrylic anhydride) polymers
US3164520A (en) * 1962-10-29 1965-01-05 Olin Mathieson Injectable steroid compositions containing at least 75% benzyl benzoate
GB1158283A (en) 1965-10-21 1969-07-16 Foster Milburn Company Composition to be Applied to Skin and Process for Preparing Same.
US3888995A (en) 1968-07-19 1975-06-10 Syntex Corp Fatty alcohol-propylene glycol vehicle
US3887699A (en) 1969-03-24 1975-06-03 Seymour Yolles Biodegradable polymeric article for dispensing drugs
US3913789A (en) * 1974-02-13 1975-10-21 United States Banknote Corp Fluid container of the flexible wall capsule type
US4009254A (en) 1974-05-06 1977-02-22 Colgate-Palmolive Company Topical compositions
US3939111A (en) 1974-07-01 1976-02-17 The B. F. Goodrich Company Stable polyurethane solutions
US3989816A (en) 1975-06-19 1976-11-02 Nelson Research & Development Company Vehicle composition containing 1-substituted azacycloheptan-2-ones
US4197316A (en) * 1975-07-23 1980-04-08 Scott Eugene J Van Treatment of dry skin
NL7510104A (en) 1975-08-27 1977-03-01 Akzo Nv PROCESS FOR THE PREPARATION OF AN ORAL ACTIVE PHARMACEUTICAL PREPARATION.
CH625702A5 (en) 1977-01-18 1981-10-15 Delalande Sa
DE2747531A1 (en) * 1977-10-22 1979-04-26 Basf Ag SUBSTITUTED 3-AMINOPYRAZOLE
US4954487A (en) 1979-01-08 1990-09-04 The Procter & Gamble Company Penetrating topical pharmaceutical compositions
US4299826A (en) 1979-10-12 1981-11-10 The Procter & Gamble Company Anti-acne composition
CA1165240A (en) 1980-07-09 1984-04-10 The Procter & Gamble Company Penetrating topical pharmaceutical compositions
US4346709A (en) 1980-11-10 1982-08-31 Alza Corporation Drug delivery devices comprising erodible polymer and erosion rate modifier
US4442094A (en) 1980-12-23 1984-04-10 Merck & Co., Inc. (3-Aralkylamino-2-or-propoxy)heterocyclic compounds
US4440777A (en) * 1981-07-07 1984-04-03 Merck & Co., Inc. Use of eucalyptol for enhancing skin permeation of bio-affecting agents
US4447562A (en) 1981-07-15 1984-05-08 Ivani Edward J Amino-polysaccharides and copolymers thereof for contact lenses and ophthalmic compositions
FR2515041A1 (en) 1981-10-26 1983-04-29 Besins Jean PROGESTERONE-BASED DRUG FOR THE TREATMENT OF MAMMARY DISEASES
FR2518879A1 (en) 1981-12-30 1983-07-01 Besins Jean Topical oestradiol compsns. - for treatment of disorders associated with menopause
FR2519252A1 (en) 1982-01-07 1983-07-08 Besins Jean Percutaneous gel contg. di:hydro:testosterone - for treating deficient secretion of testicular androgen
JPH0782147B2 (en) 1982-10-21 1995-09-06 キヤノン株式会社 Focusing method of zoom lens
US4496556A (en) 1982-08-16 1985-01-29 Norman Orentreich Topical applications for preventing dry skin
DE3315654A1 (en) * 1983-04-29 1984-10-31 Bosch Gmbh Robert POLAROGRAPHIC PROBE FOR DETERMINING THE OXYGEN CONTENT IN GASES
NL8301550A (en) 1983-05-03 1984-12-03 Gist Brocades Nv IMIDAZOLETHANOL ESTERS.
US4478822A (en) 1983-05-16 1984-10-23 Merck & Co., Inc. Drug delivery system utilizing thermosetting gels
US4557934A (en) 1983-06-21 1985-12-10 The Procter & Gamble Company Penetrating topical pharmaceutical compositions containing 1-dodecyl-azacycloheptan-2-one
US4631188A (en) 1983-08-31 1986-12-23 S.K.Y. Polymers, Ltd. (Kingston Technologies) Injectable physiologically-acceptable polymeric composition
DE3333240A1 (en) 1983-09-12 1985-03-28 Schering AG, 1000 Berlin und 4709 Bergkamen MEDIUM FOR TRANSDERMAL APPLICATION OF MEDICINAL PRODUCTS
US4690775A (en) 1983-09-30 1987-09-01 Research Corporation Emulsion-based gel and process for preparing same
WO1985002092A1 (en) 1983-11-14 1985-05-23 Bio-Mimetics Inc. Bioadhesive compositions and methods of treatment therewith
US4670254A (en) 1983-12-09 1987-06-02 Toko Yakuhin Industry Co., Ltd. Gel preparations for topical application of diclofenac sodium
FR2558373B1 (en) * 1984-01-20 1987-07-03 Mauvais Jarvis Pierre ANTI-ESTROGEN MEDICINAL PRODUCT BASED ON 4-HYDROXYTAMOXIFENE FOR PERCUTANEOUS ADMINISTRATION
GB8508404D0 (en) 1985-03-30 1985-05-09 Baylor College Medicine Therapeutic compositions
CA1248450A (en) 1984-04-05 1989-01-10 Kazuo Kigasawa Soft patch
GB8416234D0 (en) 1984-06-26 1984-08-01 Ici Plc Biodegradable amphipathic copolymers
US4704282A (en) 1984-06-29 1987-11-03 Alza Corporation Transdermal therapeutic system having improved delivery characteristics
US4725439A (en) 1984-06-29 1988-02-16 Alza Corporation Transdermal drug delivery device
US4791099A (en) 1984-10-29 1988-12-13 Chaovanee Aroonsakul Method of treatment for central nervous system diseases such as Alzheimer's's disease
GB2167408B (en) 1984-11-23 1988-05-25 Farmos Oy Substituted imidazole derivatives and their preparation and use
EP0189861A3 (en) 1985-01-26 1988-02-17 Showa Denko Kabushiki Kaisha Percutaneous absorption accelerator for ionic water-soluble medicine
DE3685895T2 (en) 1985-02-25 1992-12-24 Univ Rutgers DOSING SYSTEM FOR TRANSDERMAL ABSORPTION OF DRUG ACTIVE SUBSTANCES.
US4663157A (en) * 1985-02-28 1987-05-05 The Proctor & Gamble Company Sunscreen compositions
US4767627A (en) 1985-05-29 1988-08-30 Merck & Co., Inc. Drug delivery device which can be retained in the stomach for a controlled period of time
JPS6211675U (en) 1985-07-04 1987-01-24
US4994265A (en) 1985-09-06 1991-02-19 Aloe Scientific Labs Shaving composition
US4683242A (en) 1985-10-28 1987-07-28 A. H. Robins Company, Incorporated Transdermal treatment for pain and inflammation with 2-amino-3-aroylbenzeneacetic acids, salts and esters
DE3687692T2 (en) 1985-11-13 1993-05-19 Japan Res Dev Corp GENDER HORMONES FOR THE TREATMENT OF IMMUNE-DEFICIENT DISEASES.
EP0248885B1 (en) 1985-12-04 1992-08-05 HSIEH, Dean Transdermal delivery of drugs
US5023252A (en) 1985-12-04 1991-06-11 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery of drugs
US5731303A (en) * 1985-12-04 1998-03-24 Conrex Pharmaceutical Corporation Transdermal and trans-membrane delivery compositions
US4699779A (en) 1986-02-18 1987-10-13 Victor Palinczar Waterproof sunscreen compositions
US4820724A (en) * 1986-03-31 1989-04-11 University Of Southern California Dual phase solvent carrier system
US4780320A (en) 1986-04-29 1988-10-25 Pharmetrix Corp. Controlled release drug delivery system for the periodontal pocket
US4946870A (en) 1986-06-06 1990-08-07 Union Carbide Chemicals And Plastics Company Inc. Delivery systems for pharmaceutical or therapeutic actives
US4863911A (en) 1986-08-04 1989-09-05 University Of Florida Method for treating male sexual dysfunction
MY102980A (en) 1986-10-31 1993-03-31 Pfizer Transdermal flux enhancing compositions
US4863970A (en) 1986-11-14 1989-09-05 Theratech, Inc. Penetration enhancement with binary system of oleic acid, oleins, and oleyl alcohol with lower alcohols
US5326790A (en) 1986-11-19 1994-07-05 Dermatologic Research Corporation Administration of skin medications by use of dicarboxylic acids and derivatives
US4981696A (en) 1986-12-22 1991-01-01 E. I. Du Pont De Nemours And Company Polylactide compositions
AU601528B2 (en) 1986-12-22 1990-09-13 Ortho-Mcneil Pharmaceutical, Inc. Resilient transdermal drug-delivery device and compositions and devices employing fatty acid esters/ethers of alkanediols and percutaneous absorption enhancers
US4906169A (en) * 1986-12-29 1990-03-06 Rutgers, The State University Of New Jersey Transdermal estrogen/progestin dosage unit, system and process
US4788062A (en) 1987-02-26 1988-11-29 Alza Corporation Transdermal administration of progesterone, estradiol esters, and mixtures thereof
BE1000381A4 (en) 1987-03-13 1988-11-16 Pharlyse Sa PHARMACEUTICAL PREPARATION BASED indomethacin.
US4855305A (en) 1987-03-23 1989-08-08 Applied Medical Research Compositions and methods of effecting contraception utilizing melatonin
JPS6479103A (en) 1987-06-09 1989-03-24 Lion Corp External preparation
US5013553A (en) 1987-06-30 1991-05-07 Vipont Pharmaceutical, Inc. Drug delivery devices
US5256652A (en) 1987-11-12 1993-10-26 Pharmedic Co. Topical compositions and methods for treatment of male impotence
JPH0798935B2 (en) 1987-11-25 1995-10-25 有限会社野々川商事 Quality deterioration inhibitor
US4920203A (en) 1987-12-17 1990-04-24 Allied-Signal Inc. Medical devices fabricated from homopolymers and copolymers having recurring carbonate units
US5656286A (en) 1988-03-04 1997-08-12 Noven Pharmaceuticals, Inc. Solubility parameter based drug delivery system and method for altering drug saturation concentration
US5234957A (en) 1991-02-27 1993-08-10 Noven Pharmaceuticals, Inc. Compositions and methods for topical administration of pharmaceutically active agents
US5719197A (en) 1988-03-04 1998-02-17 Noven Pharmaceuticals, Inc. Compositions and methods for topical administration of pharmaceutically active agents
US5446070A (en) 1991-02-27 1995-08-29 Nover Pharmaceuticals, Inc. Compositions and methods for topical administration of pharmaceutically active agents
IE62871B1 (en) 1988-03-08 1995-03-08 Warner Lambert Co Compositions with enhanced penetration
US5641504A (en) 1988-06-09 1997-06-24 Alza Corporation Skin permeation enhancer compositions using glycerol monolinoleate
DE3836862A1 (en) 1988-10-27 1990-05-03 Schering Ag Composition for the transdermal administration of steroid hormones
US5332577A (en) 1988-12-27 1994-07-26 Dermamed Transdermal administration to humans and animals
CA2011423A1 (en) 1989-03-07 1990-09-07 Peter M. Taylor Pharmaceutical compositions useful as drug delivery vehicles and/or as wound dressings
US4917882A (en) 1989-03-16 1990-04-17 Amway Corporation Gel-type sunscreen composition
US5053227A (en) 1989-03-22 1991-10-01 Cygnus Therapeutic Systems Skin permeation enhancer compositions, and methods and transdermal systems associated therewith
ATE107517T1 (en) 1989-05-25 1994-07-15 Takeda Chemical Industries Ltd TRANSDERMAL THERAPEUTIC AGENT.
US5059603A (en) 1989-06-12 1991-10-22 Centuries Laboratories, Inc. Method and composition for treating impotence
US5122519A (en) 1989-06-27 1992-06-16 American Cyanamid Company Stable, cosmetically acceptable topical gel formulation and method of treatment for acne
DK0595796T3 (en) 1989-07-07 2003-05-05 Endorech Inc Method of treating androgen-related diseases
US5116828A (en) * 1989-10-26 1992-05-26 Nippon Zoki Pharmaceutical Co., Ltd. Pharmaceutical composition for treatment of osteoporosis
US5324521A (en) 1989-12-18 1994-06-28 Dermamed Systems for transdermal administration of medicaments
JP2893803B2 (en) 1990-02-27 1999-05-24 日本電気株式会社 Driving method of plasma display
ES2180690T3 (en) 1990-06-01 2003-02-16 Population Council Inc PREPARATION OF COMPOSITIONS THAT INCLUDE ST1435 FOR TOPICAL APPLICATION.
US5250534A (en) 1990-06-20 1993-10-05 Pfizer Inc. Pyrazolopyrimidinone antianginal agents
NZ240358A (en) 1990-10-29 1994-09-27 Alza Corp Transdermal contraceptive device comprising (as active agents) an estrogen and a gestodene together with a skin permeation enhancer
EP0555309A1 (en) 1990-10-31 1993-08-18 BEECHAM GROUP plc Topical composition with retinoid penetration enhancer
US5202125A (en) 1990-12-10 1993-04-13 Theratech, Inc. Method and systems for administering nitroglycerin transdermally at enhanced transdermal fluxes
EP0491076A1 (en) 1990-12-19 1992-06-24 Theratech, Inc. Penetration enhancement with multi-component system of N-aliphatic pyrrolidones with lower alcohols
JPH04261119A (en) 1991-02-13 1992-09-17 Lintec Corp Percutaneous absorption-type pharmaceutical preparation
US5340586A (en) 1991-04-12 1994-08-23 University Of Southern California Methods and formulations for use in treating oophorectomized women
US5211952A (en) 1991-04-12 1993-05-18 University Of Southern California Contraceptive methods and formulations for use therein
US5340585A (en) 1991-04-12 1994-08-23 University Of Southern California Method and formulations for use in treating benign gynecological disorders
US5326566A (en) 1991-05-17 1994-07-05 Bristol-Myers Squibb Company Use of dibutyl adipate and isopropyl myristate in topical and transdermal products
TW218849B (en) 1991-05-17 1994-01-11 Bristol Myers Squibb Co
US5208013A (en) * 1991-06-03 1993-05-04 Olympus International, Inc. Composition for skin care and protection
US5252338A (en) 1991-06-27 1993-10-12 Alza Corporation Therapy delayed
US5487898A (en) * 1991-08-26 1996-01-30 Abbott Laboratories Compositions and method for the sublingual or buccal administration therapeutic agents
US5238933A (en) 1991-10-28 1993-08-24 Sri International Skin permeation enhancer compositions
US5676968A (en) 1991-10-31 1997-10-14 Schering Aktiengesellschaft Transdermal therapeutic systems with crystallization inhibitors
JP3202777B2 (en) 1992-01-24 2001-08-27 リンテック株式会社 Transdermal absorption enhancer and tape preparation
US5629019A (en) 1992-02-27 1997-05-13 Alza Corporation Formulations with hydrophobic permeation enhancers
TW224048B (en) * 1992-03-30 1994-05-21 Hoechst Roussel Pharma
JP2960832B2 (en) 1992-05-08 1999-10-12 ペルマテック テクノロジー アクチェンゲゼルシャフト Estradiol administration system
US5605929A (en) 1992-05-27 1997-02-25 Arch Development Corp. Methods and compositions for inhibiting 5α-reductase activity
WO1993025168A1 (en) 1992-06-11 1993-12-23 Theratech, Inc. The use of glycerin in moderating transdermal drug delivery
US5446025A (en) 1992-06-12 1995-08-29 Abbott Laboratories Formulations and method of the percutaneous administration of leuprolide
US5607691A (en) * 1992-06-12 1997-03-04 Affymax Technologies N.V. Compositions and methods for enhanced drug delivery
DE4223004A1 (en) 1992-07-13 1994-01-20 Liedtke Pharmed Gmbh Single-dose semi-solid topical dosage form for transdermal therapy
WO1994002119A1 (en) 1992-07-23 1994-02-03 Hisamitsu Pharmaceutical Co., Inc. Percutaneously administrable base composition and drug composition prepared therefrom
JP3276406B2 (en) 1992-07-24 2002-04-22 富士通株式会社 Driving method of plasma display
CA2101496A1 (en) 1992-07-31 1994-02-01 Masao Kobayashi Base for transdermal administration
DE4227989A1 (en) 1992-08-21 1994-06-09 Schering Ag Agent for transdermal application containing 3-keto-desogestrel
WO1994008590A1 (en) 1992-10-20 1994-04-28 Btg Pharmaceuticals Corp. A method for ameliorating muscle weakness/wasting in a patient infected with human immunodeficiency virus-type 1
AU5449194A (en) 1992-10-26 1994-05-24 Merck & Co., Inc. Combinations of angiotensin-ii receptor antagonists and diuretics
US5883115A (en) 1992-11-09 1999-03-16 Pharmetrix Division Technical Chemicals & Products, Inc. Transdermal delivery of the eutomer of a chiral drug
US5639743A (en) 1992-11-13 1997-06-17 University Of Georgia Research Foundation Compositions and methods for treating exocrine gland atrophy
WO1994013257A1 (en) 1992-12-16 1994-06-23 Creative Products Resource Associates, Ltd. Occlusive/semi-occlusive lotion for treatment of a skin disease or disorder
ATE275957T1 (en) 1993-01-19 2004-10-15 Endorech Inc THERAPEUTIC USES AND ADMINISTRATION SYSTEMS OF DEHYDROEPIANDROSTERONE
US5881926A (en) * 1993-03-11 1999-03-16 Taro Pharmaceutical Industries, Ltd. Pharmaceutical compositions in semisolid form and a device for administration thereof
IL109036A (en) 1993-03-19 1998-12-27 Cellegy Pharamaceuticals Inc Compositions for disrupting the epithelial barrier function and use of agents for preparing such compositions
IL109037A (en) 1993-03-19 1999-01-26 Cellegy Pharma Inc Compositions for inducing phase separation of epithelial lipid bilayers and preparation of said compositions
WO1994022460A1 (en) * 1993-04-05 1994-10-13 University Patents, Inc. Diagnosis and treatment of erectile dysfunction
IT1271352B (en) 1993-04-08 1997-05-27 Boehringer Ingelheim Italia INDOLE DERIVATIVES USEFUL IN THE TREATMENT OF DISORDERS OF THE CENTRAL NERVOUS SYSTEM
EP0698393B1 (en) 1993-05-19 2002-07-03 Hisamitsu Pharmaceutical Co., Inc. 3-l-MENTHOXY-PROPANE-1, 2-DIOL AS SOLUBILIZING AGENT AND EXTERNAL PREPARATION CONTAINING THE SAME
FR2705572B1 (en) 1993-05-25 1995-08-11 Besins Iscovesco Laboratoires Dihydrotestosterone for androgen therapy.
US5648350A (en) 1993-05-25 1997-07-15 Laboratoires Besins Iscovesco Dihydrotestosterone for use in androgenotherapy
US5698589A (en) * 1993-06-01 1997-12-16 International Medical Innovations, Inc. Water-based topical cream containing nitroglycerin and method of preparation and use thereof
GB9301192D0 (en) 1993-06-09 1993-06-09 Trott Francis W Flower shaped mechanised table
US5460820B1 (en) 1993-08-03 1999-08-03 Theratech Inc Method for providing testosterone and optionally estrogen replacement therapy to women
US5362886A (en) 1993-10-12 1994-11-08 Eli Lilly And Company Asymmetric synthesis
BR9408457A (en) 1993-12-27 1997-08-05 Akzo Nobel Nv Percutaneous pharmaceutical preparation
GB9401090D0 (en) * 1994-01-21 1994-03-16 Glaxo Lab Sa Chemical compounds
US6143746A (en) 1994-01-21 2000-11-07 Icos Corporation Tetracyclic cyclic GMP-specific phosphodiesterase inhibitors, process of preparation and use
GB9514465D0 (en) 1995-07-14 1995-09-13 Glaxo Lab Sa Chemical compounds
CN1106259A (en) 1994-02-05 1995-08-09 日东制药株式会社 Antiphlogistic and analgesic gel agent for external use containing propanoic acid non steroid pharmaceutical as effective composition
FR2718372B1 (en) 1994-04-08 1996-06-28 Sofab Dispenser for fluid products.
US6395744B1 (en) 1994-04-22 2002-05-28 Queen's University At Kingston Method and compositions for the treatment or amelioration of female sexual dysfunction
ATE312609T1 (en) 1994-04-22 2005-12-15 Pentech Pharmaceuticals Inc SUBLINGUAL COMPOSITION CONTAINING APOMORPHINE FOR THE DIAGNOSTICS OF FUNCTIONAL IMPOTENCY
US5540934A (en) 1994-06-22 1996-07-30 Touitou; Elka Compositions for applying active substances to or through the skin
GB9423911D0 (en) 1994-11-26 1995-01-11 Pfizer Ltd Therapeutic agents
GB9423910D0 (en) 1994-11-26 1995-01-11 Pfizer Ltd Therapeutic agents
WO1996019205A1 (en) 1994-12-21 1996-06-27 Theratech, Inc. Transdermal delivery system with adhesive overlay and peel seal disc
US5807568A (en) 1994-12-27 1998-09-15 Mcneil-Ppc, Inc. Enhanced delivery of topical compositions containing flurbiprofen
US6024974A (en) * 1995-01-06 2000-02-15 Noven Pharmaceuticals, Inc. Composition and methods for transdermal delivery of acid labile drugs
IL116539A (en) 1995-01-06 2002-02-10 Noven Pharma Compositions for transdermal delivery of acid-labile drugs
US5629021A (en) 1995-01-31 1997-05-13 Novavax, Inc. Micellar nanoparticles
AT408067B (en) * 1995-03-17 2001-08-27 Gebro Pharma Gmbh PHARMACEUTICAL COMPOSITION FOR TOPICAL APPLICATION AND METHOD FOR THE PRODUCTION THEREOF
US5844103A (en) 1995-03-24 1998-12-01 Lever Brothers Company, Division Of Conopco, Inc. Anionic glycasuccinamide sufactants and a process for their manufacture
US5654337A (en) 1995-03-24 1997-08-05 II William Scott Snyder Topical formulation for local delivery of a pharmaceutically active agent
FR2732223B1 (en) 1995-03-30 1997-06-13 Sanofi Sa PHARMACEUTICAL COMPOSITION FOR TRANSDERMAL ADMINISTRATION
US5731339A (en) * 1995-04-28 1998-03-24 Zonagen, Inc. Methods and formulations for modulating the human sexual response
DE19517145C2 (en) 1995-05-10 2000-02-24 Hexal Pharmaforschung Gmbh Transdermal therapeutic system (TTS) for administration of testosterone
AU5671496A (en) 1995-05-15 1996-11-29 Beth Israel Hospital Use of dihydrotestosterone compounds for treating male sexual dysfunction
US5882676A (en) * 1995-05-26 1999-03-16 Alza Corporation Skin permeation enhancer compositions using acyl lactylates
NZ311304A (en) 1995-06-07 1999-03-29 Cygnus Therapeutic Systems Transdermal patch and method for administering 17-deacetyl norgestimate alone or in combination with an estrogen
US5693335A (en) 1995-06-07 1997-12-02 Cygnus, Inc. Skin permeation enhancer composition for use with sex steroids
US5785991A (en) 1995-06-07 1998-07-28 Alza Corporation Skin permeation enhancer compositions comprising glycerol monolaurate and lauryl acetate
GB9512670D0 (en) 1995-06-21 1995-08-23 Sod Conseils Rech Applic Camptothecin analogues
US5780050A (en) 1995-07-20 1998-07-14 Theratech, Inc. Drug delivery compositions for improved stability of steroids
WO1997004752A1 (en) 1995-07-26 1997-02-13 Duramed Pharmaceuticals, Inc. Pharmaceutical compositions of conjugated estrogens and methods for their use
US5906830A (en) 1995-09-08 1999-05-25 Cygnus, Inc. Supersaturated transdermal drug delivery systems, and methods for manufacturing the same
US5902603A (en) 1995-09-14 1999-05-11 Cygnus, Inc. Polyurethane hydrogel drug reservoirs for use in transdermal drug delivery systems, and associated methods of manufacture and use
US6036977A (en) * 1995-09-29 2000-03-14 L.A.M. Pharmaceutical Corp. Drug preparations for treating sexual dysfunction
US6251436B1 (en) 1995-09-29 2001-06-26 L.A.M. Pharmaceutical Corporation Drug preparations for treating sexual dysfunction
JP4036496B2 (en) 1995-10-24 2008-01-23 リンテック株式会社 Method for producing gel preparation
US5849729A (en) 1995-12-26 1998-12-15 Hershey Foods Corporation Use of hydrolyzed cocoa butter for percutaneous absorption
AU1429697A (en) 1995-12-29 1997-07-28 Cygnus, Inc. Systems and methods for the transdermal administration of androgenic agents
US5643587A (en) 1996-02-15 1997-07-01 Avon Products, Inc. Composition and method for under-eye skin lightening
US6923983B2 (en) 1996-02-19 2005-08-02 Acrux Dds Pty Ltd Transdermal delivery of hormones
US5898038A (en) 1996-03-19 1999-04-27 Board Of Regents, The University Of Texas System Treatment of osteoporosis and metabolic bone disorders with nitric oxide substrate and/or donors
US5762956A (en) 1996-04-24 1998-06-09 Rutgers, The State University Of New Jersey Transdermal contraceptive delivery system and process
AU2746397A (en) 1996-04-30 1997-11-19 Theratech, Inc. Transdermal administration of steroid hormones using diethanolamides of c12-c18 fatty acids as permeation enhancers
WO1997041865A1 (en) 1996-05-02 1997-11-13 Azupharma Gmbh Topical penile androgen application for treatment of erectile dysfunction
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
CA2256365A1 (en) 1996-05-22 1997-11-27 Diversified Pharmaceuticals, Inc. Compositions, methods and devices for the transdermal delivery of drugs
IT1283102B1 (en) * 1996-06-06 1998-04-07 Permatec Nv THERAPEUTIC COMPOSITION FOR THE TRANSDERMAL ADMINISTRATION OF AN ESTROGENIC OR PROGESTINIC ACTIVE SUBSTANCE OR OF THEIR MIXTURES
US5708038A (en) 1996-06-13 1998-01-13 Univera Pharmaceuticals, Inc. Method of using aloe vera as a biological vehicle
US5770226A (en) 1996-07-10 1998-06-23 Wake Forest University Combined pharmaceutical estrogen-androgen-progestin oral contraceptive
US6139873A (en) 1996-07-10 2000-10-31 Cedars-Sinai Medical Center Combined pharmaceutical estrogen-androgen-progestin
RU2122396C1 (en) 1996-07-12 1998-11-27 Валентина Александровна Андрюшина Bioactive addition to cosmetics
US6228852B1 (en) 1996-07-12 2001-05-08 Carolyn V. Shaak Transdermal application of naturally occurring steroid hormones
US5783208A (en) 1996-07-19 1998-07-21 Theratech, Inc. Transdermal drug delivery matrix for coadministering estradiol and another steroid
US5837289A (en) 1996-07-23 1998-11-17 Grasela; John C. Transdermal delivery of medications using a combination of penetration enhancers
GB9616700D0 (en) 1996-08-09 1996-09-25 Carey Beverly J Hormone supplement
WO1998008547A1 (en) 1996-08-29 1998-03-05 Sekisui Kagaku Kogyo Kabushiki Kaisha Stanozolol-containing percutaneously absorbable preparation
US5863560A (en) * 1996-09-11 1999-01-26 Virotex Corporation Compositions and methods for topical application of therapeutic agents
JPH1087488A (en) 1996-09-13 1998-04-07 Sekisui Chem Co Ltd Preparation for external use for treating dermatosis
US6225299B1 (en) 1996-09-16 2001-05-01 Jenapharm Gmbh & Co. Kg Hormonal agent for skin treatment
CA2241188C (en) 1996-10-24 2007-07-31 Yeong Hua Huang Hydrogel wound dressing and the method of making and using the same
US6331543B1 (en) 1996-11-01 2001-12-18 Nitromed, Inc. Nitrosated and nitrosylated phosphodiesterase inhibitors, compositions and methods of use
US6019988A (en) * 1996-11-18 2000-02-01 Bristol-Myers Squibb Company Methods and compositions for enhancing skin permeation of drugs using permeation enhancers, when drugs and/or permeation enhancers are unstable in combination during long-term storage
WO1998024451A1 (en) 1996-12-05 1998-06-11 Bio-Technology General Corp. Uses of oxandrolone
US5855920A (en) * 1996-12-13 1999-01-05 Chein; Edmund Y. M. Total hormone replacement therapy
US5807957A (en) 1996-12-23 1998-09-15 Macrochem Corporation Cationic film-forming polymer compositions, and use thereof in topical agents delivery system and method of delivering agents to the skin
US5908619A (en) * 1997-01-09 1999-06-01 Minnesota Mining And Manufacturing Company Hydroalcoholic compositions thickened using surfactant/polymer complexes
CA2275971C (en) 1997-01-09 2008-04-29 Conrex Pharmaceutical Corporation Composition for enhancing skin or hair
DE19701949A1 (en) 1997-01-13 1998-07-16 Jenapharm Gmbh Transdermal therapeutic system
GB9700878D0 (en) * 1997-01-17 1997-03-05 Scherer Ltd R P Dosage forms and method for ameliorating male erectile dysfunction
US20010023261A1 (en) 1997-01-27 2001-09-20 Lg Chemical Limited. Novel composition for the transdermal administration of drugs
KR100215027B1 (en) 1997-01-27 1999-08-16 성재갑 Composition for transdermal administration of steroid drugs and formulation containing same
KR20000070757A (en) 1997-02-07 2000-11-25 에버트 챨스 디 Composition and method for supplementing testosterone in women with symptoms of testosterone deficiency
AU735944B2 (en) 1997-02-28 2001-07-19 Minnesota Mining And Manufacturing Company Transdermal device for the delivery of testosterone
WO1998040076A1 (en) 1997-03-10 1998-09-17 Schering Aktiengesellschaft Compositions for the treatment of climacteric disorders with nitric oxide synthase substrates and/or donors, in combination with partial estrogen antagonists
DE69809222T2 (en) 1997-05-07 2003-08-14 Galen Chemicals Ltd DEVICES FOR INTRAVAGINAL ADMINISTRATION OF TESTOSTERONE AND TESTOSTERONE PRECONDITIONAL COMPOUNDS
GB2325855B (en) 1997-06-04 2001-08-29 Healthscene Ltd Preparation for topical application to the male sexual organ
US6103765A (en) 1997-07-09 2000-08-15 Androsolutions, Inc. Methods for treating male erectile dysfunction
US20020012694A1 (en) 1997-09-17 2002-01-31 Alfred J. Moo-Young Transdermal administration of ment
US6342250B1 (en) * 1997-09-25 2002-01-29 Gel-Del Technologies, Inc. Drug delivery devices comprising biodegradable protein for the controlled release of pharmacologically active agents and method of making the drug delivery devices
US5877216A (en) * 1997-10-28 1999-03-02 Vivus, Incorporated Treatment of female sexual dysfunction
US6156753A (en) 1997-10-28 2000-12-05 Vivus, Inc. Local administration of type III phosphodiesterase inhibitors for the treatment of erectile dysfunction
US6127363A (en) 1997-10-28 2000-10-03 Vivus, Inc. Local administration of Type IV phosphodiesterase inhibitors for the treatment of erectile dysfunction
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
US6037346A (en) 1997-10-28 2000-03-14 Vivus, Inc. Local administration of phosphodiesterase inhibitors for the treatment of erectile dysfunction
AU734734B2 (en) 1997-10-28 2001-06-21 Vivus, Inc. Local administration of phosphodiesterase inhibitors for the treatment of erectile dysfunction
WO1999021562A1 (en) 1997-10-28 1999-05-06 Asivi, Llc Treatment of female sexual dysfunction
US6472425B1 (en) 1997-10-31 2002-10-29 Nitromed, Inc. Methods for treating female sexual dysfunctions
US6696484B2 (en) 1997-10-31 2004-02-24 University Of Chicago Office Of Technology And Intellectual Property Method and compositions for regulation of 5-alpha reductase activity
US6046244A (en) * 1997-11-05 2000-04-04 Nexmed Holdings, Inc. Topical compositions for prostaglandin E1 delivery
JP2004515446A (en) * 1997-11-24 2004-05-27 ユニバーシティ・オブ・フロリダ・リサーチ・ファンデーション・インコーポレーテッド Testosterone inhibitors and their use for protecting neurons
GB9726916D0 (en) 1997-12-19 1998-02-18 Danbiosyst Uk Nasal formulation
WO1999032153A1 (en) 1997-12-22 1999-07-01 Alza Corporation Monoglyceride and ethyl palmitate permeation enhancer compositions
US6054446A (en) 1997-12-24 2000-04-25 Sri International Anti-estrogenic steroids, and associated pharmaceutical compositions and methods of use
EP1043020A1 (en) 1997-12-25 2000-10-11 Daiichi Pharmaceutical Co., Ltd. Medicinal composition for percutaneous administration
US5935949A (en) 1998-03-20 1999-08-10 Trustees Of Dartmouth College Use of androgen therapy in fibromyalgia and chronic fatigue syndrome
CN1644201A (en) * 1998-04-17 2005-07-27 奥索-麦克尼尔药品公司 Application of folic acid-containing pharmaceutical compositions
FR2777784B1 (en) 1998-04-27 2004-03-19 Arepa PHARMACEUTICAL COMPOSITION BASED ON ESTROGEN AND PROGESTERONE
US6124461A (en) 1998-05-26 2000-09-26 Saint Louis University, Health Services Center, Research Administration Compounds, compositions, and methods for treating erectile dysfunction
US5847128A (en) 1998-05-29 1998-12-08 Virginia Commonwealth University Water soluble derivatives of cannabinoids
US6277884B1 (en) 1998-06-01 2001-08-21 Nitromed, Inc. Treatment of sexual dysfunction with N-hydroxyguanidine compounds
US6087368A (en) 1998-06-08 2000-07-11 Bristol-Myers Squibb Company Quinazolinone inhibitors of cGMP phosphodiesterase
DE19825856A1 (en) * 1998-06-10 1999-12-16 Labtec Gmbh New topical formulation which includes active agent as liquid lipid nanoparticles in an oil-in-water emulsion
US6266560B1 (en) 1998-06-19 2001-07-24 Genetronics, Inc. Electrically assisted transdermal method and apparatus for the treatment of erectile dysfunction
EP1089736A2 (en) 1998-06-22 2001-04-11 Queen's University At Kingston Method and compositions for the treatment or amelioration of female sexual dysfunction
PL345087A1 (en) 1998-06-25 2001-12-03 Lavipharm Lab A device and method for the treatment of erectile dysfunction
JP2002519366A (en) 1998-07-07 2002-07-02 トランスダーマル・テクノロジーズ・インコーポレーテツド Compositions for rapid and non-irritating transdermal delivery of pharmaceutically active agents, and methods of dispensing such compositions and their delivery
US5880117A (en) * 1998-07-13 1999-03-09 Arnold; Patrick Use of 4-androstenediol to increase testosterone levels in humans
US6207694B1 (en) 1998-07-27 2001-03-27 Howard Murad Pharmaceutical compositions and methods for managing scalp conditions
US6284234B1 (en) 1998-08-04 2001-09-04 Johnson & Johnson Consumer Companies, Inc. Topical delivery systems for active agents
US6436950B1 (en) 1998-08-14 2002-08-20 Nastech Pharmaceutical Company, Inc. Nasal delivery of apomorphine
DK1109814T3 (en) 1998-09-04 2004-08-02 Ortho Mcneil Pharm Inc 5-Heterocyclylpyrazolo [4,3-d] pyrimidin-7-ones for the treatment of male erectile dysfunction
US6509005B1 (en) 1998-10-27 2003-01-21 Virginia Commonwealth University Δ9 Tetrahydrocannabinol (Δ9 THC) solution metered dose inhaler
US6323242B1 (en) 1998-12-02 2001-11-27 Peter Sterling Mueller Treatment of disorders secondary to organic impairments
EP1005831A3 (en) 1998-12-04 2001-05-30 Eisai Co., Ltd. A method for measurement of a penile diameter
AU2223600A (en) 1999-01-06 2000-07-24 Cedars-Sinai Medical Center Hormone replacement for breast cancer patients
US6224573B1 (en) 1999-01-15 2001-05-01 Nexmed Holdings, Inc. Medicament dispenser
JP2000212080A (en) 1999-01-26 2000-08-02 Hiroshi Azuma Erection dysfunction remedy
US6117446A (en) * 1999-01-26 2000-09-12 Place; Virgil A. Drug dosage unit for buccal administration of steroidal active agents
IL144729A0 (en) 1999-02-05 2002-06-30 Cipla Ltd Topical sprays
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6267985B1 (en) 1999-06-30 2001-07-31 Lipocine Inc. Clear oil-containing pharmaceutical compositions
US6294192B1 (en) 1999-02-26 2001-09-25 Lipocine, Inc. Triglyceride-free compositions and methods for improved delivery of hydrophobic therapeutic agents
US6087362A (en) 1999-03-16 2000-07-11 Pentech Pharmaceuticals, Inc. Apomorphine and sildenafil composition
DE29907720U1 (en) 1999-04-30 1999-08-12 H H Heim Und Haus Holding Gmbh Roller shutter arrangement for horizontally split windows
AU4825200A (en) 1999-05-07 2000-11-21 Board Of Regents, The University Of Texas System Oral steroidal hormone compositions and methods of use
EP1187618A1 (en) 1999-06-04 2002-03-20 The General Hospital Corporation Pharmaceutical formulations for treating postmenopausal and perimenopausal women, and their use
CA2376797C (en) 1999-06-11 2011-03-22 Watson Pharmaceuticals, Inc. Administration of non oral androgenic steroids to women
US6309663B1 (en) 1999-08-17 2001-10-30 Lipocine Inc. Triglyceride-free compositions and methods for enhanced absorption of hydrophilic therapeutic agents
EP1206937B1 (en) 1999-07-16 2008-01-23 Shoei Co., Ltd. Nitroimidazole external preparations for the treatment of atopic dermatitis
US6187750B1 (en) * 1999-08-25 2001-02-13 Everyoung Technologies, Inc. Method of hormone treatment for patients with symptoms consistent with multiple sclerosis
US6075028A (en) 1999-09-23 2000-06-13 Graham; Richard Method of treating Tourette's syndrome and related CNS disorders
US6980566B2 (en) 2000-03-10 2005-12-27 Lightwaves Systems, Inc. Method for routing data packets using an IP address based in GEO position
US6582724B2 (en) * 1999-12-16 2003-06-24 Dermatrends, Inc. Dual enhancer composition for topical and transdermal drug delivery
ATE290856T1 (en) 1999-12-16 2005-04-15 Dermatrends Inc HYDROXIDE RELEASING COMPOUNDS TO IMPROVE SKIN PERMEABILITY
US6323241B1 (en) 2000-01-10 2001-11-27 Nexmed (Holdings) Inc. Prostaglandin compositions and methods of treatment for male erectile dysfunction
US20020004065A1 (en) * 2000-01-20 2002-01-10 David Kanios Compositions and methods to effect the release profile in the transdermal administration of active agents
BR0108107A (en) 2000-01-28 2003-03-11 Endorech Inc Selective estrogen receptor modulators in combination with estrogens; pharmaceutical kits and compositions for practicing such a combination; as well as administering said combination
EP1259204B1 (en) 2000-02-28 2007-04-18 Radiant Medical, Inc. Disposable cassette for intravascular heat exchange catheter
DE10009423A1 (en) 2000-02-28 2001-09-06 Henkel Kgaa Cosmetic or pharmaceutical preparation especially for treatment of cellulite comprises nerve fibre stimulator or depolariser, phosphodiesterase inhibitor and antiestrogen
AU2001253054A1 (en) 2000-03-29 2001-10-08 The J. David Gladstone Institutes Methods of treating cognitive decline disease conditions with androgens
DE10015783C2 (en) * 2000-03-30 2003-12-04 Lohmann Therapie Syst Lts Transdermal therapeutic system for delivery of lerisetron and its use
IL151614A0 (en) * 2000-04-07 2003-04-10 Tap Holdings Inc Apomorphine derivatives and methods for their use
DE10019171A1 (en) 2000-04-07 2001-10-18 Schering Ag Compositions for use as penetration enhancers in transdermal formulations for highly lipophilic active ingredients
ATE485837T1 (en) * 2000-08-03 2010-11-15 Antares Pharma Ipl Ag COMPOSITION FOR TRANSDERMAL AND/OR TRANSMUCOSAL ADMINISTRATION OF ACTIVE INGREDIENTS WHICH GUARANTEES ADEQUATE THERAPEUTIC LEVELS
US20040198706A1 (en) 2003-03-11 2004-10-07 Carrara Dario Norberto R. Methods and formulations for transdermal or transmucosal application of active agents
EP1315502B1 (en) 2000-08-30 2010-03-17 Unimed Pharmaceuticals, LLC Method for treating erectile dysfunction and increasing libido in men
BR0113649A (en) 2000-08-30 2004-07-20 Unimed Pharmaceuticals Inc Process for Increasing Testosterone and Related Steroid Concentrations in Women
US20030139384A1 (en) 2000-08-30 2003-07-24 Dudley Robert E. Method for treating erectile dysfunction and increasing libido in men
JP5039252B2 (en) 2000-08-31 2012-10-03 ユニメッド ファーマシューティカルズ,リミティド ライアビリティ カンパニー Pharmaceutical compositions and methods for treating sexual dysfunction
FR2814074B1 (en) * 2000-09-15 2003-03-07 Theramex NOVEL TOPICAL ESTRO-PROGESTIVE COMPOSITIONS WITH SYSTEMIC EFFECT
US6743448B2 (en) 2000-12-11 2004-06-01 Abraham H. Kryger Topical testosterone formulations and associated methods
WO2002051421A2 (en) * 2000-12-22 2002-07-04 Dr. August Wolff Gmbh & Co. Gel composition and trans-scrotal application of a composition for the treatment of hypogonadism
JP2002212105A (en) 2001-01-22 2002-07-31 Lion Corp Aqueous skin care composition
CA2446622C (en) * 2001-05-11 2012-08-14 Elan Corporation, Plc Isostearic acid salts as permeation enhancers
US20030175329A1 (en) 2001-10-04 2003-09-18 Cellegy Pharmaceuticals, Inc. Semisolid topical hormonal compositions and methods for treatment
ATE439829T1 (en) 2001-12-07 2009-09-15 Besins Int Belgique GEL OR SOLUTION CONTAINING DIHYDROTESTOSTERONE, PROCESS OF PRODUCTION AND USE THEREOF
WO2003066130A2 (en) * 2002-02-07 2003-08-14 Massachusetts Institute Of Technology Transdermal drug delivery systems
CN1470239A (en) 2002-07-28 2004-01-28 王怀秀 Testosterone intradermic sustained preparation
FR2851470B1 (en) 2003-02-20 2007-11-16 Besins Int Belgique PHARMACEUTICAL COMPOSITION FOR TRANSDERMAL OR TRANSMUCTIVE DELIVERY
US20050042268A1 (en) * 2003-07-16 2005-02-24 Chaim Aschkenasy Pharmaceutical composition and method for transdermal drug delivery
US20050025833A1 (en) * 2003-07-16 2005-02-03 Chaim Aschkenasy Pharmaceutical composition and method for transdermal drug delivery
US7968532B2 (en) 2003-12-15 2011-06-28 Besins Healthcare Luxembourg Treatment of gynecomastia with 4-hydroxy tamoxifen
US7138389B2 (en) 2004-02-09 2006-11-21 University Of Washington Oral androgen therapy using modulators of testosterone bioavailability
US20050222106A1 (en) 2004-04-01 2005-10-06 Stefan Bracht Drospirenone-containing preparations for transdermal use
RU2275930C2 (en) * 2004-04-26 2006-05-10 ООО "РусГен" Composition for correction of human endocrine system age-related changes (variants) and method for production of pharmaceutical formulation based on the same
US8907153B2 (en) 2004-06-07 2014-12-09 Nuvo Research Inc. Adhesive peel-forming formulations for dermal delivery of drugs and methods of using the same
US20070190124A1 (en) 2004-06-07 2007-08-16 Jie Zhang Two or more solidifying agent-containing compositions and methods for dermal delivery of drugs
US20070196453A1 (en) 2004-06-07 2007-08-23 Jie Zhang Two or more non-volatile solvent-containing compositions and methods for dermal delivery of drugs
US20070196323A1 (en) 2004-06-07 2007-08-23 Jie Zhang Polyvinyl alcohol-containing compositions and methods for dermal delivery of drugs
US20070189977A1 (en) 2004-06-07 2007-08-16 Jie Zhang Spray-on formulations and methods for dermal delivery of drugs
JP5244388B2 (en) 2004-08-18 2013-07-24 ウオーターズ・テクノロジーズ・コーポレイシヨン Apparatus and method for generating or carrying fluid under pressure and seal member used in the apparatus
EP1634583A1 (en) 2004-09-09 2006-03-15 Laboratoires Besins International Testosterone gels comprising propylene glycol as penetration enhancer
EP1647271A1 (en) 2004-10-14 2006-04-19 Laboratoires Besins International 4-Hydroxy tamoxifen gel formulations
US20070082039A1 (en) * 2004-10-18 2007-04-12 Jones Gerald S Jr Synthesis of fatty alcohol esters of alpha-hydroxy carboxylic acids, the use of the same as percutaneous permeation enhancers, and topical gels for the transdermal delivery of steroids
US20070088012A1 (en) * 2005-04-08 2007-04-19 Woun Seo Method of treating or preventing type-2 diabetes
CN101217874B (en) * 2005-04-12 2012-06-06 优尼麦德药物股份有限公司 Method of treating or preventing bone deterioration or osteoporosis
JP2008536851A (en) * 2005-04-13 2008-09-11 ユニメド ファーマスーティカルズ インク How to increase testosterone and related steroid levels in women
WO2006113227A2 (en) 2005-04-13 2006-10-26 Massachusetts Institute Of Technology Compositions facilitating drug penetration and drug retention in skin
ES2930658T3 (en) 2005-04-15 2022-12-20 Tolmar Inc Pharmaceutical delivery systems for hydrophobic drugs and compositions comprising the same
ES2607454T3 (en) 2005-06-03 2017-03-31 Acrux Dds Pty Ltd Method and composition for transdermal testosterone delivery
US20070065494A1 (en) * 2005-08-03 2007-03-22 Watson Laboratories, Inc. Formulations and Methods for Enhancing the Transdermal Penetration of a Drug
DE102005044008B4 (en) * 2005-09-14 2007-07-12 Krohne Ag Method for testing a mass flowmeter
US20070254036A1 (en) 2006-04-13 2007-11-01 Besins Healthcare Sa Treatment of menopause associated symptoms
US20080220068A1 (en) 2006-07-31 2008-09-11 Laboratories Besins International Treatment and prevention of excessive scarring
EP2062575B1 (en) 2006-09-11 2012-02-15 Sekisui Chemical Co., Ltd. Adhesive preparation comprising desglymidodrine
US20080261937A1 (en) * 2007-03-23 2008-10-23 Dudley Robert E Pharmaceutical compositions and method for treating pediatric hypogonadism
ES2376675T3 (en) * 2008-07-23 2012-03-15 Telstar Technologies, S.L. SECONDARY DRYING CONTROL METHOD IN A FREEZING DRYING PROCESS.

Patent Citations (46)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US568112A (en) * 1896-09-22 Vehicle-wheel
US4078060A (en) * 1976-05-10 1978-03-07 Richardson-Merrell Inc. Method of inducing an estrogenic response
US4861764A (en) * 1986-11-17 1989-08-29 Macro Chem. Corp. Percutaneous absorption enhancers, compositions containing same and method of use
US5223261A (en) * 1988-02-26 1993-06-29 Riker Laboratories, Inc. Transdermal estradiol delivery system
US5231087A (en) * 1988-03-16 1993-07-27 Cellegy Pharmaceuticals, Inc. Treatment of skin diseases and tumors with esters and amides of monocarboxylic acids
US5200190A (en) * 1988-10-11 1993-04-06 Sekisui Kagaku Kogyo Kabushiki Kaisha Percutaneous pharmaceutical preparation
US5788984A (en) * 1988-10-27 1998-08-04 Schering Aktiengesellschaft Gestodene-containing agent for transdermal administration
US5238944A (en) * 1988-12-15 1993-08-24 Riker Laboratories, Inc. Topical formulations and transdermal delivery systems containing 1-isobutyl-1H-imidazo[4,5-c]quinolin-4-amine
US5550107A (en) * 1989-03-10 1996-08-27 Endorecherche Inc. Combination therapy for the treatment of estrogen-sensitive disease
US5788983A (en) * 1989-04-03 1998-08-04 Rutgers, The State University Of New Jersey Transdermal controlled delivery of pharmaceuticals at variable dosage rates and processes
US5232703A (en) * 1989-07-21 1993-08-03 Izhak Blank Estradiol compositions and methods for topical application
US5152997A (en) * 1990-12-11 1992-10-06 Theratech, Inc. Method and device for transdermally administering testosterone across nonscrotal skin at therapeutically effective levels
US5643899A (en) * 1992-06-19 1997-07-01 Cellegy Pharmaceuticals, Inc. Lipids for epidermal moisturization and repair of barrier function
US5955455A (en) * 1993-01-19 1999-09-21 Endorecherche, Inc. Therapeutic methods and delivery systems utilizing sex steroid precursors
US5723114A (en) * 1993-03-19 1998-03-03 Cellegy Pharmaceuticals Inc. Penetration enhancing compositions and methods of their use
US7097853B1 (en) * 1994-09-14 2006-08-29 3M Innovative Properties Company Matrix for transdermal drug delivery
US6818226B2 (en) * 1996-02-19 2004-11-16 Acrux Dds Pty. Ltd. Dermal penetration enhancers and drug delivery systems involving same
US5730987A (en) * 1996-06-10 1998-03-24 Omar; Lotfy Ismail Medication for impotence containing lyophilized roe and a powdered extract of Ginkgo biloba
US6007837A (en) * 1996-07-03 1999-12-28 Alza Corporation Drug delivery devices and process of manufacture
US5760096A (en) * 1996-10-18 1998-06-02 Thornfeldt; Carl R. Potent penetration enhancers
US5952000A (en) * 1996-10-30 1999-09-14 Theratech, Inc. Fatty acid esters of lactic acid salts as permeation enhancers
US6019997A (en) * 1997-01-09 2000-02-01 Minnesota Mining And Manufacturing Hydroalcoholic compositions for transdermal penetration of pharmaceutical agents
US5968919A (en) * 1997-10-16 1999-10-19 Macrochem Corporation Hormone replacement therapy drug formulations for topical application to the skin
US6319913B1 (en) * 1997-11-10 2001-11-20 Cellegy Pharmaceuticals, Inc. Penetration enhancing and irritation reducing systems
US5942545A (en) * 1998-06-15 1999-08-24 Macrochem Corporation Composition and method for treating penile erectile dysfunction
US6200591B1 (en) * 1998-06-25 2001-03-13 Anwar A. Hussain Method of administration of sildenafil to produce instantaneous response for the treatment of erectile dysfunction
US20020128176A1 (en) * 1999-01-27 2002-09-12 Wolf-Georg Forssmann Treatment of erectile dysfunctions with C-Type natriuretic polypeptide (CNP) as a monotherapy or in combination with phosphodiesterase inhibitors
US6586000B2 (en) * 1999-12-16 2003-07-01 Dermatrends, Inc. Hydroxide-releasing agents as skin permeation enhancers
US6562370B2 (en) * 1999-12-16 2003-05-13 Dermatrends, Inc. Transdermal administration of steroid drugs using hydroxide-releasing agents as permeation enhancers
US6562369B2 (en) * 1999-12-16 2003-05-13 Dermatrends, Inc. Transdermal administration of androgenic drugs hydroxide-releasing agents as permeation enhancers
US20110172196A1 (en) * 2000-08-30 2011-07-14 Dudley Robert E Pharmaceutical composition and method for treating hypogonadism
US20050113353A1 (en) * 2000-08-30 2005-05-26 Dudley Robert E. Pharmaceutical composition and method for treating hypogonadism
US20030022877A1 (en) * 2000-08-30 2003-01-30 Dudley Robert E. Method of increasing testosterone and related steroid concentrations in women
US6503894B1 (en) * 2000-08-30 2003-01-07 Unimed Pharmaceuticals, Inc. Pharmaceutical composition and method for treating hypogonadism
US20110201586A1 (en) * 2000-08-30 2011-08-18 Dudley Robert E Pharmaceutical composition and method for treating hypogonadism
US20120058981A1 (en) * 2000-08-30 2012-03-08 Dudley Robert E Androgen pharmaceutical composition and method for treating depression
US20030027804A1 (en) * 2001-06-27 2003-02-06 Van Der Hoop Roland Gerritsen Therapeutic combinations for the treatment of hormone deficiencies
US20040072810A1 (en) * 2001-11-07 2004-04-15 Besins International Belgique Pharmaceutical composition in the form of a gel or a solution based on dihydrotestosterone, process for preparing it and uses thereof
US20110306582A1 (en) * 2002-03-15 2011-12-15 Dudley Robert E Androgen pharmaceutical composition and method for treating depression
US7320968B2 (en) * 2002-04-19 2008-01-22 Bentley Pharmaceuticals, Inc. Pharmaceutical composition
US20040110732A1 (en) * 2002-12-10 2004-06-10 Besins International Belgique Pharmaceutical composition for transdermal or transmucosal administration comprising at least one progestin and/or at least one oestrogen, process for preparing it and uses thereof
US20050020552A1 (en) * 2003-07-16 2005-01-27 Chaim Aschkenasy Pharmaceutical composition and method for transdermal drug delivery
US20110306583A1 (en) * 2005-10-12 2011-12-15 Ramana Malladi Testosterone Gel And Method Of Use
US20120028948A1 (en) * 2005-10-12 2012-02-02 Ramana Malladi Testosterone gel and method of use
US20120028946A1 (en) * 2005-10-12 2012-02-02 Ramana Malladi Testosterone gel and method of use
US20110269729A1 (en) * 2005-10-12 2011-11-03 Ramana Malladi Testosterone Gel And Method Of Use

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9132089B2 (en) 2000-08-30 2015-09-15 Besins Healthcare Inc. Pharmaceutical composition and method for treating hypogonadism
US20110172196A1 (en) * 2000-08-30 2011-07-14 Dudley Robert E Pharmaceutical composition and method for treating hypogonadism
US9125816B2 (en) 2000-08-30 2015-09-08 Besins Healthcare Inc. Pharmaceutical composition and method for treating hypogonadism
US20060100186A1 (en) * 2003-06-18 2006-05-11 White Hillary D Trandsdermal compositions and methods for treatment of fibromyalgia and chronic fatigue syndrome
US7799769B2 (en) 2003-06-18 2010-09-21 White Mountain Pharma, Inc. Transdermal compositions and methods for treatment of fibromyalgia and chronic fatigue syndrome
US20110009318A1 (en) * 2003-06-18 2011-01-13 White Mountain Pharma, Inc. Transdermal Compositions and Methods for Treatment of Fibromyalgia and Chronic Fatigue Syndrome
US20110118227A1 (en) * 2003-06-18 2011-05-19 White Mountain Pharma, Inc. Methods for the Treatment of Fibromyalgia and Chronic Fatigue Syndrome
US8999963B2 (en) 2003-06-18 2015-04-07 White Mountain Pharma, Inc. Transdermal compositions and methods for treatment of fibromyalgia and chronic fatigue syndrome
US8883769B2 (en) 2003-06-18 2014-11-11 White Mountain Pharma, Inc. Methods for the treatment of fibromyalgia and chronic fatigue syndrome
US8754070B2 (en) 2005-10-12 2014-06-17 Unimed Pharmaceuticals, Llc Testosterone gel and method of use
US8466138B2 (en) 2005-10-12 2013-06-18 Unimed Pharmaceuticals, Llc Testosterone gel and method of use
US8741881B2 (en) 2005-10-12 2014-06-03 Unimed Pharmaceuticals, Llc Testosterone gel and method of use
US8486925B2 (en) 2005-10-12 2013-07-16 Unimed Pharmaceuticals, Llc Testosterone gel and method of use
US8759329B2 (en) 2005-10-12 2014-06-24 Unimed Pharmaceuticals, Llc Testosterone gel and method of use
US8466136B2 (en) 2005-10-12 2013-06-18 Unimed Pharmaceuticals, Llc Testosterone gel and method of use
US8466137B2 (en) 2005-10-12 2013-06-18 Unimed Pharmaceuticals, Llc Testosterone gel and method of use
US8729057B2 (en) 2005-10-12 2014-05-20 Unimed Pharmaeuticals, LLC Testosterone gel and method of use
US20070237822A1 (en) * 2005-10-12 2007-10-11 Ramana Malladi Testosterone gel and method of use
US9642863B2 (en) 2010-11-18 2017-05-09 White Mountain Pharma, Inc. Methods for treating chronic or unresolvable pain and/or increasing the pain threshold in a subject and pharmaceutical compositions for use therein
US9642862B2 (en) 2010-11-18 2017-05-09 White Mountain Pharma, Inc. Methods for treating chronic or unresolvable pain and/or increasing the pain threshold in a subject and pharmaceutical compositions for use therein
US9757388B2 (en) 2011-05-13 2017-09-12 Acerus Pharmaceuticals Srl Intranasal methods of treating women for anorgasmia with 0.6% and 0.72% testosterone gels
US10111888B2 (en) 2011-05-13 2018-10-30 Acerus Biopharma Inc. Intranasal 0.15% and 0.24% testosterone gel formulations and use thereof for treating anorgasmia or hypoactive sexual desire disorder
US10668084B2 (en) 2011-05-13 2020-06-02 Acerus Biopharma Inc. Intranasal lower dosage strength testosterone gel formulations and use thereof for treating anorgasmia or hypoactive sexual desire disorder
US11090312B2 (en) 2013-03-15 2021-08-17 Acerus Biopharma Inc. Methods of treating hypogonadism with transnasal testerosterone bio-adhesive gel formulations in male with allergic rhinitis, and methods for preventing an allergic rhinitis event
US11744838B2 (en) 2013-03-15 2023-09-05 Acerus Biopharma Inc. Methods of treating hypogonadism with transnasal testosterone bio-adhesive gel formulations in male with allergic rhinitis, and methods for preventing an allergic rhinitis event

Also Published As

Publication number Publication date
ES2326621T3 (en) 2009-10-16
BRPI0113670B8 (en) 2021-05-25
EP1313482B1 (en) 2009-04-29
EP2070517A2 (en) 2009-06-17
US20050112181A1 (en) 2005-05-26
US20050113353A1 (en) 2005-05-26
HUP0302921A2 (en) 2004-01-28
PL205875B1 (en) 2010-06-30
CY1110907T1 (en) 2015-06-10
US9125816B2 (en) 2015-09-08
CN101081203B (en) 2010-09-29
CN1527714A (en) 2004-09-08
HU230401B1 (en) 2016-04-28
CN101081203A (en) 2007-12-05
MA27127A1 (en) 2005-01-03
KR100866715B1 (en) 2008-11-03
US20150313914A1 (en) 2015-11-05
US20140011788A1 (en) 2014-01-09
US20130303501A1 (en) 2013-11-14
NZ524473A (en) 2006-09-29
NO20030961D0 (en) 2003-02-28
ZA200301705B (en) 2004-07-08
US20030050292A1 (en) 2003-03-13
UA76958C2 (en) 2006-10-16
US20020183296A1 (en) 2002-12-05
HUP0302921A3 (en) 2006-07-28
ZA200301687B (en) 2004-09-13
US20110201586A1 (en) 2011-08-18
ZA200301686B (en) 2004-04-19
AU2001290598B2 (en) 2006-07-13
US20110172196A1 (en) 2011-07-14
IL154668A (en) 2007-12-03
BRPI0113670B1 (en) 2018-02-14
DE60138553D1 (en) 2009-06-10
US20130261097A1 (en) 2013-10-03
OA12386A (en) 2006-04-17
BR0113670A (en) 2004-11-09
EP2070517A3 (en) 2009-07-29
WO2002017926A1 (en) 2002-03-07
AU9059801A (en) 2002-03-13
ATE429920T1 (en) 2009-05-15
US9132089B2 (en) 2015-09-15
EP2281553A3 (en) 2012-11-14
EP1313482A1 (en) 2003-05-28
US20030232072A1 (en) 2003-12-18
KR20030064747A (en) 2003-08-02
EA009815B1 (en) 2008-04-28
IL154668A0 (en) 2003-09-17
EA200300313A1 (en) 2004-08-26
CN101077350A (en) 2007-11-28
CN101077350B (en) 2012-06-27
NO20030961L (en) 2003-04-25
PT1313482E (en) 2009-07-29
US6503894B1 (en) 2003-01-07
SI1313482T1 (en) 2009-10-31
EP2281553B1 (en) 2020-12-02
DK1313482T3 (en) 2009-08-17
PL366117A1 (en) 2005-01-24
EP2281553A2 (en) 2011-02-09
NO335506B1 (en) 2014-12-22

Similar Documents

Publication Publication Date Title
US9125816B2 (en) Pharmaceutical composition and method for treating hypogonadism
AU2001290598A1 (en) Pharmaceutical composition and method for treating hypogonadism
US20150250801A1 (en) Androgen pharmaceutical composition and method for treating depression
US20040092494A9 (en) Method of increasing testosterone and related steroid concentrations in women
US20110306582A1 (en) Androgen pharmaceutical composition and method for treating depression
JP5039252B2 (en) Pharmaceutical compositions and methods for treating sexual dysfunction
MXPA04008988A (en) Tissue monitoring system for intravascular infusion.

Legal Events

Date Code Title Description
AS Assignment

Owner name: UNIMED PHARMACEUTICALS, INC., GEORGIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:DUDLEY, ROBERT E.;REEL/FRAME:015729/0691

Effective date: 20041116

Owner name: BESINS INTERNATIONAL-USA, INC., VIRGINIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:DROUIN, DOMINIQUE;REEL/FRAME:015729/0712

Effective date: 20050120

AS Assignment

Owner name: UNIMED PHARMACEUTICALS, LLC, GEORGIA

Free format text: CHANGE OF NAME;ASSIGNOR:UNIMED PHARMACEUTICALS, INC.;REEL/FRAME:020654/0154

Effective date: 20071228

AS Assignment

Owner name: LABORATOIRES BESINS INTERNATIONAL, SAS, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:UNIMED PHARMACEUTICALS, INC.;REEL/FRAME:025401/0190

Effective date: 20071004

STCB Information on status: application discontinuation

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